http://www.abbs.info e-mail:[email protected]

ISSN 0582-9879                             ACTA BIOCHIMICA et BIOPHYSICA SINICA 2003, 35(6): 511-517                             CN 31-1300/Q

The Effects of Inhibiting P18INK4C Expression on the Invasion of Gastric Adenocarcinoma Cell Line

WANG Chu, WANG Jian-Hua, LI Feng, LU Jian*

( Department of Biochemistry and Molecular Biology, Research Center for Human Gene Therapy, Shanghai Second Medical University, Shanghai 200025, China )

 

Abstract        Using cDNA microarray with double dots of 4096 human genes, P18INK4C, a member of CKI, was found down-regulated in a gastric adenocarcinoma metastatic cell line (RF-48), compared with the corresponding primary cancer cell line (RF-1), which implied that P18INK4C might be involved in cell invasion and metastatic progression of human gastric adenocarcinoma. Antisense RNA expression plasmid was applied to inhibit P18INK4C expression to study the effect of decreased P18INK4C expression on cell migration, invasion and proliferation ability and cell cycle of RF-1. Results showed that inhibition of P18INK4C expression could obviously enhance cell invasion ability of RF-1, but had little effect on its cell cycle and cell migration and proliferation ability. These results implied that P18INK4C might play a pivotal role in regulating cell invasion, rather than regulating cell cycle and proliferation in the progression of human gastric adenocarcinoma as expected before.

 

Key words     P18INK4C; human gastric adenocarcinoma; invasion and metastasis

 

P18INK4C, a protein of 18 kD, is a member of INK4 family of cyclin-dependent kinase inhibitors (CKIs) which also include P15INK4A, P16INK4B and P19INK4D. These CKI proteins could specifically bind not only monomeric CDK4 or CDK6 to prevent the formation of CDK-cyclin complex, but also the CDK4/6-cyclin complex to form an inactive ternary complex CKI-CDK-cyclin[18]. P18INK4C could also inhibit the activity of CDK-cyclin complex, especially CDK6-cyclinD, by distorting the ATP binding site and misaligning catalytic residues, which could block cell cycle progression[1,4,9]. Furthermore it distorted the cyclin-binding site of CDK as well, with the cyclin remaining bound to CDK at a size-reduced interface[1].

P18INK4C gene was localized on 1p32 which was frequently involved in chromosomal changes in various tumors[10]. P18INK4C was considered as a potential candidate in tumor therapy since its mutations, deletions and aberrant expressions existed in many kinds of human cancers, such as breast cancer, oligodendrogliomas, Wilms tumor and testicular cancer[1115]. Recently, it was found that P18INK4C might be also correlated with cancer invasion and metastasis. The research of Bartkova et al.[14] have suggested that increased abundance of cyclinE and particularly down-regulation or loss of P18INK4C might be one of the features of the progression from pre-invasive carcinomas in situ (CIS) to invasive germ cell tumours of the human testis.

Using cDNA microarray with double dots of 4096 human genes, it was found that the expression of P18INK4C gene was down-regulated in RF-48 which is the metastatic cell line of a gastric adenocarcinoma patient, compared with RF-1 which is the primary cancer cell line of the same person. Besides, we also found the expression of cyclin E2 gene being up-regulated in RF-48 compared with RF-1[16]. By using antisense RNA expression plasmid, we inhibited the P18INK4C expression to study its effect on cell migration, invasion and proliferation ability and cell cycle. Our findings indicated that down-regulated expression of P18INK4C was associated with the enhanced invasion ability of RF-1 and this effect was not the result of its function of cell cycle arrest. These implied that P18INK4C might be involved in cell invasion and metastatic progression of human gastric adenocarcinoma.

