Colorectal Cancer Open Access
Copyright ©2007 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Jan 21, 2007; 13(3): 341-348
Published online Jan 21, 2007. doi: 10.3748/wjg.v13.i3.341
Identification of the differential expressive tumor associated genes in rectal cancers by cDNA microarray
Xue-Qin Gao, Jin-Xiang Han, Hua-Ning Zhang, Hai-Yan Huang, Key Laboratory for Biotech-Drug, Ministry of Health, Key Laboratory of Modern Drugs and Technology of Shandong Province, Shandong Medical and Biotechnology Center, Shandong Academy of Medical Sciences, Jinan 250062, Shandong Province, China
Zhong-Fa Xu, Department of Surgery, Shandong Tumor Hospital, Shandong Academy of Medical Sciences, Jinan 250117, Shandong Province, China
Wei-Dong Zhang, Department of Experimental Pathology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan 250062, Shandong Province, China
Author contributions: All authors contributed equally to the work.
Supported by Major Project of Shandong Provincial Science and Technology, No.011100105, and Shandong Natural Sciences Funding Committee, No. Y2003C03
Correspondence to: Jin-Xiang Han, Shandong Medical and Biotechnology Center, Shandong Academy of Medical Sciences; Key Laboratory for Biotech Drugs, Ministry of Health; Key Laboratory of Modern Drugs and Technology of Shandong Province; 89 Jingshi Road, Jinan 250062, Shandong Province, China. samshjx@sina.com
Telephone: +86-531-82919888 Fax: +86-531-82951586
Received: July 27, 2006
Revised: August 25, 2006
Accepted: September 26, 2006
Published online: January 21, 2007

Abstract

AIM: To identify tumor associated genes of rectal cancer and to probe the application possibility of gene expression profiles for the classification of tumors.

METHODS: Rectal cancer tissues and their paired normal mucosa were obtained from patients undergoing surgical resection of rectal cancer. Total RNA was extracted using Trizol reagents. First strand cDNA synthesis was indirectly labeled with aminoallyl-dUTP and coupled with Cy3 or Cy5 dye NHS mono-functional ester. After normalization to total spots, the genes which background subtracted intensity did not exceed 2 SD above the mean blank were excluded. The data were then sorted to obtain genes differentially expressed by ≥ 2 fold up or down in at least 5 of the 21 patients.

RESULTS: In the 21 rectal cancer patients, 23 genes were up-regulated in at least 5 samples and 15 genes were down-regulated in at least 5 patients. Hierachical cluster analysis classified the patients into two groups according to the clinicopathological stage, with one group being all above stage II and one group all below stage II.

CONCLUSION: The up-regulated genes and down-regulated genes may be molecular markers of rectal cancer. The expression profiles can be used for classification of rectal cancer.

Key Words: Rectal cancer, Tumor associated genes, cDNA microarray, Differential expression genes



INTRODUCTION

Colorectal cancer (CRC) is one of the most common causes of cancer related deaths, the majority of which are secondary to liver metastasis. Whereas significant improvements have been made in patient survival, the mortality is still quite high in patients with metastatic cancer[1]. Clinical trials of new therapeutic agents are constantly ongoing and drug effectiveness is improving. However, a complete cure for patients with advanced colorectal cancer awaits new targets and strategies. To achieve this goal, a comprehensive understanding of the mechanisms of colorectal carcinogenesis is essential. In the last two decades these mechanisms have been intensively studied. At the molecular level, activation of oncogenes[2] and inactivation of tumor suppressor genes are processes known to be involved in colorectal cancer[3,4]. CRC typically develops over decades and involves multiple genetic events. This has led to the development of a multi-step model of colorectal tumorigenesis[5]. It is generally accepted that one of the initiating steps in colorectal carcinogenesis is mutation in APC tumor suppressor gene[6].

APC is known to bind to a cell signaling/transcription factor β catenin. Activation of the Wnt pathway normally signals the association of β-catenin with members of the T-cek factor/lymphocyte-enhancer factor family and translocation to nucleous[7,8]. This complex can activate the transcription of a variety of target genes including c-Myc[9] and cyclin D1[10,11]. Loss of APC function leads to an abnormal accumulation of β-catenin and dys-regulates Wnt signaling[12]. Traditional methods of identifying novel targets that are involved in colon cancer progression are based on the studies of individual genes. The developments of oligonucleotide or cDNA microarrays have made it possible to permit the expression levels of tens of thousands of genes to be monitored simultaneously and rapidly. These new methods have been used to identify the genes involved in colorectal cancer development. By using cDNA microarray with different number of elements, many genes have been identified. Kitahara[13] identified 235 genes of differential expression in 9216 human genes by using laser-capture micro-dissection. These differentially expressed genes include those associated with signal transduction, metabolic enzymes, production of reactive oxygen species, cell cycle, transcription, mitosis and apoptosis[13]. In another study, Williams identified 2632 genes (574 up-regulated and 2058 down-regulated) of totally 9592 genes microarray were consistently expressed in one-third patients[14]. In an effort to use gene expression profiles to find the molecular marker of rectal cancer, we constructed cDNA microarray with 373 known genes and 73 expressed sequence tags (ESTs) that are related to the pathogenesis of tumor.

