Putative tumor metastasis-associated genes in human gastric cancer

  • Authors:
    • Tsung-Fan Tuan
    • Cheng-Ta Chung
    • Hsiao-Hui Tsou
    • Fong-Wen Chen
    • Heng-Liang  Lin
    • Yiu-Kay Lai
    • Wen-Sen Lee
    • Yu-Sheng Chao
    • Ling-Ling Hwang
    • Chiung-Tong Chen
  • View Affiliations

  • Published online on: May 31, 2012     https://doi.org/10.3892/ijo.2012.1502
  • Pages: 1068-1084
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer is one of the leading causes of cancer mortality and its malignancy, resulting from disseminated cancer cells of diffuse type, is clinically manifested as metastases to the liver and peritoneum. The aim of the present study was to identify putative tumor metastasis-associated genes in human gastric cancer cells of diffuse type. An MKN45 cell line constitutively expressing green fluorescent protein (MKN45-GFP) was established and selected using the Transwell® system for invasive sublines MKN45-GFP-4, MKN45-GFP-10 and MKN45-GFP-12. MKN45-GFP-10 and MKN45-GFP-12 are highly invasive compared to the others. The mRNA levels were measured with cDNA microarrays and correlated with their invasion abilities in these sublines. Many of the genes identified with a positive or negative correlation are associated with angiogenesis, cell cycle, cytoskeleton and cell motility, protease and cell adhesion, as well as cellular signal transduction. In particular, novel genes without known functions were also noted. RT-PCR and western blot analyses were applied to verify the expression of selective genes. Following orthotopical intraperitoneal implantation, MKN45-GFP-12 demonstrated significantly higher in vivo tumor malignancies than parental MKN45-GFP in ascites induction and liver -invasion in mice. We have identified putative gastric tumor metastasis-associated, as well as novel genes. These genes and their protein products are to be further explored for their functional roles associated with tumor metastasis. The molecular profiles of these identified genes, gene transcripts and proteins in the patient specimens are likely to be useful biomarkers for diagnostic, therapeutic and/or prognostics. Most importantly, they may be used as molecular targets for the discovery of antitumor drugs against human gastric cancer metastasis.

Introduction

Gastric cancer is one of the most common types of human cancer (1) and the second most common cause of cancer mortality in the world (2). Gastric cancer has a wide variation in geographical distribution and the incidence is particularly high in Asia, South America and Eastern Europe (35). Gastric cancer patients are frequently diagnosed in advanced stages. Approximately 50% of all patients present with unresectable, locally advanced or metastatic diseases (6,7) and recurrence after curative gastrectomy remains high (8,9). The prognosis for patients with advanced gastric cancer remains dismal, and little improvement in the survival rates has been achieved in recent years (6,8,9). Gastric tumor metastases, during which the cancer cells spread to distal sites such as the liver, omentum, and lymph nodes, are the leading cause of cancer mortality. Gastric tumor of diffuse type is poorly differentiated and comprises dissemination-prone gastric cancer cells lacking the abilities of tissue cohesion and gastric biological functions. This particular type of gastric cancer cells demonstrates an increased propensity for metastatic spread via the intra- and trans-peritoneal wall and, therefore, have been associated with poor prognosis (7). The somatic inactivation of E-cadherin in diffuse type gastric cancer also increases epithelial-mesenchymal transition (EMT) ability and leads to an enhanced metastasis potential of this gastric cancer type (10). There is currently no effective chemotherapeutics, nor is a molecular-targeted drug available, for treating the advanced gastric cancerous diseases. Further research into molecular targeting drugs against gastric tumor metastasis is warranted.

In the search for target molecules for the development of anticancer drugs, research efforts to identify biological molecules involved in the gastric cancer cell specific proliferation, invasion and metastasis have been extensive. Elevated plasma osteopontin was associated with gastric cancer development and patient survival (11). Overexpression of c-met (12), LOXL2 (13), EGFR (14), HER2 (15), TNS4 (16) and phosphorylated mTOR (17) in gastric tumors has been well correlated with the prognostic factors of tumor staging, the depth of tumor invasion and/or lymph node metastasis. On the other hand, reduced expressions of nm23 (18), galectin-3 (19), or TIP30 (20), loss of RUNX3 (21) or E-cadherin (14) expressions, and abnormal expression of E-cadherin (22) in gastric tumors has been associated with gastric tumor metastasis and poor prognosis. More systemically investigated, array-based technologies were utilized to analyze and identify genes associated with gastric cancer metastasis. Several studies compared human primary to metastatic gastric cancer cell lines (23,24), human gastric cancer cell lines to a normal gastric cell line (25), unpaired tumors to normal tissues (26), paired patient gastric tumors to normal gastric tissues (25,2731), and patient gastric tumors with different clinical stages (32). However, differences in the genomic backgrounds of patients and the mix of the tumor and its surrounding cells may increase the complexities and difficulties for the identification of specific genes and for database analyses. Results of these array-based studies individually suggested that putative metastasis-associated genes are not entirely the same, nor are they similar, to a great extent, to each other. Further studies are, therefore, needed to identify biologically relevant molecular targets against which effective drugs for human gastric cancer may be discovered.

In the present study, we generated, from a single parental cell, a set of cell sublines exhibiting different metastatic abilities and used them to identify the genes whose expression levels are correlated to their metastatic abilities. We applied a genome-wide scale cDNA microarray analysis using a biological functional approach different from those previously reported. A human gastric cancer cell MKN45 originally derived from a poorly differentiated gastric adenocarcinoma of the stomach of a 62-year-old woman (33,34) was used as a model cell for its diffuse phenotype potentially prone to metastasis. A series of the human gastric cancer MKN45 cell subclones with varying invasion capabilities were established. A number of putative tumor metastasis-associated genes were identified using cDNA microarray analysis and verified with correlated mRNA levels and protein expressions.

Materials and methods

Cell line and transfection

The human diffuse-type gastric cancer cell line MKN45 (JCRB0254) was purchased from the Japanese Collection of Research Bioresources/Human Science Research Resources Bank (Osaka, Japan). The cells were grown in RPMI-1640 medium (Gibco, product no. 31800105) with 10% fetal bovine serum (FBS) (Invitrogen, Carlsbad, CA, USA) and 2 mM L-glutamine at 37°C, 5% CO2. One day before the transfection, 8×103 MKN45 cells were seeded into 24-well culture plate and grown for 16–24 h to 70–90% confluence for transfection. A mixture of enhanced green fluorescent protein (GFP) expression plasmid (pEGFP-C1, cat. no. 6084-1) encoding under the control of cytomegalovirus promoter, OPTI-MEM and Lipofectamine™ 2000 from Invitrogen at a ratio of 2 μg:2 μl:100 μl was added to the MKN45 cells. The cells were harvested following incubation for 48 h at 37°C followed by selection of the GFP-expressing clones by incubating with 1 mg/ml G418 (Invitrogen) and isolated with limiting dilution method to obtain stably GFP-expressing MKN45-GFP sublines.

Selection of highly invasive MKN45-GFP sublines

The MKN45-GFP cells of the isolated clone were selected for differential invasive characteristics using Transwell® plates from Corning (Acton, MA, USA) with a method modified from one previously described (35) Briefly, Transwell of the 24-well inserts with semi-permeable polycarbonate membrane (8 μm in pore size, Corning, cat. no. CLS3422, Corning, NY, USA) were coated with 50 μl per well of reconstituted basement-membrane matrix Matrigel™ (1:1 diluted with growth medium) from BD Biosciences (San Jose, CA, USA). MKN45-GFP cells in RPMI-1640 containing 10% FBS were seeded at 1×105 cells/200 μl/well onto the upper surface of the Matrigel-coated membrane. Following incubation for 72 h at 37°C, the inserts were removed. The cells that migrated and invaded through the membrane and attached to the surface of bottom well were harvested aseptically and subsequently amplified for further selection processes. The harvested invasive MKN45-GFP cell sublines after each selection round were named with a number correspondingly.

In vitro invasion assay

The basement membrane matrix coated Transwell system was used to measure the invasion ability of tumor cells as previously reported (36). Transwell of 24-well plates with a porous polycarbonate membrane of 8-μm pore size from Corning (Corning, NY) were coated with 50 μl Matrigel diluted (1:20 = v:v) with RPMI-1640 culture medium. Cells (5×105) suspended in 200 μl RPMI-1640 with 1% FBS were seeded into upper chamber. The lower chamber was placed with 600 μl RPMI-1640 with 10% FBS. Following incubation for 72 h at 37°C, the cells that invaded through the Matrigel-coated membrane to the lower surface of the membrane and the bottom surface of the culture well were fixed with 3.7% formaldehyde in phosphate-buffered saline (PBS) for 1 h. The cells that remained on the upper surface of the membrane were scraped with a cotton bud completely and invaded cells attached on the opposite surface of the membrane in the lower chamber were stained with hematoxylin for 1 h. The invaded cancer cells were then visualized and counted from 5 different viewing areas under 100-fold magnification using a DM IRB inverted microscope from Leica Microsystems (Wetzlar, Germany).

Cell proliferation activity

Cancer cell proliferation activity was measured using a colorimetric 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium salt (MTS)/phenazine methosulfate (PMS) assay system as previously reported (37). Cancer cells were seeded at 2,000, 4,000, and 6,000 cells/well in 96-well culture plates (Corning, Acton, MA) in triplicates. The cells were harvested and the cell numbers were measured every 24 h or 6 consecutive days by a colorimetric reaction using MTS/PMS mixture, 2 mg/ml MTS (Promega, Madison, WI, USA) and 0.38 mg/ml PMS (Sigma, St. Louis, MO, USA), in phenol red-free RPMI-1640 medium (Gibco, product no. 11835-030) followed by optical density measurements at 490 nm after a 90-min incubation at 37°C. The cell doubling time was obtained by nonlinear regression with the equation: N=Ao*2^(T/Td) using SigmaPlot® from SPSS Inc. (Chicago, IL, USA), in which N is the total cell number at time T; Ao is the initial cell number; and Td is the cell doubling time.

