Rapid Communication Open Access
Copyright ©2007 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Sep 14, 2007; 13(34): 4620-4625
Published online Sep 14, 2007. doi: 10.3748/wjg.v13.i34.4620
Changes in gene-expression profiles of colon carcinoma cells induced by wild type K-ras2
Hong Li, Hou-Fa Cao, Inpatient Department of Special Need Treatment, General Hospital of Chinese PLA, Beijing 100083, China
Yuan Li, Mei-Ling Zhu, Jun Wan, Department of South Building Gastroenterology, General Hospital of Chinese PLA, Beijing 100853, China
Author contributions: All authors contributed equally to the work.
Supported by National Natural Science Foundation of China, No. 30200326
Correspondence to: Jun Wan, Department of South Building Gastroenterology, General Hospital of Chinese PLA, Beijing 100083, China. wanjun@301hospital.com.cn
Telephone: +86-10-66937584 Fax: +86-10-66937584
Received: March 15, 2007
Revised: June 20, 2007
Accepted: June 23, 2007
Published online: September 14, 2007

Abstract

AIM: To further elucidate the possible molecular biological activity of wild type K-ras2 gene by detecting changes in wild type K-ras2 gene-induced gene-expression profiles of colon carcinoma cells using cDNA microarray techniques.

METHODS: Total RNA was isolated from peripheral blood of health volunteers. Reverse transcription of RNA and polymerase chain reaction were used to synthesize wild type K-ras2 cDNA. K-ras2 cDNA fragment was cloned into a T easy vector and sequenced. A eukaryotic expression vector pCI-neo-K-ras2 was constructed and transfected to Caco2 cell line using the liposome method. Finally, mRNA was isolated, reverse-transcribed to cDNA from pCI-neo-K-ras2 or pCI-neo blank vector-transfected Caco cells, and analyzed by cDNA microarray assay.

RESULTS: Restriction enzyme analysis and DNA sequencing verified that the constructed expression vector was accurate. High-quality RNA was extracted and reverse transcribed to cDNA for microarray assay. Among the 135 genes, the expression was up-regulated in 24 and down-regulated in 121. All these differentially expressed genes were related to cell proliferation, differentiation, apoptosis and signal transduction.

CONCLUSION: Differentially expressed genes can be successfully screened from wild type K-ras2-transfected colon carcinoma cells using microarray techniques. The results of our study suggest that wild type K-ras2 is related to the negative regulation of cell proliferation, metabolism and transcriptional control, and provide new clues to the further elucidation of its possible biological activity.

Key Words: Wild type K-ras2, Colon cancer, Microarray



INTRODUCTION

Carcinogenesis and progression of human colon carcinoma result from abnormal expression of many tumor-associated genes. Activation oncogenes and deactivation antioncogenes are known as one of their important mechanisms[1,2]. Ras gene which is closely related to carcinogenesis and progression of colon carcinoma consists of three members: Hras1, Nras and K-ras2[3,4]. It is traditionally believed that activated Ras gene plays a dominant role as an oncogene in the pathogenesis of colon carcinoma. About 30% of tumors display mutations of Ras gene members, the most frequent mutation is found in K-ras2[5,6], and the relatively high frequency of K-ras2 mutation is observed in colon, pancreas and lung carcinomas[7-9]. Recent studies indicate that frequent loss of wild type Ras gene occurs in human and mouse lung adenocarcinomas, thus questioning the dominant role of Ras gene in the pathogenesis of tumors[10]. Loss of heterozygosity on chromosome 12p12-13 in K-ras2 gene has also been found in non-small-cell lung cancer[11,12]. We have reported that frequent loss of heterozygosity occurs in this domain during carcinogenesis and progression of colon carcinoma[13]. By observing the changes in growth curve and cell cycle of colon carcinoma cells transducted with wild type K-ras2 gene, we found that wild type K-ras2 gene could step down the growth and cell cycle of colon carcinoma cells manifested as significantly increased stage G0-G1 cells and decreased stage G2-M cells, suggesting that resting cells with proliferation activity are inhibited to advance into proliferating cell cycle[14]. In the present study, in order to study the biological activity of wild type K-ras2 gene, we constructed a eukaryotic expression vector of wild type K-ras2 gene phenotype, screened differentially expressed genes of colon carcinoma cells transfected by wild K-ras2 gene with cDNA microarray, and detected the effect of wild K-ras2 on the gene-expression profiles of colon carcinoma and its function in vivo. The results provide new clues to the exploration of the pathogenesis of colon carcinoma and the functions of K-ras2 gene.