MATERIALS AND METHODS

Fabrication of tumor associated genes cDNA microarray.

Totally 447 cDNA clones were obtained from the Research Genetics (Invitrogen, life Technologies, USA). E. coli with cDNA clones were cultured with LB supplemented with ampicillin (50 mg/L) or chloramphenicol (170 mg/L). Clone plasmids were extracted with Edge BioSystems plasmid extraction kit. Clone inserts were PCR amplified from the plasmids with M13 vector specific universal primer (M13F: 5’-GGT GTA AAA CGA CGG CCA GTG-3’; M13R: 5’-CAC ACA GGA AAC AGC TAT G-3’) in 96-well PCR microtiters. The PCR products were purified according to protocols published previously, and resuspended in Arrayit Spot solution [DNA precipitations. http://cmgm.stanford.edu/pbrown/protocols/2_DNA.html[15]. The re-suspended PCR products were printed in duplicate on silianated glass slides (Cel associated) with Cartisan Spot machines and UV-cross-linked at 3500 mJ using Stratagene cross-linker. Printed slides were post-processed using an online method[16].

Tissue samples

Rectal cancer tissues and their corresponding normal rectal tissues were obtained with informed consent from 21 patients who underwent surgical resection of their tumor in Shandong Tumor Hospital. The tissues were stored at -80°C. Tissues were homogenized with Trizol (Life Technologies) in homogenizer of IKA. Total RNA was extracted according to the manufacturer’s protocols. The A(260/280) of total RNA was between 1.8-2.0.

Probe preparation and hybridization and data analysis

Twenty microgram of total RNA from matched tumor/normal sample was reverse transcribed into cDNA using Superscript II (Invitrogen, Carlsbad CA) with random hexamers (Invitrogen, Carlsbad CA). Probe preparation was done based on online protocols and as described by Yang[17] and slightly modified by our laboratory[18]. First strand cDNA synthesis was primed with 6 μg random hexamers (Life Technologies) by heating at 70°C for 10 min, snap-cooling on ice for 30 s and incubated at room temperature for an additional 5-10 min. Reverse transcription was performed in the presence of 500 μmol/L each of dATP, dCTP and dGTP, 200 μmol/L aminoalyl-dUTP (Sigma), 300 μmol/L dTTP, 1 × first strand buffer, 10 μmol/L dithiolhreitol, and 400 U superscript II (Life Technologies, Carlsbad, CA) in 30 μL reaction at 42°C overnight. Reactions were quenched with 0.5 mol/L EDTA and RNA template was hydrolyzed by addition of 10 μL of 1 mol/L NaOH followed by heating at 70°C for 10 min. Reactions were neutralized with 10 μL 1 mol/L HCl and cDNA was purified with Amicon Microcon YM100 according to the manufacturer’s protocol. cDNA was lyophilized in speed vaccum Centrifuger (Eppendorf 5301) and resuspended in 4.5 μL of 0.1 mol/L sodium carbonate( pH 9.0) buffer. NHS ester (4.5 μL) Cy3 or Cy5 dye (Amersham Pharmacia, GE Healthcare) in DMSO [dye from one tube (1 mg) was dissolved in 73 μL of DMSO] was added and reactions were incubated at room temperature in the dark for 1 h. Coupling reactions were quenched by addition of 41 μL of 0.1 mol/L sodium acetate, pH 5.2, and unincorporated dye was removed using QiaQuick PCR purification kits according to the manufacturer’s instruction.

Hybridization and image processing

Each slide was printed with duplicate microarrays. Slides were prehybridized in 1% BSA, 5 × SSC, 0.1% SDS for 45 min, washed by dipping in double distilled deionized H2O and 2-propanol twice and air dried and used in 1 h. Fluorescent cDNA probes were lyophylized to dryness and resuspended in 10 μL hybridization buffer (formamide 5 μL, 20 × SSC 2.5 μL, RGDDH 201 μL, 2% SDS 0.5 μL and 1 μL human cot-1 DNA). Combined Cy dye labeled probes were denatured at 100°C water bath for 2 min and cooled at room temperature for 5 min. Room temperature probes were applied to the pre-hybridized microarray and covered with hybridized coverslip (Sigma) and placed in the hybridization chamber (Corning). Hybridizations were carried out at 42°C water bath for 20-22 h followed by washing in 2 × SSC and 0.1% SDS for 3 min, 1 × SSC for 2 min and 0.2 × SSC for 1 min and 0.05 × SSC for 10 s, and dried by spinning in plate centrifuge at 800 rotation/min for 4 min. Arrays were scanned using a Scan-array 4000 dual color confocal laser scanner (Packard Bioscience). Data were saved as paired TIFF images. Paired images were analyzed with Quantarray 3.0 provided by the Scanner. After normalized to the total, genes of which the background subtracted intensity did not exceed 2 SD above the mean blank were excluded. The data were then sorted to obtain genes differentially expressed by ≥ 2 fold or ≤ 0.5 in at least 5 of the patients.