 

1    Materials and Methods

1.1   Materials

RF-1, the primary tumor cell line of a gastric adenocarcinoma patient, and RF-48 cell lines, its relative metastatic counterpart were obtained from ATCC. E. coli DH5α and pcDNA3-GFP were from our lab. RPMI 1640 and Opti-MEM were purchased from Gibco BRL, and FBS was from Hyclone; G418 was bought from Roche; MTT was from Sigma; Trizol reagent for RNA extraction and MLV reverse transcriptase were purchased from Gibco BRL; DNA extraction kit was bought from Gentra; EX-Taq was from TaKaRa; HindIII, BamHI and T4 DNA ligase were obtained from New England Biolabs; LipofectAMINETM 2000 was the product of Invitrogen; transwell chambers (pore size: 8 μm) were bought from Costar; Matrigel basement membrane matrix (phenol-red free) was purchased from Becton Dickinson; Northern blotting kit was from Boehringer Mannheim, Roche; Western blotting kit and anti-mouse IgG-HRP were products from Wuhan Boshide Inc; anti-P18 mouse monoclonal IgG antibody was bought from Santa Cruz Biotechnology Inc.; RIPA kit was from Shanghai Shenergy Biocolor Inc.; Primers were synthesized by Shanghai Sangon Inc.; DNA was sequenced by Shanghai Genecore Inc.. All other chemicals were of analytical grade.

1.2   Cell culture

RF-1 (ATCC No: CRL-1864), and RF-48 (ATCC No: CRL-1863) were suspending cell lines and routinely cultured in a 37 , 5% CO2 incubator with RPMI 1640 containing 10% FBS.

1.3   Construction of antisense P18INK4C RNA expres-sion plasmid

Total RNA, isolated from RF-1 using the Trizol reagent according to the manufacturer's instructions, was reversely transcribed and the product was used as the template for PCR. The antisense P18INK4C gene (product size, 550 bp) was amplified by the upstream primer (5-AGTGGATCCCGTGAACAAGGGACCCT-AAAG-3) and the downstream primer (5-AGTAAG-CTTCGTTTATTGAAGATTTGTGGCTC-3). BamHI and HindIII sites were underlined respectively. After digested by the two restriction endonucleases, the partial antisense P18INK4C gene and pcDNA3 were ligated by T4 ligase to form an antisense P18 RNA expression plasmid, named as pcDNA3-antisense-p18.

1.4   Cell transfection and screening

RF-1 was transfected with pcDNA3-GFP and pcDNA3-antisense-p18 respectively using Lipofect-AMINETM 2000 according to the manufacturer's instructions. Transfection rate was calculated according to the cells transfected with pcDNA3-GFP by using FACS analysis. A cell suspension with a density of 106 cells/mL in RPMI 1640 growth medium with 10%FBS and without antibiotics was added to a 6-well plate. 4 μg of pcDNA3-antisense-p18 or pcDNA3-GFP in 250 μL of Reduced serum medium and 12 μL LipofectAMINETM 2000 in 250 μL Opti-MEMI medium were incubated for 5 min and 20 min at room temperature respectively, then these tow solutions were mixed, and added gently to each well containing RF-1 cells. Then the cells were incubated for another 48 h before the cell activities to be detected.

Transfected cells were incubated for 2 d and then screened with 500 mg/L G418 (active concentration). After 5 d, the concentration of G418 was reduced to 250 mg/L and then maintained. G418-resistant cell clones, named RF-1-anti-p18 and RF-1-GFP were isolated and expanded respectively. Throughout the process of cell screening, the cells were kept in RPMI 1640 containing 30% FBS.

1.5   Northern blotting

All the process was referred to the manufacturer's instructions and the standard protocol of “Molecular Cloning: A Laboratory Manual” (Cold Spring Harbor Laboratory Press, 1989). β-actin was used as a control.

1.6   Western blotting

Protein was prepared by using RIPA kit and quantified by the method of BCA[17]. After separated by 12% SDS-PAGE gels30 μg of each protein sample was transferred onto nitrocellulose membranes and probed with the mouse monoclonal IgG antibody of P18INK4C and anti-mouse IgG-HRP. Bands were visualized by DAB. The detailed procedure was referred to the standard protocol of “Molecular Cloning: A Laboratory Manual” (Cold Spring Harbor Laboratory Press, 1989) and the manufacturer's instructions.