MATERIALS AND METHODS
Cells and cDNA array

Human colon adenocarcinoma cell line Caco-2 was obtained from the ATCC. DMEM, FBS, Trizol RNA isolation kit, pCI-neo mammalian expression vector and Lipofectamine2000 were purchased form Invitrogen (Carlsbad, CA). A commercial human expression cDNA array was obtained from Shanghai Biochip Company (Shanghai, China). The array includes 8568 known genes, which can be categorized into cell division, cell signaling, cell structure, gene and protein expression, metabolism and pseudogene, etc.

Transfection of Caco-2 cells

Total RNA was isolated from peripheral blood of health volunteers using Trizol RNA isolation kit. Reverse transcription of RNA and polymerase chain reaction were used to synthesize the full-length sequence of wild K-ras2 cDNA. Primers containing Mul1 and Sal1 restrictions (Y1: 5'-ACCCACGCGTATGACTGAATATAAAC-3'; Y2: 5'-AACGTCGACTTACATAATTACACACT-3') were synthesized by Shanghai Ouke Biotech Company (Shanghai, China). The PCR products were inserted into pGEM-T Easy vector (Promega) to generate pGEM-T-Ras, and positive clones were identified by blue/white color screening followed by sequencing. pGEM-T-Ras and pCI-neo eukaryotic expression vector were digested in Mul1 and Sal1 restriction enzymes and ligated using T4 DNA ligase (Promega) to produce pCI-neo-K-ras2. The recombinant pCI-neo-K-ras2 and empty pCI-neo-K-ras2 were transfected into Caco-2 cells (ATCC) using Lipofectamine2000 according to the manufacturer’s instructions, and the positive clones were selected from G418 (Amresco).

cDNA microarray analysis

Total RNA was extracted from pCI-neo-K-ras2 (transfection group) and empty pCI-neo-K-ras2 (control group) using Trizol RNA isolation kit. The purity of RNA was confirmed by agarose gel electrophoresis and absorbance (A) ratio (A260/A280). To make cDNA probes, approximately 5 μg of total RNA was labeled with Cy3-dUTP (control) or Cy5-dUTP (transfection group) by reverse transcription. The probes were precipitated using ethanol and dissolved in 5 × SSC + 2 g/L SDS at 20°C. The microarray and probes were denatured in 95°C water bath for 5 min. Hybridization was performed at 60°C for 15-17 h. Microarray was washed with 2 × SSC + 2 g/L SDS and 1 × SSC + 2 g/L SDS for 10 min respectively and dried at room temperature. Scanning was performed with ScanArray3000 (General Scanning, Inc.). The acquired image was analyzed using ImaGene 3.0 software (BioDiscovery, Inc.). The intensities of Cy3-dUTP and Cy5-dUTP were normalized by a coefficient according to the ratio of housekeeping genes. The positively expressed genes were as follows: Cy5-dUTP: Cy3-dUTP signal ratio > 2.0, red fluorescent displaying up-regulated expression; Cy5-dUTP: Cy3-dUTP signal ratio < 0.5, green fluorescent displaying down-regulated expression.

Statistical analysis

Data on gene expression were analyzed by Student’s t test using SPSS 10.0 software and P < 0.05 was considered statistically significant.

RESULTS
Validation of wild type K-ras2 and RNA

Restriction enzyme analysis and nucleotide sequencing of eukaryotic expression vector pCI-neo-K-ras2 showed that its sequence containing an integrity open reading frame was accurate (Figure 1). A 260/A280 of total RNA ranging from 1.9 to 2.1 and 28S/18S of about 2 indicated that RNA was not degraded and could be used for preparation of hybridization probe (Figure 2).