Semi-quantitative RT-PCR

Some of the differentially expressed genes were selected for validation of the microarray results with semi-quantitative RT-PCR. The sequences of the primers were designed with online primer design software (Primer 3 old Version http://www.genome.wi.mit.edu/cgi-bin/primer/primer3.cgi#PRIMER_self_any[19]. The sequences of primers used for RT-PCR are as follows: MTA1 forward, 5’-AGCTACGAGCAGCACAACGGGGT-3’, reverse, 5’-CACGCTTGGTTTCCGAGGAT-3’; β-actin forward, 5’-GTGGGGCGCCCCAGGCACCA-3’, reverse, 5’-CTCCTTAATGTCACGCACGATTTC-3′[12]. RT-PCRs were performed with TAKARA version 2.1 RT-PCR kit using random primers in 25 µL volume. The PCR reaction was performed at 94°C for 2 min, followed by 30 cycles at 94°C for 30 s, 56°C for 30 s and 72°C for 30 s and a final extension at 72°C for 5 min. β-actin was used as internal control. The products were electrophoresed in 1% agarose (Spanish).

Immunohistochemistry analysis of the BCL-2 protein expression in all patients

All the samples of rectal cancer and paired normal tissues were subject to immunohistochemistry examination for the expression of BCL-2 to validate the results of microarray. The primary antibody of BCL-2 was a product of Santa Cruz (1:100 dilution). The expressions of BCL-2 in tumor and normal tissues were read by two pathologists blindly. The image was captured with NIKON H600L microscope with digital camera.

Cluster analysis and principal component analysis

The normalized ratios of all pairs of samples of rectal cancer were put into an Excel-sheet. And every ratio was log2 transformed and saved as a tab-delimited Txt. file that was ready to be analyzed with Version 5.0 kindly provided online free of charge by Professor Leif E Peterson (http://mbcr.bcm.tmc.edu/genepi)[20].

RESULTS

Totally 21 pairs of rectal samples were analyzed. Their pathological and clinical characteristics are shown in Table 1. The tissue samples were all confirmed to be rectal cancer and normal paired tissues (Figure 1A and B). The differential expression genes were identified by a tumor associated genes microarray fabricated in our laboratory. The representative scan images are shown in Figure 1C, D and E which represented the image of normal tissue, rectal cancer and the overlay of the image respectively. In the overlay images of microarray, red color spots represented genes that were up-regulated in rectal cancer tissues, green color spots represented down-regulated genes and yellow color spots represented the expression of genes in cancer tissue, which were between 0.5-2.0. If the ratio of the duplicate spots was the same, we defined it the significantly differentially expressed gene. The expression ratios of tumor associated genes in tumor and paired normal rectal tissues were analyzed with QuantaArray 3.0 and normalized to the total. The 2-fold up- or down-regulated genes were defined as differential expression genes.

Table 1 Clinical data of the rectal cancer patients.
Patient No.Age (yr)SexTNMClinical stage
131MaleT4N2M0III
251FemaleT3N1M0III
347MaleT3N1M0III
467FemaleT3N0M0II
649MaleT2N2M0III
738MaleT3N0M0II
859FemaleT3N0M0II
953MaleT3N0M0II
1062MaleT2N0M0I
1162MaleT3N1M0III
1241MaleT3N1M0III
1333MaleT4N0M0II
1476MaleT3N1M0III
1575FemaleT2N0M0I
1632FemaleT3N1MoIII
1765FemaleT3N1MoII
1848FemaleT3N0MoII
1932MaleT3N2M0III
2068MaleT2N0M0I
2176MaleT3N0M0II
2276MaleT3N0M0II
Figure 1
Figure 1 Histopathological examination of the tumor tissues of rectal cancer. A: Normal rectal mucosa tissue; B: Adenocarcinoma of rectal tissues; C, D and E: images of normal tissue, tumor tissues and overlay image of the tumor to normal of patient No.19 respectively. The density of the spot reflects the fluorescent intensity of the genes. In the overlay images, the yellow spots represent the expression of the genes in tumor tissues and normal tissues were not significant. The red spots represent the up-regulated genes in rectal cancer tissues. The green spots represent the down-regulated genes in rectal cancer tissues.