1.7   RT-PCR

Semi-quantitative RT-PCR was adopted to detect the results of transfection and screening of RF-1-anti-p18 and the change of P18INK4C expression at mRNA level. GAPDH was used as an internal control. Total RNA, isolated from RF-1, RF-1-GFP and RF-1-anti-p18 using the Trizol reagent, was reversely transcribed and the products were used as the templates for PCR. neo (product size, 410 bp) was amplified by the upstream primer (5-GAAGGGACTGGCTGCTATTG-3) and the downstream primer (5-AGCCAACGCT-ATGTCCTGAT-3). Endogenous P18INK4C gene in cells (product size, 452 bp) was amplified by the upstream primer (5-GGTGGAGTTCCTGGTGAAGC-3) and the downstream primer (5-CAACTTGGGTGTTG-AGAT-3). GAPDH (product size, 233 bp) was amplified by the upstream primer (5-TGGGGAA-GGTGAAGGTCGG-3) and the downstream primer (5-CTGGAAGATGGTGATGGGA-3).

1.8   PCR

To verify the insertion of antisense P18INK4C gene into the genome, PCR was performed using genomic DNAs of RF-1, RF-1-GFP and RF-1-anti-p18 as the templates. The genomic DNAs were prepared by using Gentra DNA extraction kit. The upstream primer was 5-ACCCACTGCTTACTGGCTTATCG-3, and the downstream primer was 5-TTACAGACTTTGCTGG-AGTTTCA-3. PCR product size was 314 bp.

1.9   MTT assay of cell proliferation

MTT assay was conducted to determine the cell proliferation. 103 cells in 2 mL of RPMI 1640 growth medium with 10%FBS were plated into a 96-well plate and incubated for 24 h. After that, 20 μL of 5 g/L MTT solution was added to each well and incubated for another 4 h in a 37 °C incubator. 150 μL DMSO was added to dissolve the crystal after the growth medium was removed. Then the absorbance of 200 μL solution from each well was measured at 570 nm by a microplate reader. Each sample was assayed in octuple for 7 d consecutively. Cell growth curves of RF-1, RF-48, RF-1-GFP and RF-1-anti-p18 were obtained based on the corresponding values of A570 respectively.

1.10 Cell cycle analysis

The detailed procedure was followed as previously reported[18]. 105 cells were collected by centrifugation, washed, and suspended in ice-cold PBS. Cells were then fixed with 70% ethanol for 10 min in room temperature, followed by staining with propidium iodide (PI). Cell cycle was assessed by FACS.

1.11 Tumor cell migration and invasion assay

Cell migration assays were performed using 6.5 mm transwell chambers (pore size: 8 μm) as described previously[19,20]. Briefly, conditioned NIH-3T3 medium was added to the bottom well, and the filters were coated by the conditioned NIH-3T3 medium for 30 min at 37 . Cells were resuspended in the appropriate buffer at a concentration of 106 cells/mL and 105 cells were added to the top well of transwell chambers. After 4 h incubation, the cells that had not migrated were removed from the upper face of the filters using cotton swabs, and the cells that had migrated to the lower surface of the filters were fixed in methanol and stained by hematoxylin. Migration was determined by the count of the cells that had migrated to the lower surface of the filter with a microscope at ×400. Six visual fields were counted for each essay. Assays were repeated for three times.

Matrigel invasive assays were performed using 6.5 mm transwell chambers (pore size: 8 μm)[19,20]. Matrigel (Matrigel basement membrane matrix, phenol-red free, Becton Dickinson) was diluted in serum-free medium, added to the top well of transwell chambers (19.6 μg/well), and dried under a sterile hood. The Matrigel was then reconstituted with medium for 2 h at 37 °C before the addition of cells. Cells were resuspended in serum-free medium containing 0.1% BSA. 2×105 cells were added to each well. Conditioned NIH-3T3 medium was added to the bottom well of transwell chambers. After 24 h, the cells that had not migrated were removed from the upper face of the filters using cotton swabs, and the cells that had invaded to the lower surface of the filters were fixed by methanol, and stained and counted as described above, then the number of cells that invaded to the lower side of the filter was measured as a parameter of the invasive ability of the cells. Assays were repeated for three times.

1.12 Statistical analysis

Data are presented as x±s and significance was determined by the Student's t-test.