Orthotopic tumor growth and metastasis in nude mice

Adult male athymic Nu-Fox1nu nude mice purchased from BioLasco (Ilan, Taiwan) were housed in sterilized cages under 12-h light/dark cycles with water and food ad libitum. As previously described (38), nude mice were intraperitoneally inoculated with 5×106 MKN45-GFP or MKN45-GFP-12 cells/mouse, suspended in 0.2 ml phenol-red free RPMI-1640 medium using 24G syringe needles at 35 nude mice per cancer cell type. After cancer cell inoculation, the occurrence of ascites was monitored weekly. The progression of the tumor growth was monitored by a fluorescence stereomicroscope MZ FLIII from Leica and animal body weights were recorded twice a week. GFP-expressing tumors were visualized with a D470/40× bandpass filter and a 495 DCLP dichroic filter. On the second week after the inoculation, 7 nude mice per cell type were euthanized weekly and the disseminated and metastasized tumors growing in the peritoneal cavities were visualized after laparotomy and the numbers of tumor nodules were counted using a fluorescent imaging system LT-9500 Illumatool TLS from Lightools Research (Encinitas, CA, USA). Metastasized tumors that had invaded into the livers of the nude mice were surgically removed and sectioned for microscopic examination following hematoxylin and eosin staining. The use of the animals was approved by The Institutional Animal Care and Use Committee of The National Health Research Institutes.

cDNA microarray assay

Total RNAs were isolated from the cancer cells using TRIzol® reagent (product no. 15596026, Invitrogen) according to the manufacturer’s protocol. The purified RNA was quantified in optical density at 260 nm by an ND-1000 spectrophotometer from Nanodrop Technology (Wilmington, DE, USA) and qualified by Agilent 2100 Bioanalyzer. Total RNA of 0.5 μg was amplified by a low RNA input fluorescent linear amplification kit (cat. no. 5184–3523) from Agilent Technologies (Santa Clara, CA, USA) using Cyanine 3 (Cy3)-labeled CTP (cat. no. NEL580001EA) or Cyanine 5 (Cy5)-labeled CTP (cat. no. NEL581001EA) from Perkin Elmer (Waltham, MA, USA) during the in vitro transcription. The cancer cell RNAs were labeled with Cy5 and Universal Human Reference RNAs (cat. no. 636538) from Clontech (Mountain View, CA, USA) were labeled with Cy3. The Cyanine-labeled RNAs (cRNAs) of 2 μg were fragmented to an average size of 50–100 nucleotides by incubation with fragmentation buffer at 60°C for 30 min. The fragmented labeled cRNAs were then pooled and hybridized to Human 1A Oligo Microarray (V2) from Agilent Technologies at 60°C for 17 h. After they were washed, the arrays were dried using a nitrogen-filled air gun and scanned with Agilent dual DNA microarray scanner at 535 nm for Cy3 and 625 nm for Cy5, respectively. The scanned images were extracted by Feature Extraction 8.1 from Agilent Technologies and analyzed to quantify signal and background intensity and to substantially normalize the data by rank-consistency-filtering LOWESS method. The microarray data, including 4 analyzed samples (MKN45-GFP, MKN45-GFP-4, MKN45-GFP-10, and MKN45-GFP-12), have been deposited to the Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo/; accession number GSE33570).

RT-PCR analysis

Samples of the total RNA (1–5 μg) from the cancer cells were reverse-transcribed in a total volume of 20 μl by the SuperScript III First-Strand Synthesis System (cat. no. 18080–400) from Invitrogen. The reverse transcription products (1 μg) were used directly for PCR amplification. PCR amplification was performed with the PCR Reagent System (cat. no. 10198-018) from Invitrogen according to the manufacturer’s instructions in a PC818 Program Temp. Control System from ASTEC (Fukuoka, Japan). Oligonucleotide primers used for the amplification of cDNAs for oxytocin receptor (OTR) were 5′-CCTTCATCGTGTGCTGGACG-3′ (forward) and 5′-CTAGGAGCAGAGCACTTATG-3′ (reverse); leucine-rich repeat-containing G protein-coupled receptor 4/G protein-coupled receptor 48 (LGR4/GPR48) were 5′-GGGAAGCT GGATGATTCGTCTTACT-3′ (forward) and 5′-GAAAAGGG GAAAACAGCCTGCT-3′ (reverse); trefoil factor 3 (TFF3) were 5′-AGAGCCTTCCCCAAGCAAACA-3′ (forward) and 5′-GCAGGGGCTTGAAACACCAA-3′ (reverse); brain expressed X-linked 2 (BEX2) were 5′-CCTTGGCCCTACCT TTGAATGT-3′ (forward) and 5′-TGCTGACTGCCCGCA AACTA-3′ (reverse); sarcoglycan-ε (SGCE) were 5′-TTCTCC AAGGTACACTCCGATCG-3′ (forward) and 5′-GGCCGAT GTGATGTTTATGGC-3′ (reverse); insulin-like growth factor binding protein 3 (IGFBP3) were 5′-ACGAGTCTCAGAGC ACAGATACCC-3′ (forward) and 5′-TATCCACACACCAG CAGAAGCC-3′ (reverse); and internal control glyceraldehyde-3-phosphate dehydrogenase (GAPDH) were 5′-TCCACCACC CTGTTGCTGTA-3′ (forward) and 5′-ACCACAGTCCATGC CATCAC-3′ (reverse), respectively. The reaction mixtures were subjected to PCR cycles of denaturation at 94°C for 1 min, annealing at 54–59°C for 45 sec, and extension at 72°C for 1.5 min. RT-PCR-amplified cDNAs were visualized on 1.2% agarose gels stained with ethidium bromide and the images were captured using UVP GDS-8000 BioImaging System (Ultra Violet Products, Cambridge, UK) for further analysis using Image Pro Plus 6.0 software. The integrated optical density (IOD) = area × average density of each gene expression band was measured and normalized to that of the corresponding GAPDH expression. The RNA expression levels in the cancer cells relative to those in MKN45-GFP cells were calculated.

Western blot analysis

The cancer cells were lysed by mixing with Radio-Immunoprecipitation Assay (cat. no. R0278) buffer containing Proteinase Inhibitor Cocktail (cat. no. P8340) from Sigma. The protein amounts in the cell lysates were quantified by bicinchoninic acid protein assay kit (Pierce, Rockford, IL, USA) and mixed with electrophoresis loading buffer (50 mM Tris-HCl, 2% sodium dodecyl sulfate (SDS), 0.1% bromophenol blue, 10% glycerol, and 1 mM dithiothreitol). The prepared samples were electrophoresed on 12% SDS-polyacrylamide gel under reducing conditions. After electrophoresis, the proteins were transferred electrophoretically to the PVDF membrane (Immobilon™-P) from Millipore. The membrane was blocked with 5% skim milk for 30 min at 37°C. After blocking, the membrane was incubated overnight at 4°C with 1:200 diluted rabbit polyclonal antibody H-300 (cat. no. sc-28215) against human fanconi anemia complementation group A (FANCA) and C-19 (cat. no. sc-540) against metallopanstimulin 1/TTK protein kinase (MPS1/TTK) from Santa Cruz (Santa Cruz, CA, USA); and 1:500 diluted mouse monoclonal antibodies clone 294216 (cat. no. MAB2050) against human melanoma-inhibitory activity (MIA) and clone 84728 (cat. no. MAB305) against IGFBP3 from R&D Systems (Minneapolis, MN, USA) in PBS-T buffer. The membrane was washed 3 times with PBS with 0.5% Triton X-100 buffer (PBS-T) for 5 min each, and then incubated with 1:5,000 diluted horse-radish peroxidase-conjugated bovine anti-rabbit IgGs (cat. no. sc-2370) from Santa Cruz or peroxidase-conjugated AffiniPure donkey anti-mouse IgGs (cat. no. 715035150) from Jackson ImmunoResearch (West Grove, PA, USA) in PBS-T buffer for 1 h at room temperature. After washing with PBS-T, the membrane was incubated with the Western Lightning™ ECL detecting reagent from Perkin Elmer (Waltham, MA, USA) and exposed to X-ray film (cat. no. 50880) from Fuji Medical (Tokyo, Japan). The images were captured by scanning exposed X-ray film using an HP Scanjet 4850 scanner (Hewlett-Packard, Palo Alto, CA, USA) and further analyzed by ImagePro Plus 6.0 software. The IOD = area × averaged density of each expressed protein band was measured and normalized to that of the corresponding GAPDH levels. The expressed protein levels in the cancer cells relative to those in MKN45-GFP cells were calculated.

Statistical analysis

Data were analyzed for significant differences using ANOVA followed by multiple comparisons with Student-Newman-Keuls test. A significant difference in the incidences of ascites and liver metastasis among the groups of nude mice was examined by using Fisher’s exact test. A difference in the averaged number of tumor nodules per mouse between the two mouse groups of cancer cells inoculated was detected using Wilcoxon signed rank test. All statistical analyses were conducted using SPSS® Statistics software from SPSS Inc. (Chicago, IL, USA). A significant difference between groups was set at p<0.05.

Results

Stable GFP-expressing MKN45 cells

Following transfection with GFP expression plasmids using lipofectamine 2000, human gastric cancer MKN45 cell clones constitutively expressing GFP were collected. Stable GFP-expressing MKN45 cell clones were selected by G418 in culture medium and a stable GFP-expressing cell clone was isolated by the limiting dilution process. A stable MKN45 cell clone with constitutive expression of the enhanced GFP, MKN45-GFP, was established (Fig. 1). There was no difference in morphology between the parental MKN45 and MKN45-GFP cells (Fig. 1A and B).