Figure 1
Figure 1 Restriction enzyme analysis of PCI-neo with K-ras2 genes showing a 576 bp K-ras2 gene and a 5600 bp carrier.
Figure 2
Figure 2 Electropherogram of total RNA from control (A) and transfection (B) groups.
Results verified by microarray hybridization system

To monitor the whole process of microarray hybridization, we set up 6 negative controls and 10 positive controls. Scanning of hybridization array and report of array detection showed that hybridization array and sample RNA were intact with good background value and well-distributed noise. The hybridization reaction system was normal and the results were reliable. To compare gene profiles between transfection and control groups, a scatter profile was plotted for the probe signal values, showing that most genes were distributed around the regression line, and their expression in two tissue samples was similar, but a few genes had a different expression. When the difference in gene expression increased, the number of differently expressed genes decreased (Figure 3). The data were confirmed by the low hybridization signal of these genes. Cy5 fluorescein (red) and Cy3 fluorescein (green) were used to mark the probes of experimental and control groups, and the difference in color was expressed as the difference in gene expression between the two groups. Yellow indicated no expression difference. According to the experimental protocol, the expression of 24 genes with their cy5/cy3 ≥ 2 (Table 1) was up-regulated, accounting for 17.76% of all the differentially expressed genes, and the expression of 121 genes with their cy5/cy3 ≤ 0.5 was down-regulated, accounting for 89.63% of all the differentially expressed genes. The top 30 down-regulated genes are listed in Table 2.

Table 1 Biological function of genes with down-regulated expression.
GeneChromosomal localizationBiological functionCy5/Cy3
NDPXp11.4Cross-cell signal transmission, signal transduction, NS development4.509
SSX4Xp11.23Transcription regulation, immunoreaction4.044
CASP111q23Positive regulation of I-kappaB kinase/NF-kappaB cascadereaction, signal transduction, apoptosis3.754
HPRT1Xq26.1Cytolysis, lymphocyte proliferation, purine nucleotide synthesis3.715
TM4SF2Xp11.4ECM3.554
DXS1283EXp22.3Unkown3.418
DPYS8q22NS development, signal transduct-ion, nucleotide metabolism3.386
TYRP19p23Cell metabolism, melanin synthesis3.358
CADPS3p21.1Calcium-regulated Exocytosis3.238
MCF2Xq27Cytokeleton3.187
COL4A6Xq22ECM and ECM synthesis3.201
F13A16p25.3-p24.3Transcriptional control3.108
HTR2CXq24IP3-induced signal pathway3.061
IL13RA1Xq24ECM3.050
OGTXq13Signal transduction3.030
TOSO1q32.1Defence reaction, anti-apoptosis2.994
MAB21L113q13Positive regulation of cell proliferation, visual development2.990
CYBBXp21.1Cross-cell signal transmission, chemotaxis, inflammation, signal transduction2.972
STK9/CDKL5Xp22Microduct skeleton and its synthesis, histogenesis2.946
PTPRG3p21-p14PTK signal pathway2.878
ELAVL29p21Transcriptional control2.853
GLAXq22Osteoclast differentce regulation,bone resorption, cell adhere2.851
APXLXp22.3Channel protein of sodium ion2.839
SERPINA7Xq22.2ECM, transport of TH2.833
Table 2 Biological function of genes with up-regulated expression.
GeneChromosomal localizationBiological functionCy5/Cy3
SZF13p21Transcriptional control0.181
MMP1414q11-q12Incision enzyme, proteolysis0.186
WIT-111p13Cell proliferation0.189
LOC2530127q21.3Gas exchange, cell adhere, early nerve difference and axonogenesis0.201
OPCML11q25Cell adhere, nerve identification0.239
PLAU10q24Chemotaxis, proteolysis, transduction0.258
MYC8q24.12-q24.13Positive-regulated cell proliferation, inhibit cell cycle, balance of iron ion, mRNA synthesis control0.274
SNTB18q23-q24Muscle contraction, skelet-matrix adhere0.281
SERPINESL7q21.3-q22Ser incision enzyme inhibitor, cell component, angiogenetic regulation0.290
C20A12q11-q14PCD transport, cell component0.294
SPTBN12p21Cytokeleton0.298
GNAS20q13.2-q13.3Energy metabolism, G-protein signal pathway, signal transduction0.302
PCOLCE7q22Proteolysis, cell component0.303
ENPP28q24.1Cell movement, G-protein signal pathway, phosphorylation metabolism, lytic activity p'tase activity, transcription0.309
CDK67q21-q22Proliferation, cell cycle, protein conjunct0.315
HIST2H2BE1q21-q23Chromosome component and synthesis0.323
ZNF13719q13.4Transcription control, ion binding0.332
IL1B2q14Inflammation, proliferation, chemotaxis0.336
IL1A2q14Apoptosis, proliferation, transduction, chemotaxis0.343
MTSS18p22Actin component and synthesis, cell adhere, cell movement, PTK signal pathway, muscle and NS development, internalization0.346
ACHE7q22Muscle contraction, transduction0.353
EPHB47q22Proliferation, organ-formation, angiogenesis0.368
AZGP17q22.1Cell member component0.376
PDE4DIP1q12Protein synthesis, actin component0.377
PITPN17p13.3PHL transport0.379
MEOX117q21Growth0.386
GNB2L15q35.3Intercellular signal cascade reaction, protein localization, transduction, PKC0.394
MERTK2q14.1Transduction, cross-cell signal transmission, protein phosphorylation0.405
CUTL17q22.1Transcription control0.419
FCGR2A1q23Immune reaction, signal switch, defence reaction, B cell proliferation0.428
PAX82q12-q14Organ-formation, transcription control, metanephros development, mRNA synthesis0.442
Figure 3
Figure 3 Scatter bar of gene-expression profiles of Caco2 cells transfected (A) and non-transfected (B) with wild-type K-ras2 gene.