 

2    Results

2.1   Analysis of P18INK4C expression in RF-1 and RF-48

Using Northern blotting and Western blotting, the expression of P18INK4C gene was found to be down-regulated in RF-48, the metastatic cell line of a gastric adenocarcinoma patient, compared with RF-1, the primary cancer cell line of the same person, which confirmed the results of cDNA microarray[16] (Fig.1). These results implied that the reduced expression of P18INK4C gene might be involved in the progression of human gastric adenocarcinoma metastasis.

Fig.1       The expression of P18INK4C in RF-1 and RF-48

(A) Northern blotting analysis of P18INK4C and β-actin expressed in RF-1 and RF-48. 1, P18INK4C and β-actin expressed in RF-1; 2, P18INK4C and β-actin expressed in RF-48. (B) Western blotting analysis of P18INK4C expressed in RF-1 and RF-48. 1, P18INK4C expressed in RF-48; 2, P18INK4C expressed in RF-1.

 

2.2   Analysis of the effects of cell transfection and screening

Using the total RNA of RF-1, RF-1-GFP and RF-1-anti-p18 as templates, RT-PCR was adopted to amplify the neo gene from these three kind cells respectively. It was found that neo could only be amplified from RF-1-GFP and RF-1-anti-p18 (Fig.2), which indicated the process of cell transfection and screening was successful.

Fig.2       RT-PCR analysis of transfection and screening of RF-1-GFP and RF-1-anti-p18

1, 100 bp DNA ladder marker; 2, neo expressed in RF-1; 3, neo expressed in RF-1-GFP; 4, neo expressed in RF-1-anti-p18. GAPDH was used as an internal control.

To verify the insertion of antisense P18INK4C gene into the genome, PCR was performed using genomic DNAs of RF-1, RF-1-GFP and RF-1-anti-p18 as the templates. The upstream primer was designed complementary to the cytomegalo virus (CMV) promoter sequence in the pcDNA3, whereas the downstream primer was complementary to the antisense P18INK4C sequence. The desired PCR product (314 bp) was obtained from RF-1-anti-p18 (Fig.3), which indicated the integration of antisense P18INK4C gene into the genome of RF-1.

 

 

Fig.3       PCR detection of a 314 bp fragment from the genomic DNAs of RF-1, RF-1-GFP and RF-1-anti-p18

1, 100 bp DNA ladder marker; 2, PCR result of RF-1; 3, PCR result of RF-1-GFP; 4, PCR result of RF-1-anti-p18.

 

2.3   Expression of P18INK4C in RF-1-anti-p18

Through analyzing the expression of P18INK4C in RF-1, RF-1-GFP and RF-1-anti-p18, the effect of antisense P18INK4C could be evaluated. The expression of endogenous P18INK4C, amplified by RT-PCR, was obviously inhibited in RF-1-anti-p18, compared with RF-1 and RF-1-GFP (Fig.4). The result of Western blotting also showed that the protein level in RF-1-anti-p18 was lower than that in the other two (Fig.4). All of these results indicated that the expression of P18INK4C was inhibited by antisense P18INK4C.

Fig.4       The expression of P18INK4C in RF-1, RF-1-GFP and RF-1-anti-p18

(A) RT-PCR analysis of P18INK4C expressed in RF-1, RF-1-GFP and RF-1-anti-p18. 1, 100 bp DNA ladder marker; 2, P18INK4C expressed in RF-1; 3, P18INK4C expressed in RF-1-GFP; 4, P18INK4C expressed in RF-1-anti-p18. (B) Western blotting analysis of P18 expressed in RF-1, RF-1-GFP and RF-1-anti-p18. 1, P18INK4C expressed in RF-1; 2, P18INK4C expressed in RF-1-GFP; 3, P18INK4C expressed in RF-1-anti-p18.

 

2.4   Cell proliferation and cell cycle analysis

The proliferation ability of RF-1, RF-48, RF-1-GFP and RF-1-anti-p18 was evaluated by MTT assay. According to the A570 values of 7 d, cell growth curves were made (Fig.5). There was no detectable difference in the proliferation ability among them, which, at the same time, suggested that the proliferation of RF-1-anti-p18 cells was not affected by the change in the expression of P18INK4C.