Establishment of MKN45-GFP cell sublines

The MKN45-GFP cells collected after the 4th, 10th, and 12th round of 72 h-selection periods using Transwell system with Matrigel-coated porous membrane were designated as MKN45-GFP-4, MKN45-GFP-10, and MKN45-GFP-12, respectively. The cell morphologies among MKN45-GFP and MKN45-GFP cell sublines were not different from each other as shown in Fig. 1B (MKN45-GFP) and 1D (MKN45-GFP-12). Furthermore, the cell proliferation doubling times of the parental MKN45 and the established MKN45-GFP cell sublines ranged from 1.4 to 1.7 days, as shown in Table I, and were not significantly different from each other (p=0.70).

Table I

Doubling time of MKN45 and MKN45-GFP sublines.

Table I

Doubling time of MKN45 and MKN45-GFP sublines.

Cell lineDoubling time (day)
MKN451.4±0.3*
MKN45-GFP1.7±0.1
MKN45-GFP-41.5±0.5
MKN45-GFP-101.7±0.4
MKN45-GFP-121.5±0.4

* Mean ± SD (n=6). No significant difference among the cells, p=0.70.

MKN45-GFP cell sublines with different in vitro invasion abilities

The three established MKN45-GFP sublines exhibited different invasive abilities in an increasing tendency, sequentially as MKN45-GFP, MKN45-GFP-4, MKN45-GFP-10 and MKN45-GFP-12. Fig. 2A–E shows representative images of hematoxylin-stained invaded cells on the surface of the porous membrane of the lower chamber of Transwell cultures. There was no difference in invasion ability between the parental MKN45 and MKN45-GFP cells, whereas significantly more MKN45-GFP-10 and MKN45-GFP-12 cells invaded through the membrane onto the other side of the membrane compared to MKN45 and MKN45-GFP cells. In comparison to that of MKN45-GFP, the invasion ability was significantly increased by 2-, 6-, and 6-fold for MKN45-GFP-4, MKN45-GFP-10 and MKN45-GFP-12 cells, respectively, as shown in Fig. 2F.

Incidences of ascites caused by MKN45- GFP and MKN45-GFP-12 cells in nude mice

The incidences of ascites in the MKN45-GFP and MKN45-GFP-12 cells inoculated nude mice were monitored as an index of tumorigenic aggressiveness and tumor burden caused by the cells. As the data summarized in Table II show, MKN45-GFP-12 cells caused a significant on-set of ascites on week 4 after the inoculation, which is one week earlier than that caused by MKN45-GFP cells. The higher incidence of ascites at week 4 caused by MKN45-GFP-12 indicated a higher tumorigenic aggressiveness of MKN45-GFP-12 than MKN45-GFP cells.

Table II

Incidence of ascites in the nude mice intraperitoneally inoculated with the cancer cells.

Table II

Incidence of ascites in the nude mice intraperitoneally inoculated with the cancer cells.

Week 2*Week 3Week 4Week 5

MKN45-GFP0/71/71/76/7
MKN45-GFP-120/71/76/7**6/7

* Time when animals were sacrificed following inoculation.

** p=0.015 vs. MKN45-GFP, Fisher’s exact test.

Orthotopic tumorigenesis and liver metastasis of MKN45-GFP and MKN45-GFP-12 cells in nude mice

Adult male nude mice intraperitoneally inoculated with MNK45-GFP or MKN45-GFP-12 cells were observed for intraperitoneal tumorigenesis and the numbers of tumor nodules were counted by visualizing the GFP-expressing tumors using a fluorescence imaging system. The orthotopic intraperitoneal inoculation of gastric cancer cells of diffuse type in mice is to mimic the dissemination and metastasis conditions of the cancer cells in the peritoneum of gastric cancer patients. After the inoculation, MKN45-GFP-12 tumor cells grew inside the peritoneal cavity and metastasized to and grew in the mouse liver. Representative images of the intraperitoneally disseminated growing MKN45-GFP-12 tumors in a nude mouse after laparotomy in bright and fluorescent fields are shown in Fig. 3A and B, respectively. Growing MKN45-GFP-12 tumors that invaded into the mouse liver are shown in Fig. 3C (bright field) and D (fluorescent field). MKN45-GFP cells also grew into tumors in the mouse peritoneum as shown in Fig. 3E (bright field) and F (fluorescent field). On Day 14 after the cancer cell inoculation, we observed that MKN45-GFP cells caused 14–25 (median 17; mean 18; n=7) intraperitoneally orthotopic growing tumors, whereas MKN45-GFP-12 cells caused 10–40 (median 25; mean 24; n=7) intraperitoneally orthotopic growing tumors in the laparotomized nude mice. The number of intraperitoneal tumor nodules caused by the orthotopic inoculation was compared and MKN45-GFP-12 cells were found to cause more, though not statistically significant (p=0.128), intraperitoneally disseminated growing tumors in the nude mice. Furthermore, both intraperitoneally inoculated MKN45-GFP and MKN45-GFP-12 cells caused metastatic tumor growth in the nude mouse livers during the observation period (Fig. 3G–I). MKN45-GFP-12 cells metastasized to the mouse liver and established metastatic growth in more nude mouse livers (p=0.042) than MKN45-GFP cells as detailed in the liver metastasis incidence summary in Table III. The results showed that MKN45-GFP-12 cells exhibited a higher metastasizing potential in vivo than MKN45-GFP cells.

Table III

Incidence of liver metastasis in the nude mice intraperitoneally inoculated with the cancer cells.

Table III

Incidence of liver metastasis in the nude mice intraperitoneally inoculated with the cancer cells.

No metastasisMetastasis

MKN45-GFP33/35 (94%)2/35 (6%)
MKN45-GFP-1227/35 (77%)8/35 (23%)*

* p=0.042 vs. MKN45-GFP, Fisher’s exact test.

cDNA microarray analyses of genes in MKN45-GFP cell sublines

Total RNAs from the established MKN45-GFP and the selected MKN45-GFP-4, MKN45-GFP-10, and MKN45-GFP-12 cells were cDNA microarray analyzed. The gene expression levels and patterns of MKN45-GFP-4, MKN45-GFP-10, and MKN45-GFP-12 cells were compared in relation to those of the MKN45-GFP cells (GEO accession number GSE33570). We used the PvalueLogRatio method to sieve out significant spots of whole microarray analysis. PvalueLogRatio gives the statistical significance on the log ratio for each spot between experimental and control signals. In order to focus on the desired data, we setup the criteria for removal steps and filtering procedure to identify positive or negative expression-correlated 514 genes (184 ascending and 330 descending genes) of the examined genes by hierarchical clustering calculation. We found that only <3% of all genes may be involved in human gastric cancer invasion and metastasis. Fig. 4 displays a hierarchical clustering analysis image with 514 genes (accounting for 2.75% of the total number of 18,716 genes examined) from significant expression levels by using the selection criteria described above, which share a similar tendency containing 184 ascending (positively correlated with invasiveness) and 330 descending genes (negatively correlated with invasiveness). The expression levels were pseudo-color encoded showing the descending and ascending expression levels of genes in the MKN45-GFP cell sublines. Based on the hierarchical clustering analyses, the expression patterns of these 514 genes were subjected to regulating tendency selection processes.

The 514 genes identified in the hierarchical clustering analysis were further categorized into groups such as angiogenesis-related genes, cell cycle regulators, cytoskeleton and motility molecules, protease and adhesion proteins, and signal transduction molecules, based on known molecular and biological functions associated with tumor progression. A total of 133 genes were found to be associated with these biological functions and 30 of them are shown in Fig. 5 and Table IV. The 5 selective groups were (A) angiogenesis-related genes such as angiogenic inducer Cyr61 and vascular endothelial growth factor; (B) cell cycle regulators such as MPS1/TTK and cyclin B2; (C) cytoskeleton and motility molecules such as OTR and catenin α-like 1; (D) protease and adhesion molecules such as laminin γ 2 and collagen type XII α 1, and (E) signal transduction molecules such as LGR4/GPR48 and heparin-binding growth factor binding protein. Genes associated with multiple biological functions were included in the corresponding categorized functional groups accordingly. A number of genes that have not been previously reported to correlate with cancer progression or tumor metastasis were also identified and are listed in Table V. The levels of these genes differed more than two-fold between highly metastatic and parental cells, indicating that these genes may play significant roles in gastric cancer metastasis.

Table IV

Genes of known functions differentially expressed between parental and highly metastatic MKN45 cell lines.

Table IV

Genes of known functions differentially expressed between parental and highly metastatic MKN45 cell lines.