Biological function classification of differentially expressed wild type K-ras2 genes Biological function classification of differentially expressed wild type K-ras2 genes was performed based on the biological classification of genes in Affymetrix gene ontology database. Eleven subtypes were found to be closely related to carcinogenesis (Table 3).

Table 3 Functional classification of differentially expressed genes.
Type of geneDown-regulatedgenes (n)Rate ofdown-regulatedgenes (%)Up-regulatedgenes (n)Rate ofup-reguletedgenes (%)
Metabolism-associated1814.88312.50
Cell proliferation3024.79312.50
Cell cycle1613.2200
Signal transduction1814.88833.30
Cytokelet86.6114.17
Transcription control1814.88416.67
Cell adhere119.0914.17
Cell apoptosis86.6128.33
Cell differentiation1411.57416.67
Immune-associated1411.5728.33
ECM64.9614.17
DISCUSSION

Carcinogenesis and progression of colon cancer represent its phases from normal mucosa to atypical hyperplasia (including intestinal metaplasia) of adenoma and adenocarcinoma, involving multiple genes and factors[15]. K-ras2 gene plays a dominant role as an oncogene in promoting carcinogenesis because of point mutation[16,17]. In the present study, in vitro experiments demonstrated that carcinogenic agents used in the treatment of loss of heterozygosity in mice with wild type K-ras2 gene facilitated the development of cancer but not in those with normal wild phenotype K-ras2 gene. Moreover, the size of poorly-differentiated adenocarcinoma in mice with loss of heterozygosity was significantly larger than that of adenoma in mice with abnormal wild phenotype K-ras2 gene. It was reported that cell lines activated by wild type K-ras2 gene-transfected ras can inhibit cell growth, clone formation and tumorigenesis in nude mice, indicating that wild type K-ras2 gene may be a potential anti-oncogene[9,18]. Changes in gene-expression of Caco2 cells induced by wild type K-ras2 gene were found in our study, showing the possible biological activity of wild type K-ras2 gene.

In our study, genes related to signal transduction, transcription control and cell differentiation were dominant, accounting for 33.33% of the total up-regulated genes. The top 30 down-regulated genes related to cell proliferation accounted for 24.79% of the total up-regulated genes. The expression of genes related to cell metabolism, cell cycle and transcription control was up-regulated. Wild type ras may inhibit cell proliferation by promoting differentiation. In fact, it has long been known that Ras proteins can induce differentiation of some cell types, such as neurons, under certain conditions[19]. Our findings suggest that K-ras2 can negatively regulate cell proliferation, metabolism and transcription control, and inhibit the growth of colon carcinoma.

The expression of NDP is most significant. As a genetic locus, its mutation may give rise of genetic-correlated Norrie disease caused by two molecular defects in NDP gene. One is 265 C>G missense mutation in the 97th codon by changing arginine into praline, the other is excalation in 3'-non-translated region of the third exon[20]. It was reported that patients with gene excalation present relatively severe symptoms, whereas patients with gene mutation display relatively mild symptoms[21]. The SSX4 gene (a member of the node point protein family) whose expression was significantly up-regulated in our study, can inhibit cell transcription, cause humor- and cell-mediated immune reaction, and may be a very valuable target for vaccine therapy of tumors[22]. Caspase-1 encoding apoptosis-associated thioserinase (a member of the caspase family) can lead to proteolysis and activate pro-IL-1, thus playing an important role in cell apoptosis[23]. Its up-regulated expression in wild-type K-ras2-transducted cells may be related to apoptosis of tumor cells, suggesting that caspase-1 is one of the human p53-dependent cell modulators[24].