Fig.5       Cell growth curves of RF-1, RF-1-GFP, RF-1-anti-p18 and RF-48 in 7 d

The cell growth curve was measured by MTT assay. Values were expressed as x±s n= 8.

 

Using FACS, cell cycle of RF-1RF-48RF-1-GFP and RF-1-anti-p18 were analyzed. No significant difference of cell cycle distribution was found among these different cells (Fig.6).

Fig.6       Cell cycle analysis of RF-1, RF-1-GFP, RF-1-anti-p18 and RF-48 by FACS

1, RF-1; 2, RF-1-GFP; 3, RF-1-anti-p18; 4, RF-48.

 

2.5   Cell migration and invasion abilities in vitro

According to the results of cell migration assay in vitro, it was found that RF-1, RF-1-GFP and RF-1-anti-p18 could all migrate to the lower surface of the transwell filter under the same condition. As to the migrated cells stained by hematoxylin under a microscope at ×400, there were (91±3) cells of RF-1, (95±3) cells of RF-1-GFP and (94±3) cells of RF-1-anti-p18 (Fig.7), and no significant difference was found among the groups, which implied that cell migration abilities of these different cells were similar.

 

Fig.7       Comparison of cell migration ability of RF-1, RF-1-GFP and RF-1-anti-p18.

Cell number in each visual field was counted under a phase contrast microscope at ×400. Values are expressed as x±s, n=3.

 

Under the same condition we analyzed the cell invasion ability of the RF-1, RF-1-GFP and RF-1-anti-p18 in vitro. There were (15±2) cells of RF-1, (16±1) cells of RF-1-GFP and (34±3) cells of RF-1- anti-p18 each visual field under a phase contrast microscope at ×400(Fig.8). We could find significant difference among the cell numbers. The result suggested that RF-1-anti-p18 could easily migrate though Matrigel, compared with the other two.

 

Fig.8       Comparison of cell invasion abilities of RF-1, RF-1-GFP and RF-1-anti-p18

Cell number in each visual field was counted under a phase contrast microscope at ×400. Values were expressed as x±s, *P<0.01 vs. RF-1-anti-p18, n=3.

 

3    Discussion

Invasion and metastasis of tumor cells were the main causes of cancer patients’ death[21,22]. The mechanisms of acquiring the capabilities of invasion and metastasis were different from the primary mechanisms of inducing malignant transformation. In various experimental systems, the expression of a considerable amount of genes affected metastatic ability, and several model systems had been established to study mechanisms that induced tumor cells to metastasize[2329]. However, despite recent progress in the knowledge about metastatic pathways, the mechanisms hadn't been fully understood yet.

In order to identify the possible metastasis-associated genes of human gastric adenocarcinoma, cDNA microarray with double dots of 4096 human genes was performed and the expression of P18INK4C was found to be down-regulated in RF-48, the metastatic cell line of a gastric adenocarcinoma patient, compared with RF-1, the primary cancer cell line[16]. The result was also substantiated by Northern blotting and Western blotting. Bartkova et al.[14] demonstrated that P18INK4C was considerably reduced or absent at the protein level in invasive germ cell tumors (GCTs), which was different from its abundant expression in the CIS cells, and suggested that down-modulation or loss of P18INK4C might contribute to progression from pre-invasive lesions to overt germinal tumors. All of these gave us a hint that the down-regulation of P18INK4C expression might be associated with the progression of invasion and metastasis of gastric adenocarcinoma.