The results showed that 23 genes were up-regulated in at least 5 samples (Table 2) and 15 genes were down regulated at least in 5 samples (Table 3). The up-regulated genes included growth factors, growth factor receptor, nuclear transcription factor, oncogenes, adhesion molecules, and some metabolic enzymes.

Table 2 Commonly up-regulated genes in rectal cancer patients showing ≥ 2 fold over expression.
AccessionNo.Gene descriptionPatients(n)
NM_000184Hs.23763, hemoglobin, gamma G (186624)8
NM_000633B-cell CLL/lymphoma 2 (BCL2), nuclear gene encoding mitochondrial protein, transcript variant alpha (3950016)6
Not foundHs.77579, ESTs (2811005)6
Not foundHs.87497, ESTs (1216395)5
BB366468Hs.126701, ESTs, Weakly similar to homolog of the Aspergillus nidulans sudD gene product [H.sapiens] (1082187)5
NM_000600Interleukin 6 (interferon, beta 2) (IL6) (3876176)5
NM_004469c-fos induced growth factor (vascular endothelial growth factor D) (FIGF) (160946)5
BM440387KIAA0712 gene product (539641)5
BB217505Hs.106513, Homo sapiens mRNA; cDNA DKFZp586I1518 (from clone DKFZp586I1518) (585388)5
NM_002210Integrin, alpha V (vitronectin receptor, alpha polypeptide, antigen CD51) (ITGAV) (3878920)5
L12168Adenylyl cyclase-associated protein (2217802)5
NM_007047Hs.19301, butyrophilin, subfamily 3, member A2 (1070574)5
NM_018844b-cell receptor associated protein (2746778)5
U41371Spliceosome associated protein 145 (1964680)5
NM_001022Hs.5621, ribosomal protein S19 (728984)5
NM_002506Nerve growth factor, beta polypeptide (NGFB) (72869)5
NM_000121Erythropoietin receptor (EPOR) (114048)5
NM_003371Vav 2 oncogene (VAV2) (3861692)5
NM_000267Neurofibromin 1 (neurofibromatosis, von Recklinghausen disease, Watson disease) (NF1) (3868563)5
AF071400Plasminogen activator inhibitor, type II (arginine-serpin) (PAI2) (323255)5
NM_006059Laminin, gamma 3 (LAMC3) (2497685)5
NM_002865Hs.234726, RAB2, member RAS oncogene family (728947)5
NM_013230Hs.180414, CD24 antigen (small cell lung carcinoma cluster 4 antigen) (115306)5
Table 3 Commonly down-regulated genes in rectal cancers showing < 0.5 under expression.
AccessionNo.Gene descriptionPatients(n)
Not foundHs.170311, EST (126182)5
NM_004689Metastasis associated 1 (MTA1) (4054392)5
BC001766S100 calcium-binding protein, beta (neural) (S100B) (759948)5
NM_005246Fer (fps/fes related) tyrosine kinase (phosphoprotein NCP94) (FER) (3886018)5
NM_001920Decorin (DCN) (274397)5
NM_001951E2F transcription factor 5, p130-binding (E2F5) (701492)5
NM_002211Homo sapiens integrin, beta 1 (fibronectin receptor, beta polypeptide, antigen CD29 includes MDF2, MSK12) (ITGB1) (531839)5
Not foundHs.23954, ESTs (132543)5
Not foundHs.5621, ESTs (2947053)5
NM_007191Wnt inhibitory factor-1 (WIF-1) (40908)5
NM_021785Retinoic acid induced 2 (RAI2) (501868)5
Not foundTachykinin, precursor 1 (substance K, substance P, neurokinin 1, neurokinin 2, neuromedin L, neurokinin alpha, neuropeptide K, neuropeptide gamma) (TAC1), transcript variant beta (784179)5
NM_004345Cathelicidin antimicrobial peptide (CAMP) (3057931)5
AF385430Rho GTPase activating protein 1 (ARHGAP1) (3896574)5
NM_005252v-fos FBJ murine osteosarcoma viral oncogene homolog (FOS) (755279)5

To validate the results of microarray examination, the expression of MTA1 mRNA was measured with semi-quantitative RT-PCR using β-actin as internal control. The results showed that the MTA1 expression was down-regulated (Figure 2). The up-regulation of BCL-2 was also validated by immunohistochemistry and significant expression of BCL-2 was shown in tumor tissues and less expression in normal rectal mucosa (Figure 3A-D).

Figure 2
Figure 2 Semi-quantitative RT-PCR of MTA1 mRNA. A: Electrophoresis image of MTA1 expression, lanes 1-8 represent the mRNA expression of MTA1 in 4 pairs of rectal cancer patients, respectively. Lanes 1, 3, 5, 7 were normal tissues and 2, 3, 6, 8 were tumor tissues. B: The semi-quantitative analysis results with spot intensity software. The ratios of MTA1 in tumor tissues were lower than that of paired normal tissues.
Figure 3
Figure 3 Validation of the expression of BCL-2 with immunohistochemistry method. A: The normal rectal mucosa. There was no expression of BCL-2 in all the cells; B: The expression of BCL-2 in well differentiated rectal cancer. The tumor cells were stained yellow-brown by DAB; C: The expression of BCL-2 in another intermediately differentiated rectal cancer. The expression of BCL-2 was also significant in tumor cells; D: The expression of BCL-2 in poorly differentiated rectal tissues.