Probe nameAccession no.Sequence homology (GenBank/EMBL)Gene symbolChromosome location Log2[E12/Ep]
Angiogenesis related genes

A_23_P257296NM_003226Trefoil factor 3TFF321q22.32.74
A_23_P132987NM_002619Platelet factor 4PF44q12-q212.06
A_23_P387668NM_003376Vascular endothelial growth factorVEGFA6p121.40
A_23_P87564NM_001731B-cell translocation gene 1, anti-proliferativeBTG112q221.24
A_23_P421423NM_006291Tumor necrosis factor, α-induced protein 2TNFAIP214q32−1.76
A_23_P46429NM_001554Cystein-rich, angiogenic inducer, 61CYR611p31-p22−2.12

Cell cycle regulation related genes

A_23_P22735NM_032621Brain expressed X-linked 2BEX2Xq223.99
A_23_P122197NM_031966Cyclin B1CCNB15q12−1.56
A_23_P80032NM_005225E2F transcription factorE2F120q11.2−1.66
A_23_P65757NM_004701Cyclin B2CCNB215q22.2−1.78
A_23_P259586NM_003318Metallopanstimulin 1TTK6q13-q21−1.85
A_23_P139881NM_001759Cyclin D2CCND212p13−2.33

Cytoskeleton and motility related genes

A_23_P19663NM_001901Connective tissue growth factorCTGF6q23.11.56
A_23_P254626NM_003919Sarcoglycan, epsilonSGCE7q21-q221.46
A_23_P374844NM_015973GalaninGAL11q13.3−1.52
A_23_P121533NM_012445Spondin 2, extracellular matrix proteinSPON24p16.3−1.57
A_23_P96149NM_000422Keratin 17KRT1717q12-q21−1.59
A_23_P157795NM_003798Catenin, α-like 1CTNNAL19q31.2−2.14
A_23_P132619NM_000916Oxytocin receptorOXTR3p25−2.90

Protease and cell adhesion related genes

A_23_P257296NM_003226Trefoil factor 3TFF321q22.32.74
A_23_P19663NM_001901Connective tissue growth factorCTGF6q23.11.56
A_23_P254626NM_003919Sarcoglycan, epsilonSGCE7q21-q221.46
A_23_P387668NM_003376Vascular endothelial growth factorVEGFA6p121.40
A_23_P121533NM_012445Spondin 2, extracellular matrix proteinSPON24p16.3−1.57
A_23_P6935NM_001777CD47 antigenCD473q13.1-q13.2−1.89
A_23_P201636NM_005562Laminin, gamma 2LAMC21q25-q31−2.00
A_23_P118815NM_001168Baculoviral IAP repeat-containing 5BIRC517q25−2.12
A_23_P157795NM_003798Catenin, α-like 1CTNNAL19q31.2−2.14
A_23_P218441NM_002483Carcinoembryonic antigene-related CAM 6CEACAM619q13.2−2.61
A_23_P214168NM_004370Collagen, type XII, α 1COL12A16q12-q13−2.74

Signal transduction related genes

A_23_P42869NM_000596Insulin-like growth factor binding protein 1IGFBP17p13-p124.19
A_23_P215634NM_000598Insulin-like growth factor binding protein 3IGFBP37p13-p124.10
A_23_P4714NM_006533Melanoma inhibitory activityMIA 19q13.32-q13.332.73
A_23_P65918NM_002220Inositol 1,4,5-trisphosphate 3-kinase AITPKA15q14-q212.44
A_23_P9571NM_018098ECT sequence 2 oncogeneECT23q26.1-q26.2−1.13
A_23_P203767NM_018490G protein-coupled receptor 48LGR411p14-p13−1.35
A_23_P206441NM_000135Fanconi anemia, complementation group AFANCA16q24.3−1.38
A_23_P374844NM_015973GalaninGAL11q13.3−1.52
A_23_P6935NM_001777CD47 antigenCD473q13.1-q13.2−1.89
A_23_P218441NM_002483Carcinoembryonic antigene-related CAM 6CEACAM619q13.2−2.61
A_23_P132619NM_000916Oxytocin receptorOXTR3p25−2.90
A_23_P30126NM_005130FGF binding proteinFGFBP14p16-p15−3.20

[i] Log2[E12/Ep] corresponds to 2 logarithm of mean expression ration of MKN45-GFP-12 (E12) to MKN45 parental cell line (Ep).

Table V

Novel genes of unknown functions differentially expressed between parental and highly metastatic MKN45 cell lines.

Table V

Novel genes of unknown functions differentially expressed between parental and highly metastatic MKN45 cell lines.

Probe nameAccession no.Sequence homology (GenBank/EMBL)Gene symbolChromosome location Log2[E12/Ep]
A_23_P209360NM_052920Kelch-like 29KLHL292p24.13.26
A_23_P20876NM_152422Protein tyrosine phosphatase domain containing 1PTPDC19q22.322.51
A_23_P83339NM_145051Ring finger protein 183RNF1839q322.14
A_23_P108823NM_145739Oxysterol binding protein-like 6OSBPL62q32.11.92
A_23_P154740NM_018474Polo-like kinase 1 substrate 1PLK1S120p11.231.80
A_23_P79302NM_177964LY6/PLAUR domain containing 6BLYPD6B2q23.21.44
A_23_P52727NM_182964Neuron navigator 2NAV211p15.1−1.49
A_23_P51966NM_003035SCL/TAL1 interrupting locusSTIL1q32; 1p32−1.55
A_23_P379778NM_017594DIRAS family, GTP-binding RAS-like 2DIRAS29q22.2−1.56
A_23_P69179NM_018192Leprecan-like 1LEPREL13q28−1.57
A_23_P340909NM_145061Spindle and kinetochore associated complex subunit 3SKA313q12.11−1.66
A_23_P118582NM_013290PSMC3 interacting proteinPSMC3IP17q21.2−1.70
A_23_P48835NM_138555Kinesin family member 23KIF2315q23−1.80
A_23_P166526NM_015653RIB43A domain with coiled-coils 2RIBC222q13.31−1.90
A_23_P80718NM_144642SynaptoporinSYNPR3p14.2−1.92
A_23_P49878NM_019013Family with sequence similarity 64, member AFAM64A17p13.2−1.93
A_23_P323751NM_030919Family with sequence similarity 83, member DFAM83D20q11.22-q12−1.95
A_23_P42811NM_176813Anterior gradient homolog 3AGR37p21.1−2.06
A_23_P388812NM_152515Cytoskeleton associated protein 2-likeCKAP2L2q13−2.07
A_23_P25964NM_000153 GalactosylceramidaseGALC14q31−2.16
A_23_P355525NM_173497HECT domain containing 2HECTD210q23.32−2.39
A_23_P250853N/ASM3A_HUMAN (Q14563) Semaphorin 3A precursor (Semaphorin III) (Sema III), partial (8%)THC2052903N/A−2.41
A_23_P35995NM_024769Adipocyte-specific adhesion moleculeASAM11q24.1−2.49
A_23_P3302NM_018365Meiosis-specific nuclear structural 1MNS115q21.3−2.50
A_23_P217785NM_016500Chromosome X open reading frame 26CXorf26Xq13.3−2.54
A_23_P130027NM_017957Epsin 3EPN317q21.33−2.60
A_23_P301360NM_152412Zinc finger protein 572ZNF5728q24.13−2.62
A_23_P254842NM_012080Haloacid dehalogenase-like hydrolase domain containing 1HDHD1Xp22.32−2.68
A_23_P329254NM_153032Archaelysin family metallopeptidase 2 pseudogene 1AMZ2P117q24.1−2.69
A_23_P35871NM_024680E2F transcription factor 8E2F811p15.1−2.75
A_23_P154279NM_032309 Coiled-coil-helix-coiled-coil-helix domain containing 5CHCHD52q13−3.01
A_23_P252432NM_004617Transmembrane 4 L six family member 4TM4SF43q25−3.06
A_23_P75915NM_024557Resistance to inhibitors of cholinesterase 3 homolog (C. elegans)RIC311p15.4−3.06
A_23_P252388NM_024867Sperm flagellar 2SPEF25p13.2−3.20
A_23_P396765NM_173582Phosphoglucomutase 2-like 1PGM2L111q13.4−3.41
A_23_P139604NM_003805CASP2 and RIPK1 domain containing adaptor with death domainCRADD12q21.33-q23.1−4.71

[i] Log2[E12/Ep] corresponds to 2 logarithm of mean expression ration of MKN45-GFP-12 (E12) to MKN45 parental cell line (Ep).

mRNA transcript levels in MKN45-GFP cell sublines

Several putative tumor metastasis-associated genes were further examined by using RT-PCR analysis. Four genes, TFF3, BEX2, SGCE and IGFBP3, positively correlated with the invasion ability, and two genes, OTR and LGR4/GPR48, negatively correlated with the invasion ability of the gastric cancer MKN45-GFP cell sublines, were RT-PCR analyzed. The housekeeping gene GAPDH was selected as the internal control. Results showed that all four examined genes TFF3, BEX2, SGCE, and IGFBP3 exhibited an increasing order of the mRNA transcript levels from MKN45-GFP, MKN45-GFP-4, MKN45-GFP-10, to MKN45-GFP-12 cells. On the other hand, mRNA transcript levels of the two genes OTR and LGR4/GPR48 were in a decreasing order in these MKN45-GFP cell sublines in relation to their invasion ability (Fig. 6). Results of the RT-PCR analyses on these selective genes were consistent with those from the cDNA microarray analyses.

Protein expression levels in MKN45-GFP cell sublines

The expression levels of several proteins of the microarray-identified genes were also examined using western blot analysis. FANCA, MPS1/TTK, MIA, and IGFBP3 protein levels in the MKN45-GFP cell sublines were analyzed in triplicates. FANCA and MPS1/TTK protein levels were downregulated with the increase of the invasive potentials of the MKN45-GFP cell sublines. By contrast, the protein levels of (precursor and/or mature) MIA and IGFBP3 were upregulated with the increase of the invasive potentials of the MKN45-GFP cell sublines (Fig. 7). Both the mRNA transcript and protein expression levels of the IGFBP3 gene were positively correlated with the invasive abilities of the MKN45-GFP cell sublines. The results demonstrated that the expression levels of these proteins examined were consistent with those from the cDNA microarray analyses.