The myc gene whose expression was most significantly up-regulated in our study, is closely related to tumors. It is adjusted by many factors, and can promote cell mitosis and make target cells proliferate and immortalize. This gene involving cell apoptosis is related to tumorigenesis and progression of diverse tumors[25]. Amplification of correlated sequence of myc has been observed in diverse human tumor cell lines including cell lines of granulocytic leukemia, retinoblastoma, neuroblastoma, breast and lung cancer, as well as in human colon carcinoma cell line[26,27]. The MMP14 gene (MT1-MMP) is a member of the matrix metalloprotease family. Its function is modified and regulated by O-glycosylation, interaction with CD44, internalization and recycling, depending on its proper expression on the cell surface[28]. It can invade tumors by activating MMP2 protein. It was reported that up-regulated expression of MMP2 and MT1-MMP is related to invasion of glioblastoma[29], while the expression of MT1-MMP is related to local invasion of and metastasis to lymphonodes of oral squamous cell carcinoma[30], supporting its function in colon carcinoma LoVo cells[31]. The WIT-1 gene is localized in the upstream of Wilm’s tumor gene sharing the same promoter. Methylation of the WIT-1 gene is related to chemotherapy-resistant tumors and acute leukemia[32]. The EPHB4 gene whose expression was remarkably down-regulated in our study is a member of the biggest receptor tyrosine kinase (RTK) family. Its encoding protein, a receptor of ephrin-B2, promotes microvascular endothelial cell migration and/or proliferation, thus playing an important role in angiogenesis of tumors[33]. It has been shown that EPHB4 expresses in diverse tumors such as prostate carcinoma and astrocytoma, and involves phenotype transformation post-metastasis[34,35].

In summary, K-ras2 seems to have a dual function. On the one hand, it promotes cancer development as a gain of function oncogene. On the other hand, it inhibits cancer as a loss-of-function tumor suppressor gene. There are some interesting parallels between the Trp53 tumor suppressor gene and the unfolding story of K-ras2. Trp53 was initially described as an oncogene carrying point mutations in tumors. Later, it was found that it is in fact the wild type copy of the gene that functions as a tumor suppressor gene and is capable of reducing cell proliferation. In this case, the Trp53 mutation may, in a sense, also be considered an activating (but not necessarily gain-of-function) mutation in that it produces a dominant-negative effect over the wild type p53 protein. The two major players in human cancer have more in common than they were previously thought[18].

COMMENTS
Background

Studies indicate that mutation of the K-ras2 gene plays an important role in carcinogenesis and progression of tumors. It was reported that K-ras2 gene not only induces tumors but also inhibits tumor growth. We have reported that loss of heterozygosity on chromosome 12p12-13 K-ras2 gene occurs in colon carcinoma and wild type K-ras2 gene can effectively inhibit its growth. This study was to construct a eukaryotic expression vector of the wild type K-ras2 gene phenotype, screen differentially expressed genes of colon carcinoma cells transfected by the wild K-ras2 gene with cDNA microarray, and detect the effect of wild K-ras2 gene on the gene-expression profiles of colon carcinoma and its function in vivo.

Research frontiers

Based on the results of recent studies, it is hypothesized that K-ras gene plays a role both in carcinogenesis and in inhibition of cancer. It is a poto-oncogene in normal physiological conditions. However, when mutations occur, the activated K-ras2 gene changes into an oncogene and wild type K-ras2 gene becomes an anti-oncogene. This is what we want to prove in this study.

Innovations and breakthroughs

Since the inhibitory effect of wild type K-ras2 gene on tumors was reported by Zhong-Qiu Zhang, its role in suppressing long cancer has been extensively studied by foreign scholars. This is the first time to study the inhibitory effect of wild type K-ras2 gene on colon carcinoma in China.

Applications

The possible molecular pathway of K-ras2 gene in suppressing tumor cell proliferation found in this study may contribute to finding the genes closely related with colon cancer.

Peer review

This interesting paper investigated the importance of K-ras cascade at mRNA level. The major finding of this study is that wild-type K-ras results in both complex induction and more common inhibition of several genes, and may have a dual role in carcinogenesis. The new association elucidated herein may provide further insight into the carcinogenesis and may identify potentially important therapeutic targets.