For the cluster analysis, the Claufavor version 6.0 supplied kindly by Professor Leif Peterson was used to classify the patients or genes by gene expression profiles. As a result, the patients were classified into two groups, with patients No 22, 13, 12, 1, 11, 21, 19, 2, 6, 16, 15, 3 into one group which were all above grade II except patient No 15 and the other patients into another group. The top 100 genes that were related with rectal cancer are shown in Figure 4A and B. Principal component analysis showed that all the genes were belonged to 7 factors, in which 17 genes were positively correlated with factor 1 (r > 0.45). They are as follows: apoptosis associated protein, xeroderma pigmentosum, complementation group A (XPA), early endosome antigen 1162 kDa (EEA), lactate dehydrogenase A, spectrin, alpha non-erythrocytic 1 (alpha-fodrin), small nuclear ribonucleoprotein associated protein n (human), phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1) , RAS p21 protein activator (GTPase activating protein) 1 (RASA1), cathepsin D (lysosomal aspartyl protease) (CTSD), v-ski avian sarcoma viral oncogene homolog (SKI), Hs.271616, ESTs, small nuclear ribonucleoprotein associated proteins b and b’ (human); contains mer22.t1 mer22 repetitive element, CD83 antigen (activated B lymphocytes, immunoglobulin superfamily) (CD83), bone marrow stromal cell antigen 1 (BST1), Wnt inhibitory factor-1 (WIF-1), histone deacetylase 1 (HDAC1), microvascular endothelial differentiation gene 1 (MDG1).

Figure 4
Figure 4 Hierarchial cluster analysis and principal component analysis results. A: Hierachial cluster analysis of patients and genes. The patients could be classified into two groups: one group of clinicopathological stage above grade II and one group below grade II. B: Principal component analysis of the genes related to rectal cancer.

Nine genes were negatively correlated with factor 3 (r < -0.45) (Figure 4B); they are as follows: apoptosis associated protein, phospholipase C, delta 1 (PLCD1), small nuclear ribonucleoprotein associated protein n (human), RAS p21 protein activator (GTPase activating protein) 1 (RASA1), cathepsin D (lysosomal aspartyl protease) (CTSD), met proto-oncogene (hepatocyte growth factor receptor) (MET), 65 KDa yes-associated protein; contains Alu repetitive element; contains element MER31 repetitive element, KIAA0712 gene product, ESTs (Weakly similar to homolog of the Aspergillus nidulans sudD gene product). These results showed that microarray expression profiles may be used for the classification of rectal cancer.

DISCUSSION

Differential gene expression can be detected in several ways, including differential screening of cDNA libraries[21], subtraction hybridization[22] differential display of RNA[23], serial analysis of gene expression (SAGE)[24] and cDNA microarray[25]. Among these cDNA microarray is the most commonly used technique now for surveying many samples with thousands of genes[26]. By using cDNA microarray methods and laser capture microdissection, 235 differential expression genes were identified from colorectal cancers[14]. A colonchip has been constructed for the identification of genes that are related to colorectal cancer tumorigenesis, 59 genes showing two-fold increase of altered expression with 23 genes commonly up-regulated and 36 genes commonly down-regulated in colorectal cancer[27]. All these studies were about colorectal carcinoma, while few were concerned with rectal cancer. Moreover, rectal cancer has the tendency to occur in young age. To identify the molecular markers of rectal cancer we constructed a tumor associated genes cDNA microarray for the evaluation of the predisposition to metastasis with gene expression profiles. All the tumor associated genes were selected from literatures that are related with the growth, invasion and metastasis process of tumor and some ESTs. We used this cDNA microarray to detect the differential expression genes that play important roles in the pathogenesis of rectal cancer. We found that 23 genes were up-regulated in rectal cancers compared with their paired normal tissues. The most commonly expressed gene in rectal cancers was BCL-2. It was validated by immunohistochemistry. We also found that oncogene vav-2 was up-regulated in 5 of the 21 patients. Vav-2 acts as a guanosine nucleotide exchange factor (GEF) for RhoG and RhoA-like GTPases in a phosphotyrosine-dependent manner. Vav-2 oncogenic activation correlates with the acquisition of phosphorylation-independent exchange activity. In vivo, wild-type Vav-2 is activated oncogenically by tyrosine kinases, an effect enhanced further by co-expression of RhoA. Likewise, the Vav-2 oncoprotein synergizes with RhoA and RhoB proteins in cellular transformation. Transient transfection assays in NIH-3T3 cells show that phosphorylated wild-type Vav-2 and the Vav-2 oncoprotein induce cytoskeletal changes resembling those observed by the activation of the RhoG pathway. In contrast, the constitutive expression of the Vav-2 oncoprotein in rodent fibroblasts leads to major alterations in cell morphology and to highly enlarged cells in which karyokinesis and cytokinesis frequently are uncoupled[28].