By using antisense RNA technology, we successfully inhibited the expression of P18INK4C gene in RF-1 with pcDNA-antisense-p18 and studied its effects on cell migration and invasion ability as well as cell cycle and proliferation. The results of cell migration and invasion assay indicated that although the migration abilities of RF-1, RF-1-GFP and RF-1-anti-p18 were similar, RF-1-anti-p18 apparently migrate though Matrigel and filter more easily than the other two cells. But these results could not assure us that the invasion ability of RF-1-anti-p18 was actually enhanced because the mechanism of cell invasion assay was that the invasion ability was determined by the total number of the cells having migrated through Matrigel and 8 μm pores to the lower surface of the filter. As a negative regulator of cell cycle, reduced expression of P18INK4C gene may increase the number of the cells having migrated through the transwell filter by increasing the total cell number of RF-1-anti-p18 in a certain period without affecting cell invasion ability. Recent studies indicated that although P18INK4C, as a member of INK4 family, could block cell cycle progression at G1/S phrase, it did not play a pivotal role in controlling cell cycle and proliferation due to the compensatory roles by the Cip/Kip family of CKI, and loss of P18INK4C did not appear to confer any proliferative advantage to some kind of cell lines[15, 30]. Our results of MTT assay and cell cycle analysis also showed that there were no significant difference in cell cycle and proliferation among RF-1, RF-1-GFP and RF-1-anti-p18. Therefore, through all the researches above, it can be concluded that the inhibition of P18INK4C expression could enhance the invasion ability of RF-1 without affecting its migration ability, which implied that P18INK4C might play an important role in regulating cell invasion ability in the progression of human gastric adenocarcinoma metastasis. Besides, the primary function of P18INK4C might not be to regulate cell cycle and proliferation during the development of gastric adenocarcinoma from CIS to invasive cancer.

 

References

1     Jeffrey PD, Tong L, Pavletich NP. Structural basis of inhibition of CDK-cyclin complexes by INK4 inhibitors. Genes Dev, 2000, 14(24): 31153125

2     Noh SJ, Li Y, Xiong Y, Guan KL. Identification of functional elements of p18INK4C essential for binding and inhibition of cyclin-dependent kinase (CDK) 4 and CDK6. Cancer Res, 1999, 59(3): 558564

3    Serrano M, Hannon GJ, Beach D. A new regulatory motif in cell cycle control causing specific inhibition of cyclin D/cdk4. Nature, 1993, 366(6456): 704707

4     Guan KL, Jenkins CW, Li Y, Nichols MA, Wu X, O'Keefe CL, Matera AG et al. Growth suppression by p18, a p16INK4/MTS1-and p14INK4B/MTS2-related CDK6 inhibitor, correlates with wild-type pRb function. Genes Dev, 1994, 8(24): 29392952

5     Hannon GJ, Beach D. p15INK4B is a potential effector of TGF-beta-induced cell cycle arrest. Nature, 1994, 371(6494): 257261

6     Chan FK, Zhang J, Chen L, Shapiro DN, Winoto A. Identification of human and mouse p19, a novel CDK4/CDK6 inhibitor with homology to p16ink4. Mol Cell Biol, 1995, 15: 26822688

7     Hirai H, Roussel MF, Kato J, Ashmun RA, Sherr CJ. Novel INK4 proteins, p19 and p18, are specific inhibitors of cyclin D-dependent kinases CDK4 and CDK6. Mol Cell Biol, 1995 15: 26722681

8     Guan KL, Jenkins CW, Li Y, O'Keefe CL, Noh S, Wu X, Zariwala M et al. Isolation and characterization of p19INK4d, a p16-related inhibitor specific to CDK6 and CDK4. Mol Biol Cell, 1996, 7: 5770

9     Aprelikova O, Xiong Y, Liu ET. Both p16 and p21 families of cyclin-dependent kinase (CDK) inhibitors block the phosphorylation of cyclin-dependent kinases by the CDK-activating kinase. J Biol Chem, 1995, 270: 1819518197

10    Platz A, Hansson J, Ringborg U. Screening of germline mutations in the CDK4, CDKN2C and TP53 genes in familial melanoma: A clinic-based population study. Int J Cancer, 1998, 78(1): 1315

11    He J, Hoang-Xuan K, Marie Y, Leuraud P, Mokhtari K, Kujas M, Delattre JY et al. P18 tumor suppressor gene and progression of oligodendrogliomas to anaplasia. Neurology, 2000, 55(6): 867869

12    Arcellana-Panlilio MY, Egeler RM, Ujack E, Pinto A, Demetrick DJ, Robbins SM, Coppes MJ. Decreased expression of the INK4 family of cyclin-dependent kinase inhibitors in Wilms tumor. Genes Chromosomes Cancer, 2000, 29(1): 6369

13    Sotillo R, Garcia JF, Ortega S, Martin J, Dubus P, Barbacid M, Malumbres M. Invasive melanoma in Cdk4-targeted mice. Proc Natl Acad Sci USA, 2001, 98(23): 1331213317