Discussion

It is well known that differential expression of multiple genes and dynamic interactions of a number of cellular proteins are involved in the multiple progressive steps of cancer metastasis including gastric cancer. cDNA microarray has been applied to explore the expression profiles of a massive number of genes between testing cancer samples. In the present study, we used cDNA microarray to identify gastric cancer invasion/metastasis-associated genes on a genome-wide scale in human gastric cancer sublines that have demonstrated different meta-static abilities. As the poorly differentiated diffuse type gastric cancer remains an unmet medical need, a gastric cancer cell line of diffuse type MKN45, originally isolated from a poorly differentiated adenocarcinoma of a patient’s stomach (33,34), was used as the model cell. By taking advantage of the fluorescence-emitting ability of GFP, we established an MKN45 cell line constitutively expressing GFP which can be visualized, and applied it to select the MKN45-GFP sublines with differential invasion abilities by using the Transwell system. Genes in relation to the invasiveness of these sublines were identified using cDNA microarray analysis and verified by RT-PCR and western blot analyses. Furthermore, the invasiveness of the highly invasive subline MKN45-GFP-12 was in vivo demonstrated by the incidence of ascites and organ invasive abilities in nude mice. Meanwhile, we observed that a number of the identified genes are also related to angiogenesis, cell cycle, cytoskeleton and motility, protease and cell adhesion, and signal transduction, which are cellular events known to be associated with invasion and tumor metastasis (39). Accordingly, a number of novel genes without previously known cancer-related biological activities were also identified in relation to gastric tumor metastasis in the present study.

Systemically analyzed using genomic microarray technologies and biological activity assays, genes putatively associated with tumor cell invasion and metastasis have been identified in various human cancer cells, including gastric cancer cells. Many studies utilized microarray technologies to discover genes specifically associated with tumor invasion and metastasis. Hippo et al observed a number of differentially expressed genes by using a set of human gastric scirrhous cancer cells prone to peritoneal and lymph node metastasis in nude mice (23). Although the array-based oligonucleotide hybridization method is a powerful tool for identifying potential genes associated with tumor metastasis, the genes reported in these above-mentioned studies are different from each other. The intrinsic heterogeneity of the cancer cell populations in patient tumor tissues likely mixed with normal stromal and/or vascular cells may contribute to these observed discrepancies in the gene expression profiles (40). Herein we used a human gastric cancer cell line of diffuse type in vitro to normalize and minimize the potentially existing heterogeneity in order to identify human gastric tumor metastasis-associated genes.

Chronic infection of Helicobacter P. has been proven to be an important etiologic factor of gastric cancer (41) and accounts for 18% of all cancers caused by infections (42). Helicobacter infection has been found to be associated with β-catenin, MMP-1, ERK (p38), EGFR, AP-1, Shh, CDX2, E-cadherin (CDH1), p16, APC, MLH1, COX2, IL-1, IL-6, IL-10, CTLR4, BCL-2, FOXP1 and p53 (43), and IL-8, ras family and Ras p21 are involved in the TNFα/NF-κB/IκB-mediated gastric tumorigenesis (44). Helicobacter colonization is usually asymptomatic and gastric tumorigenesis occurs only in a subset of individuals depending on the host response and genetic variation of the bacteria. The infection alone could not attribute entirely to the gastric tumorigenesis as the clinically observed human gastric cancers are genetically and histologically diversifying (43,45). Although several studies reported that Helicobacter infection increases tumor invasiveness and metastasis via multiple molecular pathways (4650), in the present study we did not find significant changes in the expressions of these mentioned genes and/or the genes in the TNFα/NF-κB/IκB-mediated signaling pathways (43,44). Therefore, the results suggest that the genes identified in the present study are not Helicobacter infection-related and are thus directly correlated to the metastatic abilities of human gastric cancer tumors.

GFP has been utilized as a reporter molecule and has been widely applied in vitro and in vivo to visualize the developmental processes of tumors, including primary and metastatic tumor growths, tumor cell metastases, tumor angiogenesis, and interactions between tumor and the host microenvironment (38,5154). Chu et al established a set of human lung cancer cells with differential invasive abilities (35), Chen et al identified a number of genes associated with human lung cancer metastasis (55), and Chen et al demonstrated a significant clinical association for some of these identified genes (56). Accordingly, we took advantage of the fluorescence-emitting GFP and established the human GFP-expressing MKN45 sublines with differential invasive abilities in the present study. The morphology and proliferation rates were not different from each other among these established MKN45-GFP sublines including the parental MKN45 cells. One of the important advantages is that we are able to monitor the tumor growth and peritoneal dissemination in individual nude mice without laparotomy. The appropriate timing for animal euthanization can then easily be determined and therefore the number of animals used in a study can be reduced. The identification of the genes previously known with biological activities related to tumor progression and metastasis suggests the validity of the approach utilized in this study. The observations indicate that the genes differentially expressed among these sublines are likely to be associated with their invasive and, thus, metastatic abilities.

In the present study, 184 and 330 genes were found positively and negatively expressed, respectively, with regard to the increasing order of the invasive abilities of these model cell sublines. The identified genes are likely to be associated with cancer progression and/or tumor metastasis. They are found to be associated with the biological activities pertaining to tumor metastasis such as angiogenesis-, cell cycle regulation-, cytoskeleton and motility-, protease and cell adhesion-, and signal transduction-related genes as shown in Fig. 5 and Table IV. Angiogenesis is one of the essential steps of tumor growth and metastasis (39). Proteins of the trefoil factor family genes may induce tumor vascularity and be associated with tumor metastatic phenotype and recurrence in human gastric cancer (57). Trefoil factor 3 (TFF3) is connected with multiple oncogenic pathways such as COX2 and EGFR and participates in the blood vessel formation during tumor progression (58). IGFBP3 is a protein of multi-functions. IGFBP3 induces IGF-independent anti-angiogenic effects and exerts tumor suppressive effects against prostate cancer (59) and non-small cell lung cancer (60), whereas its overexpression is associated with the increasing metastatic ability of well-differentiated pancreatic endocrine tumors (61). We report here that IGFBP3 expression is positively correlated with the invasive ability of human gastric cancer cells suggesting that IGFBP3 is involved in gastric cancer metastasis. In agreement with a previous study (62), we found that vascular endothelial growth factor (VEGF) genes were positively regulated in accordance with the development of tumor invasiveness. Overexpression of B-cell translocation gene 1 (BTG1) increases tube formation and migration of the endothelial cells (63). Accordingly, we observed that BTG1 is upregulated during the development of human gastric cancer cell invasiveness. Further studies are required to explore the pathological roles of these angiogenesis-related genes during gastric tumor progression.

During cell cycle progression, MPS1/TTK is a protein kinase and is involved in the regulation of centrosomal control of cytokinesis in the mitotic checkpoint process (64). MPS1/TTK kinase affects the chromosomal stabilities in the M phase of the cell cycles in zebra fish and mammalian cells (65,66). Chromosomal instability may induce changes in the cellular functions and thus increase the invasive and metastatic abilities of tumor cells (67). In accordance with the implications of these observations, we found that MPS1/TTK expression was downregulated in the invasive cell sublines. BEX2 participates in G1 phase regulation during cell cycle progression and its over-expression protects breast cancer cells against mitochondrial apoptosis (68). We herein observed that upregulation of BEX2 was associated with the development of the invasion and metastasic abilities of gastric tumor cells.

Cellular cytoskeleton and motility also play important roles in tumor cell metastasis. Connective tissue growth factor (CTGF), whose transcript levels were moderately increased, as shown in the present study, modulates the motility of human breast cancer cells through activation of the ERK1/2 signaling pathway (69). Catenin-α is anti-metastatic in human squamous carcinoma and breast cancer by forming a complex with E-cadherin and thus enhancing the cell-cell adhesion (70,71). In agreement with these observations, catenin-α was down-regulated in the invasive cell sublines in this study. Oxytocin receptor was found in the smooth muscles of the gastric/intestinal system and mediates gastric motility (72). A previous study showed that oxytocin inhibited ovarian carcinoma cell metastasis in vitro and in vivo (73). Furthermore, we observed that oxytocin receptors are downregulated in the highly metastatic gastric cancer cell sublines. The results suggest that the oxytocin/oxytocin receptor system may play a significant role in gastric cancer metastasis.

Proteases and adhesion proteins play key roles in gastric tumor metastasis. The ability of tumor cells to detach from a primary tumor tissue and degradation of the extracellular matrix and basement membrane structures are associated with the progress of tumor cell invasion and metastasis. TFF3 modulates E-cadherin/catenin-mediated cell-cell contact and increases cell motility in cancer progression (74). We found that TFF3 was upregulated in the gastric cancer cells with high invasive ability. Involvement of SGCE in adhesion activity has been reported to inhibit the invasion of colorectal tumors (75). Contrary to that observed in colorectal tumors, SGCE expression was positively correlated with the gastric cancer cell invasion ability shown in the present study. Laminin family proteins involved in the cell-laminin interactions are related to tumor angiogenesis, invasion and metastasis (76). We found that laminin-γ was downregulated in the gastric cancer cells of high invasiveness. Collagen XII is an adhesion protein reported to have critical roles in the interactions between cancer cells and bone marrow, in cell adhesion, and in cell motility and is thus involved in the bone metastasis of prostate cancer (77). In gastric cancer cells, we observed that collagen XII expression is inversely correlated with cancer cell invasion ability.

Cellular and subcellular signaling pathways regulate multiple and complex cellular activities, including tumor cell proliferation, invasion and metastasis. IGFBP3 plays significant roles in tumor cell proliferation, invasion and metastasis in different human cancer diseases. Compared to the primary melanoma cells, IGFBP3 was overexpressed in the metastatic melanoma cells and may serve as a biomarker in the early diagnosis of melanoma (78). On the other hand, Torng et al reported that IGFBP3 plays a major role as an invasion-metastasis suppressor via an insulin growth factor-independent pathway in human ovarian endometrioid carcinomas (79). These previous observations suggested possible dual biological roles of the IGFBP3 signal pathways in various cancers (5961,78,79). Nonetheless, our results showed a positive correlation between IGFBP3 protein expression and metastasis potential of the gastric cancer cells. Contrary to that reported in colorectal cancer HCT116 cells (80), we found in gastric cancer MKN45 cells that LGR4 expression was inversely correlated with the cell invasion ability.