There were 15 down-regulated genes including PIGF gene products, MTA1, WIF-1 ras-guanine releasing protein and all pre-mRNA cleavage factor Im. There was a controversial report that MTA1 are down-regulated in rectal cancer and it has been validated by RT-PCR. Toh et al[29] reported a relative over-expression of MTA1 mRNA (tumor/normal ratio ≥ 2) was observed in 14 of 36 (38.9%) colorectal carcinomas. Tumors over-expressing MTA1 mRNA exhibited a significantly deeper wall invasion and a higher rate of metastasis to lymph nodes, and tended to be at an advanced Dukes’ stage with frequent lymphatic involvement[29].

In addition, Nicolson has reported the over-expression of MTA1 in several cell lines (the breast, ovarian, lung, gastric and colorectal cancer but not melanoma or sarcoma) and cancerous tissues (breast, esophageal, colorectal, gastric and pancreatic cancer)[30]. Giannini et al[31] have reported that MTA1 increased in tumor tissues than in normal tissues by real-time PCR. This difference may be attributed to the different function MTA1 exerts in different tumor stage. Further studies are needed at protein levels.

To investigate the possibility of classification of tumor with gene expression profiles, hierarchical cluster analysis was done and the results showed that the patients could be divided into two groups: clinicopathological grade higher than grade II and the other below grade II.

Many studies have done to uncover mechanisms underlying progression of colorectal carcinogenesis and to identify genes associated with the liver metastasis. Li et al[32] analyzed expression profiles of 14 primary colorectal cancers with liver metastases, and compared them with profiles of 11 non-metastatic carcinomas and those of 9 adenomas of the colon. A hierarchical cluster analysis using data from a cDNA microarray containing 23 040 genes indicated that the cancers with metastasis had different expression profiles from those without metastasis although a number of genes were commonly up-regulated in primary cancers of both categories. They found that 54 genes were frequently up-regulated and 375 genes were frequently down-regulated in primary tumors with metastases to liver, but not in tumors without metastasis. Subsequent quantitative PCR experiments confirmed that PRDX4, CKS2, MAGED2, and an EST (GenBank accession number BF696304) were expressed at significantly higher levels in tumors with metastasis[32]. Koehler has studied the expression profiles of 25 colorectal carcinomas (CRCs, pT1-4), corresponding normal colonic mucosa, and 14 liver metastases using cDNA arrays containing 1176 cancer-related genes (Clontech). They found no specific expression signature in matching metastases, but a set of 23 classifier genes with statistically significant expression patterns in high- and low-stage tumors was identified[33].

Komori et al[34] constructed the gene expression profiles of 50 colorectal cancers (CRCs) and 12 normal colorectal epithelia using a cDNA microarray specially constructed for CRC. Hierarchical clustering analysis and principal component analysis could clearly distinguish the gene profiles of cancer tissues from those of normal tissues. They identified 22 up-regulated genes and 32 down-regulated genes in CRC. Many of these genes have been previously identified in association with human carcinogenesis, being 68% and 78%, respectively[34].

Moreover, D’Arrigo et al[35] compared the transcriptional profiles of 10 radically resected primary CRCs from patients who did not develop distant metastases within a 5-year follow-up period with those of 10 primary/metastatic tumor pairs from patients with synchronous liver metastases. Arrays of 7864 human cDNAs were utilized using laser-microdissected bioptic tissues. Of 37 gene expression differences found between the 2 groups of primary tumors, 29 were also different between non-metastasizing tumors from metastases. The gene encoding mannosyl (alpha-1, 3-)-glycoprotein beta-1, 4-N-acetyl-glucosaminyl-transferase (GnT-IV) became significantly up-regulated in primary/metastatic tumor pairs (P < 0.001)[35]. All these results were obtained from colorectal cancer. However, colon cancer and rectal cancer may have different molecular markers. Our methods have similar ability in classifying tumor with gene expression profiles.

In conclusion, we have screened 23 up-regulated genes and 15 down-regulated genes from 21 rectal cancers. Hierarchical cluster analysis divided the patients into one group with clinicopathological stage above II, and one group below stage II. These genes may be the molecular markers of rectal cancer. In addition, we have also found gene expression profiles in different stage of rectal cancers and to further screen the molecular markers in each stage of the cancer larger samples are needed.

ACKNOWLEDGMENTS

We thank Professor Leaf Peterson at Baylor College of Medicine for his free CLUSFAVOR 5.0 software and his instruction for the study. We also thank Dr Feng-Wei Wang in Medical College of Wisconsin for the language correction.