14    Bartkova J, Thullberg M, Rajpert-De Meyts E, Skakkebaek NE, Bartek J. Cell cycle regulators in testicular cancer: Loss of p18INK4C marks progression from carcinoma in situ to invasive germ cell tumours. Int J Cancer, 2000, 85(3): 370375

15    Latres E, Malumbres M, Sotillo R, Martin J, Ortega S, Martin-Caballero J, Flores JM et al. Limited overlapping roles of P15(INK4b) and P18(INK4c) cell cycle inhibitors in proliferation and tumorigenesis. EMBO J, 2000, 19(13): 34963506

16    Wang JH, Chen SS. Screening and identification of gastric adenocarcinoma metastasis-related genes by using cDNA microarray coupled to FDD-PCR. Acta Biochim Biophys Sin 2002, 34(4): 475481

17    Smith PK, Krohn RI, Hermanson GT, Hermanson GT, Mallia AK, Gartner FH, Provenzano MD, Fujimoto EK et al. Measurement of protein using bicinchoninic acid. Anal Biochem, 1985, 150(1): 7685

18    Lei QY, Wang LY, Dai ZY, Zha XL. The relationship between PTEN expression and anoikis in human lung carcinoma cell lines. Acta Biochim Biophys Sin, 2002, 34(4): 463468

19    Mensing H, Pontz BF, Muller PK, Gauss-Muller V. A study on fibroblast chemotaxis using fibronectin and conditioned medium as chemoattractants. Eur J Cell Biol 1983, 29(2): 268273

20    Albini A, Iwamoto Y, Kleinman HK, Martin GR, Aaronson SA, Kozlowski JM, McEwan RN. A rapid in vitro assay for quantitating the invasive potential of tumor cells. Cancer Res, 1987, 47: 32393245

21    Pepper MS. Lymphangiogenesis and tumor metastasis: Myth or reality? Clin Cancer Res, 2001, 7: 462468

22    John A, Tuszynski G. The role of matrix metalloproteinases in tumor angiogenesis and tumor metastasis. Pathol Oncol Res, 2001, 7: 1423

23    Sobel ME. Metastasis suppressor genes. J Natl Cancer Inst, 1990, 82(4): 267276

24    Keller SM, Adak S, Wagner H, Herskovic A, Komaki R, Brooks BJ, Perry MC et al. A randomized trial of postoperative adjuvant therapy in patients with completely resected stage II or IIIA non-small-cell lung cancer. Eastern Cooperative Oncology Group. N Engl J Med, 2000, 343: 12171222

25    Sleeman Jp. The lymph node as a bridgehead in the metastatic dissemination of tumors. Recent Result Cancer Res, 2000, 157: 5581

26    Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T et al. Invovlement of chemokine receptors in breast cancer metastsis. Nature, 2001, 410: 5056

27    Hibi K, Liu Q, Beaudry GA, Madden SL, Westra WH, Wehage SL, Yang SC et al. Serial analysis of gene expression in non-small cell lung cancer. Cancer Res, 1998, 58: 56905694

28    Weston WM, LeClair EE, Trzyna W, McHugh KM, Nugent P, Lafferty CM, Ma L et al. Differential display identification of plunc, a novel gene expressed in embryonic palate, nasal epithelium, and adult lung. J Biol Chem, 1999, 274: 1369813703

29    Krus-Berthier L, Jan M, Guilbaud N, Naze M, Pierre A, Atassi G. Histology and sensitivity to anticancer drugs of two human non-small cell lung carcinomas implanted in the pleural cavity of nude mice. Clin Cancer Res, 2000, 6: 297304

30    Franklin DS, Godfrey VL, Lee H, Kovalev GI, Schoonhoven R, Chen-Kiang S, Su L et al. CDK inhibitors p18(INK4c) and p27(Kip1) mediate two separate pathways to collaboratively suppress pituitary tumorigenesis. Genes Dev, 1998, 12(18): 28992911

 


Received: March 3, 2003Acccepted: April 4, 2003

*Corresponding author: Tel, 86-21-63846590-776441; e-mail, [email protected]