A number of the genes identified in our studies have been verified for their transcriptional or translational activities in the mRNA or protein levels by semi-quantified RT-PCR and western blotting, respectively, such as TFF3, IGFBP3, BEX2, OTR, SGCE, LGR4, FANCA, MIA, and MPS1/TTK. FANCA has been reported to participate in the BRCA signaling pathway and is involved in the chromosome stability of cancer cells (81). MIA proteins binding to integrins promote detachment of melanoma cells from extracellular matrix structures and increase the migration ability of the cells, which could be inhibited by treatments with chemicals (82). MIA may play an important role in gastric tumor cell invasion and metastasis. The mRNA or protein expression profiles verified in the present study are in agreement with those previously observed.

In addition, we have also identified a number of novel genes, listed in Table V, using the same evaluation processes. These novel genes have not been previously demonstrated with any biological activity nor are they known with any cancer progression-related or tumor metastasis-associated activities. Similarly, these novel genes are likely to be involved in important biological processes in the development and progression of human gastric tumor invasion and metastasis. Their possible cellular/subcellular mechanisms involving tumor progression or metastasis are to be further explored in the future. By using gene-silencing approaches such as siRNA treatments to the MKN45-GFP-12 cells, one may further investigate the functional roles of the pro-metastasis genes in the tumor metastatic process and their underlying molecular mechanisms. For the genes against metastasis, one can restore the downregulated gene functions by gene transfection in MKN45-GFP-12 cells to demonstrate a decrease in their metastatic abilities. Furthermore, one can look into the gene expression levels in the clinical patient specimens and search for clinical relevance in terms of tumor progression as well as patient survival.

Furthermore, we established a highly aggressive MKN45-GFP-12 cell line and adopted an orthotopic model in which human gastric cancer cells of diffuse type were intraperitoneally inoculated in nude mice to mimic the pathological condition of peritoneal metastasis in patients. The malignancies of MKN45-GFP and MKN45-GFP-12 cells were compared using the orthotopic model. Compared to MKN45-GFP cells, MKN45-GFP-12 cells tend to invade internal organs, such as the liver, and to cause ascites at a shorter onset time, indicating a higher malignant property of the MKN45-GFP-12 cells. More tumor nodules were growing in the abdominal cavity of MKN45-GFP-12-inoculated nude mice at Day 14 (data not show). By using an in vivo selection process, we previously established a human gastric cancer cell line MKN45-GFP-ip4 (38). Whether the two malignant cell lines MKN45-GFP12 and MKN45-GFP-ip4 established via the in vitro and in vivo selections, respectively, share a similar gene expression profile requires further investigation.

In this study, we established several human diffuse type gastric cancer cell sublines with different invasion abilities and identified a number of putative gastric tumor metastasis-associated genes, including novel genes without any known biological activity (GEO accession number GSE33570). These genes and protein products are to be further investigated for their roles in the molecular and cellular actions related to tumor metastasis. The molecular profiles of these identified genes, gene transcripts, and/or proteins in the patient specimens are likely to be useful for diagnostic, therapeutic, and/or prognostic purposes. In particular, they may be used as molecular targets for the development of drugs against tumor metastasis. These further research efforts could ultimately lead to the identification of tumor metastasis-associated molecules from which diagnostics and therapeutics for the human gastric cancer may be discovered.

Acknowledgements

This study was supported by the following grants, NHRIBP-091-CF03, NHRI-BP-093-CB06, NHRI-BP-094-PP11, NHRI-BP-094-PP13, NHRI-095-PP07, and NSC-92-2218-E-400-001, awarded to C.T. Chen from The National Health Research Institutes, Miaoli and The National Science Council, Taiwan, R.O.C.

References

1. 

Archie V, Kauh J, Jones DV Jr, Cruz V, Karpeh MS Jr and Thomas CR Jr: Gastric cancer: standards for the 21st century. Crit Rev Oncol Hematol. 57:123–131. 2006. View Article : Google Scholar : PubMed/NCBI

2. 

Brenner H, Rothenbacher D and Arndt V: Epidemiology of stomach cancer. Methods Mol Biol. 472:467–477. 2009. View Article : Google Scholar

3. 

Yeh KH and Cheng AL: Recent advances in therapy for gastric cancer. J Formos Med Assoc. 103:171–185. 2004.

4. 

Parkin DM: The global health burden of infection-associated cancers in the year 2002. Int J Cancer. 118:3030–3044. 2006.PubMed/NCBI

5. 

Vital and Health Statistics Division, Statistics and Information Department, Ministry of Health, Labour and Welfare: Vital statistics in Japan (1950–2007.

6. 

Horner MJ, Ries LAG, Krapcho M, Neyman N, Aminou R, Howlader N, Altekruse SF, Feuer EJ, Huang L, Mariotto A, Miller BA, Lewis DR, Eisner MP, Stinchcomb DG and Edwards BK: SEER Cancer statistics review, 1975–2006. National Cancer Institute; Bethesda: 2008

7. 

Lee KH, Lee JH, Cho JK, Kim TW, Kang YK, Lee JS, Kim WK, Chung JG, Lee IC and Sun HS: A prospective correlation of Laurén’s histological classification of stomach cancer with clinicopathological findings including DNA flow cytometry. Pathol Res Pract. 197:223–229. 2001.

8. 

Whiting J, Sano T, Saka M, Fukagawa T, Katai H and Sasako M: Follow-up of gastric cancer: a review. Gastric Cancer. 9:74–81. 2006. View Article : Google Scholar : PubMed/NCBI

9. 

The EUROCARE-4 database on cancer survival in Europe. [http://www.eurocare.it/Results/tabid/79/Default.aspx#eu4dB].

10. 

Guarino M: Epithelial-mesenchymal transition and tumour invasion. Int J Biochem Cell Biol. 39:2153–2160. 2007. View Article : Google Scholar : PubMed/NCBI

11. 

Wu CY, Wu MS, Chiang EP, Wu CC, Chen YJ, Chen CJ, Chi NH, Chen GH and Lin JT: Elevated plasma osteopontin associated with gastric cancer development, invasion and survival. Gut. 56:782–789. 2007. View Article : Google Scholar : PubMed/NCBI

12. 

Kuniyasu H, Yasui W, Yokozaki H, Kitadai Y and Tahara E: Aberrant expression of c-met mRNA in human gastric carcinomas. Int J Cancer. 55:72–75. 1993. View Article : Google Scholar : PubMed/NCBI

13. 

Ran Y, Peng L, Hu H, Yu L, Liu Q, Zhou Z, Sun YM, Sun LC, Pan J, Sun LX, Zhao P and Yang ZH: Secreted LOXL2 is a novel therapeutic target that promotes gastric cancer metastasis via the Src/FAK pathway. Carcinogenesis. 30:1660–1669. 2009. View Article : Google Scholar : PubMed/NCBI

14. 

Kim JH, Kim MA, Lee HS and Kim WH: Comparative analysis of protein expressions in primary and metastatic gastric carcinomas. Hum Pathol. 40:314–322. 2009. View Article : Google Scholar : PubMed/NCBI

15. 

Gravalos C and Jimeno A: HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol. 19:1523–1529. 2008. View Article : Google Scholar : PubMed/NCBI

16. 

Sakashita K, Mimori K, Tanaka F, Kamohara Y, Inoue H, Sawada T, Hirakawa K and Mori M: Prognostic relevance of Tensin4 expression in human gastric cancer. Ann Surg Oncol. 15:2606–2613. 2008. View Article : Google Scholar : PubMed/NCBI

17. 

Yu G, Wang J, Chen Y, Wang X, Pan J, Li G, Jia Z, Li Q, Yao JC and Xie K: Overexpression of phosphorylated mammalian target of rapamycin predicts lymph node metastasis and prognosis of Chinese patients with gastric cancer. Clin Cancer Res. 15:1821–1829. 2009. View Article : Google Scholar : PubMed/NCBI

18. 

Nakayama H, Yasui W, Yokozaki H and Tahara E: Reduced expression of nm23 is associated with metastasis of human gastric carcinomas. Jpn J Cancer Res. 84:184–190. 1993. View Article : Google Scholar : PubMed/NCBI

19. 

Okada K, Shimura T, Suehiro T, Mochiki E and Kuwano H: Reduced galectin-3 expression is an indicator of unfavorable prognosis in gastric cancer. Anticancer Res. 26:1369–1376. 2006.PubMed/NCBI

20. 

Li X, Zhang Y, Cao S, Chen X, Lu Y, Jin H, Sun S, Chen B, Liu J, Ding J, Wu K and Fan D: Reduction of TIP30 correlates with poor prognosis of gastric cancer patients and its restoration drastically inhibits tumor growth and metastasis. Int J Cancer. 124:713–721. 2009. View Article : Google Scholar : PubMed/NCBI

21. 

Hsu PI, Hsieh HL, Lee J, Lin LF, Chen HC, Lu PJ and Hsiao M: Loss of RUNX3 expression correlates with differentiation, nodal metastasis, and poor prognosis of gastric cancer. Ann Surg Oncol. 16:1686–1694. 2009. View Article : Google Scholar : PubMed/NCBI

22. 

Kim DY, Joo JK, Park YK, Ryu SY, Kim HS, Noh BK, Lee HK and Lee HJ: E-cadherin expression in early gastric carcinoma and correlation with lymph node metastasis. J Surg Oncol. 96:429–435. 2007. View Article : Google Scholar : PubMed/NCBI

23. 