Footnotes

S- Editor Wang GP L- Editor Zhu LH E- Editor Bi L

References
1.  Gutman M, Fidler IJ. Biology of human colon cancer metastasis. World J Surg. 1995;19:226-234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 92]  [Article Influence: 3.2]  [Reference Citation Analysis (1)]
2.  Forrester K, Almoguera C, Han K, Grizzle WE, Perucho M. Detection of high incidence of K-ras oncogenes during human colon tumorigenesis. Nature. 1987;327:298-303.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 745]  [Cited by in F6Publishing: 726]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
3.  Baker SJ, Fearon ER, Nigro JM, Hamilton SR, Preisinger AC, Jessup JM, vanTuinen P, Ledbetter DH, Barker DF, Nakamura Y. Chromosome 17 deletions and p53 gene mutations in colorectal carcinomas. Science. 1989;244:217-221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1354]  [Cited by in F6Publishing: 1390]  [Article Influence: 39.7]  [Reference Citation Analysis (0)]
4.  Fearon ER, Cho KR, Nigro JM, Kern SE, Simons JW, Ruppert JM, Hamilton SR, Preisinger AC, Thomas G, Kinzler KW. Identification of a chromosome 18q gene that is altered in colorectal cancers. Science. 1990;247:49-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1202]  [Cited by in F6Publishing: 1309]  [Article Influence: 38.5]  [Reference Citation Analysis (0)]
5.  Kinzler KW, Vogelstein B. Lessons from hereditary colorectal cancer. Cell. 1996;87:159-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3455]  [Cited by in F6Publishing: 3301]  [Article Influence: 117.9]  [Reference Citation Analysis (0)]
6.  Groden J, Thliveris A, Samowitz W, Carlson M, Gelbert L, Albertsen H, Joslyn G, Stevens J, Spirio L, Robertson M. Identification and characterization of the familial adenomatous polyposis coli gene. Cell. 1991;66:589-600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1920]  [Cited by in F6Publishing: 1824]  [Article Influence: 55.3]  [Reference Citation Analysis (0)]
7.  Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, Vogelstein B, Kinzler KW. APC mutations occur early during colorectal tumorigenesis. Nature. 1992;359:235-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1308]  [Cited by in F6Publishing: 1270]  [Article Influence: 39.7]  [Reference Citation Analysis (0)]
8.  Behrens J, von Kries JP, Kühl M, Bruhn L, Wedlich D, Grosschedl R, Birchmeier W. Functional interaction of beta-catenin with the transcription factor LEF-1. Nature. 1996;382:638-642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2241]  [Cited by in F6Publishing: 2234]  [Article Influence: 79.8]  [Reference Citation Analysis (0)]
9.  He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B, Kinzler KW. Identification of c-MYC as a target of the APC pathway. Science. 1998;281:1509-1512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3533]  [Cited by in F6Publishing: 3516]  [Article Influence: 135.2]  [Reference Citation Analysis (0)]
10.  Shtutman M, Zhurinsky J, Simcha I, Albanese C, D'Amico M, Pestell R, Ben-Ze'ev A. The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc Natl Acad Sci USA. 1999;96:5522-5527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1701]  [Cited by in F6Publishing: 1730]  [Article Influence: 69.2]  [Reference Citation Analysis (0)]
11.  Tetsu O, McCormick F. Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature. 1999;398:422-426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2796]  [Cited by in F6Publishing: 2806]  [Article Influence: 112.2]  [Reference Citation Analysis (0)]
12.  Sparks AB, Morin PJ, Vogelstein B, Kinzler KW. Mutational analysis of the APC/beta-catenin/Tcf pathway in colorectal cancer. Cancer Res. 1998;58:1130-1134.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Kitahara O, Furukawa Y, Tanaka T, Kihara C, Ono K, Yanagawa R, Nita ME, Takagi T, Nakamura Y, Tsunoda T. Alterations of gene expression during colorectal carcinogenesis revealed by cDNA microarrays after laser-capture microdissection of tumor tissues and normal epithelia. Cancer Res. 2001;61:3544-3549.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Williams NS, Gaynor RB, Scoggin S, Verma U, Gokaslan T, Simmang C, Fleming J, Tavana D, Frenkel E, Becerra C. Identification and validation of genes involved in the pathogenesis of colorectal cancer using cDNA microarrays and RNA interference. Clin Cancer Res. 2003;9:931-946.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  DNA precipitations  Available from: http: //cmgm.stanford.edu/pbrown/protocols/2_DNA.html.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Post processing of microarrays  Available from: http://www.umbi.umd.edu/~cbr/postprocessing601.html.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Yang IV, Chen E, Hasseman JP, Liang W, Frank BC, Wang S, Sharov V, Saeed AI, White J, Li J. Within the fold: assessing differential expression measures and reproducibility in microarray assays. Genome Biol. 2002;3:research0062.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Gao XQ, Han JX, Huang HY, Yan S, Song CZ, Huang HN. Effects of aspirin on metastasis-associated gene expression detected by cDNA microarray. Acta Pharmacol Sin. 2004;25:1327-1333.