Footnotes

S- Editor Liu Y L- Editor Wang XL E- Editor Lu W

References
1.  Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990;61:759-767.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8087]  [Cited by in F6Publishing: 7708]  [Article Influence: 226.7]  [Reference Citation Analysis (1)]
2.  Frattini M, Balestra D, Suardi S, Oggionni M, Alberici P, Radice P, Costa A, Daidone MG, Leo E, Pilotti S. Different genetic features associated with colon and rectal carcinogenesis. Clin Cancer Res. 2004;10:4015-4021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in F6Publishing: 174]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
3.  Bos JL. ras oncogenes in human cancer: a review. Cancer Res. 1989;49:4682-4689.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Barbacid M. ras genes. Annu Rev Biochem. 1987;56:779-827.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3375]  [Cited by in F6Publishing: 3396]  [Article Influence: 91.8]  [Reference Citation Analysis (0)]
5.  Kranenburg O. The KRAS oncogene: past, present, and future. Biochim Biophys Acta. 2005;1756:81-82.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Bezieau S, Devilder MC, Avet-Loiseau H, Mellerin MP, Puthier D, Pennarun E, Rapp MJ, Harousseau JL, Moisan JP, Bataille R. High incidence of N and K-Ras activating mutations in multiple myeloma and primary plasma cell leukemia at diagnosis. Hum Mutat. 2001;18:212-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 148]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
7.  Halatsch ME, Hirsch-Ernst KI, Weinel RJ, Kahl GF. Differential activation of the c-Ki-ras-2 proto-oncogene in human colorectal carcinoma. Anticancer Res. 1998;18:2323-2325.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Deramaudt T, Rustgi AK. Mutant KRAS in the initiation of pancreatic cancer. Biochim Biophys Acta. 2005;1756:97-101.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Kim DH, Kim JS, Park JH, Lee SK, Ji YI, Kwon YM, Shim YM, Han J, Park J. Relationship of Ras association domain family 1 methylation and K-ras mutation in primary non-small cell lung cancer. Cancer Res. 2003;63:6206-6211.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Zhang Z, Wang Y, Vikis HG, Johnson L, Liu G, Li J, Anderson MW, Sills RC, Hong HL, Devereux TR. Wildtype Kras2 can inhibit lung carcinogenesis in mice. Nat Genet. 2001;29:25-33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 227]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
11.  Li J, Zhang Z, Dai Z, Plass C, Morrison C, Wang Y, Wiest JS, Anderson MW, You M. LOH of chromosome 12p correlates with Kras2 mutation in non-small cell lung cancer. Oncogene. 2003;22:1243-1246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 47]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
12.  De Gregorio L, Manenti G, Incarbone M, Pilotti S, Pastorino U, Pierotti MA, Dragani TA. Prognostic value of loss of heterozygosity and KRAS2 mutations in lung adenocarcinoma. Int J Cancer. 1998;79:269-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
13.  Li H, Wan J, Li Y, Zhu ML, Zhao P. Loss of heterozygosity on chromosome 12p12-13 region in Chinese patients with colon carcinoma. Zhonghua YiXue YiChuanXue ZaZhi. 2005;22:694-697.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Zhu ML, Wan J, Li H, Li Y. Growth inhibitary effect of wild-type K-ras2 gene on colon adenocarcinoma cell. Junyi Jinxiu Xueyuan Xuebao. 2006;27:18-19.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Wang WS, Chen PM, Su Y. Colorectal carcinoma: from tumorigenesis to treatment. Cell Mol Life Sci. 2006;63:663-671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
16.  Geido E, Sciutto A, Rubagotti A, Oliani C, Monaco R, Risio M, Giaretti W. Combined DNA flow cytometry and sorting with k-ras2 mutation spectrum analysis and the prognosis of human sporadic colorectal cancer. Cytometry. 2002;50:216-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
17.  Kozma L, Kiss I, Nagy A, Szakáll S, Ember I. Investigation of c-myc and K-ras amplification in renal clear cell adenocarcinoma. Cancer Lett. 1997;111:127-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
18.  Pfeifer GP. A new verdict for an old convict. Nat Genet. 2001;29:3-4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
19.  Borasio GD, John J, Wittinghofer A, Barde YA, Sendtner M, Heumann R. ras p21 protein promotes survival and fiber outgrowth of cultured embryonic neurons. Neuron. 1989;2:1087-1096.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 160]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