Hippo Y, Yashiro M, Ishii M, Taniguchi H, Tsutsumi S, Hirakawa K, Kodama T and Aburatani H: Differential gene expression profiles of scirrhous gastric cancer cells with high metastatic potential to peritoneum or lymph nodes. Cancer Res. 61:889–895. 2001.PubMed/NCBI

24. 

Wang J and Chen S: Screening and identification of gastric adenocarcinoma metastasis-related genes using cDNA micro-array coupled to FDD-PCR. J Cancer Res Clin Oncol. 128:547–553. 2002. View Article : Google Scholar

25. 

Kim JM, Sohn HY, Yoon SY, Oh JH, Yang JO, Kim JH, Song KS, Rho SM, Yoo HS, Kim YS, Kim JG and Kim NS: Identification of gastric cancer-related genes using a cDNA microarray containing novel expressed sequence tags expressed in gastric cancer cells. Clin Cancer Res. 11:473–482. 2005.PubMed/NCBI

26. 

Myllykangas S, Junnila S, Kokkola A, Autio R, Scheinin I, Kiviluoto T, Karjalainen-Lindsberg ML, Hollmén J, Knuutila S, Puolakkainen P and Monni O: Integrated gene copy number and expression microarray analysis of gastric cancer highlights potential target genes. Int J Cancer. 123:817–825. 2008. View Article : Google Scholar : PubMed/NCBI

27. 

Hippo Y, Taniguchi H, Tsutsumi S, Machida N, Chong JM, Fukayama M, Kodama T and Aburatani H: Global gene expression analysis of gastric cancer by oligonucleotide microarrays. Cancer Res. 62:233–240. 2002.PubMed/NCBI

28. 

Inoue H, Matsuyama A, Mimori K, Ueo H and Mori M: Prognostic score of gastric cancer determined by cDNA microarray. Clin Cancer Res. 8:3475–3479. 2002.PubMed/NCBI

29. 

Hasegawa S, Furukawa Y, Li M, Satoh S, Kato T, Watanabe T, Katagiri T, Tsunoda T, Yamaoka Y and Nakamura Y: Genome-wide analysis of gene expression in intestinal-type gastric cancers using a complementary DNA microarray representing 23,040 genes. Cancer Res. 62:7012–7017. 2002.PubMed/NCBI

30. 

Yang S, Jeung HC, Jeong HJ, Choi YH, Kim JE, Jung JJ, Rha SY, Yang WI and Chung HC: Identification of genes with correlated patterns of variations in DNA copy number and gene expression level in gastric cancer. Genomics. 89:451–459. 2007. View Article : Google Scholar : PubMed/NCBI

31. 

Chang W, Ma L, Lin L, Gu L, Liu X, Cai H, Yu Y, Tan X, Zhai Y, Xu X, et al: Identification of novel hub genes associated with liver metastasis of gastric cancer. Int J Cancer. 125:2844–2853. 2009. View Article : Google Scholar : PubMed/NCBI

32. 

Oue N, Aung PP, Mitani Y, Kuniyasu H, Nakayama H and Yasui W: Genes involved in invasion and metastasis of gastric cancer identified by array-based hybridization and serial analysis of gene expression. Oncology. 69:17–22. 2005. View Article : Google Scholar : PubMed/NCBI

33. 

Motoyama T, Hojo H and Watanabe H: Comparison of seven cell lines derived from human gastric carcinomas. Acta Pathol Jpn. 36:65–83. 1986.PubMed/NCBI

34. 

Yokozaki H: Molecular characteristics of eight gastric cancer cell lines established in Japan. Pathol Int. 50:767–777. 2000. View Article : Google Scholar : PubMed/NCBI

35. 

Chu YW, Yang PC, Yang SC, Shyu YC, Hendrix MJ, Wu R and Wu CW: Selection of invasive and metastatic subpopulations from a human lung adenocarcinoma cell line. Am J Respir Cell Mol Biol. 17:353–360. 1997. View Article : Google Scholar : PubMed/NCBI

36. 

Huang YC, Chen CT, Chen SC, Lai PH, Liang HC, Chang Y, Yu LC and Sung HW: A natural compound (Ginsenoside Re) isolated from Panax Ginseng as a novel angiogenic agent for tissue regeneration. Pharm Res. 22:636–646. 2005. View Article : Google Scholar : PubMed/NCBI

37. 

Li WT, Hwang DR, Chen CP, Shen CW, Huang CL, Chen TW, Lin CH, Chang YL, Chang YY, Lo YK, et al: Synthesis and biological evaluation of N-heterocyclic indolyl glyoxylamides as orally active anticancer agents. J Med Chem. 46:1706–1715. 2003. View Article : Google Scholar : PubMed/NCBI

38. 

Tuan TF, Tsai ML, Yeh KC, Huang HC, Chung CT, Huang CL, Han CH, Chen CP, Wang MH, Shen CC, et al: Intravenous paclitaxel against metastasis of human gastric tumors of diffuse type. Cancer Chemother Pharmacol. 66:773–783. 2010. View Article : Google Scholar : PubMed/NCBI

39. 

Geiger TR and Peeper DS: Metastasis mechanisms. Biochim Biophys Acta. 1796:293–308. 2009.PubMed/NCBI

40. 

Chen X, Leung SY, Yuen ST, Chu KM, Ji J, Li R, Chan AS, Law S, Troyanskaya OG, Wong J, So S, Botstein D and Brown PO: Variation in gene expression patterns in human gastric cancers. Mol Biol Cell. 14:3208–3215. 2003. View Article : Google Scholar : PubMed/NCBI

41. 

Hansson LE, Engstrand L, Nyrén O, Evans DJ Jr, Lindgren A, Bergström R, Andersson B, Athlin L, Bendtsen O and Tracz P: Helicobacter pylori infection: independent risk indicator of gastric adenocarcinoma. Gastroenterology. 105:1098–1103. 1993.PubMed/NCBI

42. 

Piazuelo MB, Epplein M and Correa P: Gastric cancer: an infectious disease. Infect Dis Clin North Am. 24:853–869. 2010. View Article : Google Scholar : PubMed/NCBI

43. 

Ferreira AC, Isomoto H, Moriyama M, Fujioka T, Machado JC and Yamaoka Y: Helicobacter and gastric malignancies. Helicobacter. 13(Suppl 1): 28–34. 2008. View Article : Google Scholar

44. 

Suganuma M, Kuzuhara T, Yamaguchi K and Fujiki H: Carcinogenic role of tumor necrosis factor-alpha inducing protein of Helicobacter pylori in human stomach. J Biochem Mol Biol. 39:1–8. 2006. View Article : Google Scholar : PubMed/NCBI

45. 

Smith MG, Hold GL, Tahara E and El-Omar EM: Cellular and molecular aspects of gastric cancer. World J Gastroenterol. 12:2979–2990. 2006.

46. 

Chan AO, Lam SK, Wong BC, Wong WM, Yuen MF, Yeung YH, Hui WM, Rashid A and Kwong YL: Promoter methylation of E-cadherin gene in gastric mucosa associated with Helicobacter pylori infection and in gastric cancer. Gut. 52:502–506. 2003. View Article : Google Scholar : PubMed/NCBI

47. 

Iwamoto J, Mizokami Y, Takahashi K, Nakajima K, Ohtsubo T, Miura S, Narasaka T, Takeyama H, Omata T, Shimokobe K, Ito M, Takehara H and Matsuoka T: Expressions of urokinase-type plasminogen activator, its receptor and plasminogen activator inhibitor-1 in gastric cancer cells and effects of Helicobacter pylori. Scand J Gastroenterol. 40:783–793. 2005. View Article : Google Scholar : PubMed/NCBI

48. 

Schmausser B, Endrich S, Brändlein S, Schär J, Beier D, Müller-Hermelink HK and Eck M: The chemokine receptor CCR7 is expressed on epithelium of non-inflamed gastric mucosa, Helicobacter pylori gastritis, gastric carcinoma and its precursor lesions and up-regulated by H. pylori. Clin Exp Immunol. 139:323–327. 2005. View Article : Google Scholar

49. 

Yasumoto K, Koizumi K, Kawashima A, Saitoh Y, Arita Y, Shinohara K, Minami T, Nakayama T, Sakurai H, Takahashi Y, Yoshie O and Saiki I: Role of the CXCL12/CXCR4 axis in peritoneal carcinomatosis of gastric cancer. Cancer Res. 66:2181–2187. 2006. View Article : Google Scholar : PubMed/NCBI

50. 

Zhao C, Lu X, Bu X, Zhang N and Wang W: Involvement of tumor necrosis factor-alpha in the upregulation of CXCR4 expression in gastric cancer induced by Helicobacter pylori. BMC Cancer. 10:4192010. View Article : Google Scholar : PubMed/NCBI

51. 

Amoh Y, Katsuoka K and Hoffman RM: Color-coded fluorescent protein imaging of angiogenesis: the AngioMouse models. Curr Pharm Des. 14:3810–3819. 2008. View Article : Google Scholar : PubMed/NCBI

52. 

Bouvet M, Tsuji K, Yang M, Jiang P, Moossa AR and Hoffman RM: In vivo color-coded imaging of the interaction of colon cancer cells and splenocytes in the formation of liver metastases. Cancer Res. 66:11293–11297. 2006. View Article : Google Scholar : PubMed/NCBI

53. 

Hoffman RM: The multiple uses of fluorescent proteins to visualize cancer in vivo. Nat Rev Cancer. 5:796–806. 2005. View Article : Google Scholar : PubMed/NCBI

54. 

Hoffman RM: Dual-color imaging of tumor angiogenesis. Methods Mol Biol. 515:45–61. 2009. View Article : Google Scholar : PubMed/NCBI

55. 