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Online primer design software  Available from: http://www.genome.wi.mit.edu/cgi-bin/primer/primer3.cgi#PRIMER_self_any.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Peterson LE. CLUSFAVOR 5.0: hierarchical cluster and principal-component analysis of microarray-based transcriptional profiles. Genome Biol. 2002;3:SOFTWARE0002.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Höög C. Isolation of a large number of novel mammalian genes by a differential cDNA library screening strategy. Nucleic Acids Res. 1991;19:6123-6127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 83]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
22.  Sagerström CG, Sun BI, Sive HL. Subtractive cloning: past, present, and future. Annu Rev Biochem. 1997;66:751-783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 101]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
23.  Liang P, Pardee AB. Differential display of eukaryotic messenger RNA by means of the polymerase chain reaction. Science. 1992;257:967-971.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3487]  [Cited by in F6Publishing: 3160]  [Article Influence: 98.8]  [Reference Citation Analysis (0)]
24.  Velculescu VE, Zhang L, Vogelstein B, Kinzler KW. Serial analysis of gene expression. Science. 1995;270:484-487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2999]  [Cited by in F6Publishing: 2557]  [Article Influence: 88.2]  [Reference Citation Analysis (0)]
25.  Schena M, Shalon D, Davis RW, Brown PO. Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science. 1995;270:467-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6477]  [Cited by in F6Publishing: 5032]  [Article Influence: 173.5]  [Reference Citation Analysis (0)]
26.  DeRisi J, Penland L, Brown PO, Bittner ML, Meltzer PS, Ray M, Chen Y, Su YA, Trent JM. Use of a cDNA microarray to analyse gene expression patterns in human cancer. Nat Genet. 1996;14:457-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1283]  [Cited by in F6Publishing: 1367]  [Article Influence: 48.8]  [Reference Citation Analysis (0)]
27.  Takemasa I, Higuchi H, Yamamoto H, Sekimoto M, Tomita N, Nakamori S, Matoba R, Monden M, Matsubara K. Construction of preferential cDNA microarray specialized for human colorectal carcinoma: molecular sketch of colorectal cancer. Biochem Biophys Res Commun. 2001;285:1244-1249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 47]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
28.  Schuebel KE, Movilla N, Rosa JL, Bustelo XR. Phosphorylation-dependent and constitutive activation of Rho proteins by wild-type and oncogenic Vav-2. EMBO J. 1998;17:6608-6621.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 225]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
29.  Toh Y, Oki E, Oda S, Tokunaga E, Ohno S, Maehara Y, Nicolson GL, Sugimachi K. Overexpression of the MTA1 gene in gastrointestinal carcinomas: correlation with invasion and metastasis. Int J Cancer. 1997;74:459-463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
30.  Nicolson GL, Nawa A, Toh Y, Taniguchi S, Nishimori K, Moustafa A. Tumor metastasis-associated human MTA1 gene and its MTA1 protein product: role in epithelial cancer cell invasion, proliferation and nuclear regulation. Clin Exp Metastasis. 2003;20:19-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 112]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
31.  Giannini R, Cavallini A. Expression analysis of a subset of coregulators and three nuclear receptors in human colorectal carcinoma. Anticancer Res. 2005;25:4287-4292.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Li M, Lin YM, Hasegawa S, Shimokawa T, Murata K, Kameyama M, Ishikawa O, Katagiri T, Tsunoda T, Nakamura Y. Genes associated with liver metastasis of colon cancer, identified by genome-wide cDNA microarray. Int J Oncol. 2004;24:305-312.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Koehler A, Bataille F, Schmid C, Ruemmele P, Waldeck A, Blaszyk H, Hartmann A, Hofstaedter F, Dietmaier W. Gene expression profiling of colorectal cancer and metastases divides tumours according to their clinicopathological stage. J Pathol. 2004;204:65-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 76]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
34.  Komori T, Takemasa I, Higuchi H, Yamasaki M, Ikeda M, Yamamoto H, Ohue M, Nakamori S, Sekimoto M, Matsubara K. Identification of differentially expressed genes involved in colorectal carcinogenesis using a cDNA microarray. J Exp Clin Cancer Res. 2004;23:521-527.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  D'Arrigo A, Belluco C, Ambrosi A, Digito M, Esposito G, Bertola A, Fabris M, Nofrate V, Mammano E, Leon A. Metastatic transcriptional pattern revealed by gene expression profiling in primary colorectal carcinoma. Int J Cancer. 2005;115:256-262.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 79]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]