20.  Rivera-Vega MR, Chiñas-Lopez S, Vaca AL, Arenas-Sordo ML, Kofman-Alfaro S, Messina-Baas O, Cuevas-Covarrubias SA. Molecular analysis of the NDP gene in two families with Norrie disease. Acta Ophthalmol Scand. 2005;83:210-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 5]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
21.  Khan AO, Shamsi FA, Al-Saif A, Kambouris M. A novel missense Norrie disease mutation associated with a severe ocular phenotype. J Pediatr Ophthalmol Strabismus. 2004;41:361-363.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Ayyoub M, Merlo A, Hesdorffer CS, Rimoldi D, Speiser D, Cerottini JC, Chen YT, Old LJ, Stevanovic S, Valmori D. CD4+ T cell responses to SSX-4 in melanoma patients. J Immunol. 2005;174:5092-5099.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
23.  Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10:417-426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4047]  [Cited by in F6Publishing: 4286]  [Article Influence: 194.8]  [Reference Citation Analysis (0)]
24.  Gupta S, Radha V, Sudhakar Ch, Swarup G. A nuclear protein tyrosine phosphatase activates p53 and induces caspase-1-dependent apoptosis. FEBS Lett. 2002;532:61-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
25.  Kim YH, Girard L, Giacomini CP, Wang P, Hernandez-Boussard T, Tibshirani R, Minna JD, Pollack JR. Combined microarray analysis of small cell lung cancer reveals altered apoptotic balance and distinct expression signatures of MYC family gene amplification. Oncogene. 2006;25:130-138.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Kowara R, Gołebiowski F, Chrzan P, Skokowski J, Karmolinski A, Pawełczyk T. Abnormal FHIT gene transcript and c-myc and c-erbB2 amplification in breast cancer. Acta Biochim Pol. 2002;49:341-350.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Pelengaris S, Khan M, Evan G. c-MYC: more than just a matter of life and death. Nat Rev Cancer. 2002;2:764-776.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 825]  [Cited by in F6Publishing: 829]  [Article Influence: 37.7]  [Reference Citation Analysis (0)]
28.  Itoh Y, Seiki M. MT1-MMP: an enzyme with multidimensional regulation. Trends Biochem Sci. 2004;29:285-289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 65]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
29.  Guo P, Imanishi Y, Cackowski FC, Jarzynka MJ, Tao HQ, Nishikawa R, Hirose T, Hu B, Cheng SY. Up-regulation of angiopoietin-2, matrix metalloprotease-2, membrane type 1 metalloprotease, and laminin 5 gamma 2 correlates with the invasiveness of human glioma. Am J Pathol. 2005;166:877-890.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 88]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
30.  Myoung H, Kim MJ, Hong SD, Lee JI, Lim CY, Hong SP. Expression of membrane type I-matrix metalloproteinase in oral squamous cell carcinoma. Cancer Lett. 2002;185:201-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
31.  Deryugina EI, Ratnikov BI, Yu Q, Baciu PC, Rozanov DV, Strongin AY. Prointegrin maturation follows rapid trafficking and processing of MT1-MMP in Furin-Negative Colon Carcinoma LoVo Cells. Traffic. 2004;5:627-641.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
32.  Gessler M, Bruns GA. Sequence of the WT1 upstream region including the Wit-1 gene. Genomics. 1993;17:499-501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
33.  Steinle JJ, Meininger CJ, Forough R, Wu G, Wu MH, Granger HJ. Eph B4 receptor signaling mediates endothelial cell migration and proliferation via the phosphatidylinositol 3-kinase pathway. J Biol Chem. 2002;277:43830-43835.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 139]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
34.  Xia G, Kumar SR, Masood R, Zhu S, Reddy R, Krasnoperov V, Quinn DI, Henshall SM, Sutherland RL, Pinski JK. EphB4 expression and biological significance in prostate cancer. Cancer Res. 2005;65:4623-4632.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 114]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
35.  Xiao HL, Li ZP, Yin YS, Zhang QH, Jiang XF, Bian XW. Expressions of EphB4 receptor and its ligand ephrinB2 in brain astrocytoma and their value in prognosis. Disan Junyi Daxue Xuebao. 2005;18:49-52.  [PubMed]  [DOI]  [Cited in This Article: ]