Chen JJ, Peck K, Hong TM, Yang SC, Sher YP, Shih JY, Wu R, Cheng JL, Roffler SR, Wu CW and Yang PC: Global analysis of gene expression in invasion by a lung cancer model. Cancer Res. 61:5223–5230. 2001.PubMed/NCBI

56. 

Chen HY, Yu SL, Chen CH, Chang GC, Chen CY, Yuan A, Cheng CL, Wang CH, Terng HJ, Kao SF, et al: A five-gene signature and clinical outcome in non-small-cell lung cancer. N Engl J Med. 356:11–20. 2007. View Article : Google Scholar : PubMed/NCBI

57. 

Dhar DK, Wang TC, Tabara H, Tonomoto Y, Maruyama R, Tachibana M, Kubota H and Nagasue N: Expression of trefoil factor family members correlates with patient prognosis and neoangiogenesis. Clin Cancer Res. 11:6472–6478. 2005. View Article : Google Scholar : PubMed/NCBI

58. 

Rodrigues S, Van Aken E, Van Bocxlaer S, Attoub S, Nguyen QD, Bruyneel E, Westley BR, May FE, Thim L, Mareel M, Gespach C and Emami S: Trefoil peptides as proangiogenic factors in vivo and in vitro: implication of cyclooxygenase-2 and EGF receptor signaling. FASEB J. 17:7–16. 2003. View Article : Google Scholar : PubMed/NCBI

59. 

Liu B, Lee KW, Anzo M, Zhang B, Zi X, Tao Y, Shiry L, Pollak M, Lin S and Cohen P: Insulin-like growth factor-binding protein-3 inhibition of prostate cancer growth involves suppression of angiogenesis. Oncogene. 26:1811–1819. 2007. View Article : Google Scholar : PubMed/NCBI

60. 

Akhtar S, Meeran SM, Katiyar N and Katiyar SK: Grape seed proanthocyanidins inhibit the growth of human non-small cell lung cancer xenografts by targeting insulin-like growth factor binding protein-3, tumor cell proliferation, and angiogenic factors. Clin Cancer Res. 15:821–831. 2009. View Article : Google Scholar : PubMed/NCBI

61. 

Hansel DE, Rahman A, House M, Ashfaq R, Berg K, Yeo CJ and Maitra A: Met proto-oncogene and insulin-like growth factor binding protein 3 overexpression correlates with metastatic ability in well-differentiated pancreatic endocrine neoplasms. Clin Cancer Res. 10:6152–6158. 2004. View Article : Google Scholar

62. 

Saaristo A, Karpanen T and Alitalo K: Mechanisms of angiogenesis and their use in the inhibition of tumor growth and metastasis. Oncogene. 19:6122–6129. 2000. View Article : Google Scholar : PubMed/NCBI

63. 

Iwai K, Hirata K, Ishida T, Takeuchi S, Hirase T, Rikitake Y, Kojima Y, Inoue N, Kawashima S and Yokoyama M: An anti-proliferative gene BTG1 regulates angiogenesis in vitro. Biochem Biophys Res Commun. 316:628–635. 2004. View Article : Google Scholar : PubMed/NCBI

64. 

Fisk HA, Mattison CP and Winey M: A field guide to the Mps1 family of protein kinases. Cell Cycle. 3:439–442. 2004.PubMed/NCBI

65. 

Dorer RK, Zhong S, Tallarico JA, Wong WH, Mitchison TJ and Murray AW: A small-molecule inhibitor of Mps1 blocks the spindle-checkpoint response to a lack of tension on mitotic chromosomes. Curr Biol. 15:1070–1076. 2005. View Article : Google Scholar : PubMed/NCBI

66. 

Poss KD, Nechiporuk A, Stringer KF, Lee C and Keating MT: Germ cell aneuploidy in zebrafish with mutations in the mitotic checkpoint gene mps1. Genes Dev. 18:1527–1532. 2004. View Article : Google Scholar : PubMed/NCBI

67. 

Takayama T, Miyanishi K, Hayashi T, Sato Y and Niitsu Y: Colorectal cancer: genetics of development and metastasis. J Gastroenterol. 41:185–192. 2006. View Article : Google Scholar : PubMed/NCBI

68. 

Naderi A, Liu J and Bennett IC: BEX2 regulates mitochondrial apoptosis and G1 cell cycle in breast cancer. Int J Cancer. 126:1596–1610. 2010.PubMed/NCBI

69. 

Chen PS, Wang MY, Wu SN, Su JL, Hong CC, Chuang SE, Chen MW, Hua KT, Wu YL, Cha ST, et al: CTGF enhances the motility of breast cancer cells via an integrin-alphavbeta3-ERK1/2-dependent S100A4-upregulated pathway. J Cell Sci. 120:2053–2065. 2007. View Article : Google Scholar : PubMed/NCBI

70. 

Bracke ME, Van Roy FM and Mareel MM: The E-cadherin/catenin complex in invasion and metastasis. Curr Top Microbiol Immunol. 213:123–161. 1996.PubMed/NCBI

71. 

Mauro L, Bartucci M, Morelli C, Ando S and Surmacz E: IGF-I receptor-induced cell-cell adhesion of MCF-7 breast cancer cells requires the expression of junction protein ZO-1. J Biol Chem. 276:39892–39897. 2001. View Article : Google Scholar : PubMed/NCBI

72. 

Qin J, Feng M, Wang C, Ye Y, Wang PS and Liu C: Oxytocin receptor expressed on the smooth muscle mediates the excitatory effect of oxytocin on gastric motility in rats. Neurogastroenterol Motil. 21:430–438. 2009. View Article : Google Scholar : PubMed/NCBI

73. 

Morita T, Shibata K, Kikkawa F, Kajiyama H, Ino K and Mizutani S: Oxytocin inhibits the progression of human ovarian carcinoma cells in vitro and in vivo. Int J Cancer. 109:525–532. 2004. View Article : Google Scholar : PubMed/NCBI

74. 

Meyer zum Büschenfelde D, Hoschützky H, Tauber R and Huber O: Molecular mechanisms involved in TFF3 peptide-mediated modulation of the E-cadherin/catenin cell adhesion complex. Peptides. 25:873–883. 2004.PubMed/NCBI

75. 

Ortega P, Moran A, Fernandez-Marcelo T, De Juan C, Frias C, Lopez-Asenjo JA, Sanchez-Pernaute A, Torres A, Diaz-Rubio E, Iniesta P and Benito M: MMP-7 and SGCE as distinctive molecular factors in sporadic colorectal cancers from the mutator phenotype pathway. Int J Oncol. 36:1209–1215. 2010.PubMed/NCBI

76. 

Patarroyo M, Tryggvason K and Virtanen I: Laminin isoforms in tumor invasion, angiogenesis and metastasis. Semi Cancer Biol. 12:197–207. 2002. View Article : Google Scholar : PubMed/NCBI

77. 

Wang J, Levenson AS and Satcher RL Jr: Identification of a unique set of genes altered during cell-cell contact in an in vitro model of prostate cancer bone metastasis. Int J Mol Med. 17:849–856. 2006.PubMed/NCBI

78. 

Xi Y, Nakajima G, Hamil T, Fodstad O, Riker A and Ju J: Association of insulin-like growth factor binding protein-3 expression with melanoma progression. Mol Cancer Ther. 5:3078–3084. 2006. View Article : Google Scholar : PubMed/NCBI

79. 

Torng PL, Lee YC, Huang CY, Ye JH, Lin YS, Chu YW, Huang SC, Cohen P, Wu CW and Lin CT: Insulin-like growth factor binding protein-3 (IGFBP-3) acts as an invasion-metastasis suppressor in ovarian endometrioid carcinoma. Oncogene. 27:2137–2147. 2007. View Article : Google Scholar : PubMed/NCBI

80. 

Gao Y, Kitagawa K, Hiramatsu Y, Kikuchi H, Isobe T, Shimada M, Uchida C, Hattori T, Oda T, Nakayama K, Nakayama KI, Tanaka T, Konno H and Kitagawa M: Up-regulation of GPR48 induced by down-regulation of p27Kip1 enhances carcinoma cell invasiveness and metastasis. Cancer Res. 66:11623–11631. 2006. View Article : Google Scholar : PubMed/NCBI

81. 

D’Andrea AD and Grompe M: The Fanconi anaemia/BRCA pathway. Nat Rev Cancer. 3:23–34. 2003.

82. 

Schmidt J and Bosserhoff AK: Processing of MIA protein during melanoma cell migration. Int J Cancer. 125:1587–1594. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September 2012
Volume 41 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tuan T, Chung C, Tsou H, Chen F, Lin H, Lai Y, Lee W, Chao Y, Hwang L, Chen C, Chen C, et al: Putative tumor metastasis-associated genes in human gastric cancer. Int J Oncol 41: 1068-1084, 2012
APA
Tuan, T., Chung, C., Tsou, H., Chen, F., Lin, H., Lai, Y. ... Chen, C. (2012). Putative tumor metastasis-associated genes in human gastric cancer. International Journal of Oncology, 41, 1068-1084. https://doi.org/10.3892/ijo.2012.1502
MLA
Tuan, T., Chung, C., Tsou, H., Chen, F., Lin, H., Lai, Y., Lee, W., Chao, Y., Hwang, L., Chen, C."Putative tumor metastasis-associated genes in human gastric cancer". International Journal of Oncology 41.3 (2012): 1068-1084.
Chicago
Tuan, T., Chung, C., Tsou, H., Chen, F., Lin, H., Lai, Y., Lee, W., Chao, Y., Hwang, L., Chen, C."Putative tumor metastasis-associated genes in human gastric cancer". International Journal of Oncology 41, no. 3 (2012): 1068-1084. https://doi.org/10.3892/ijo.2012.1502