Acessibilidade / Reportar erro

Combination of meta-analysis and graph clustering to identify prognostic markers of ESCC

Abstract

Esophageal squamous cell carcinoma (ESCC) is one of the most malignant gastrointestinal cancers and occurs at a high frequency rate in China and other Asian countries. Recently, several molecular markers were identified for predicting ESCC. Notwithstanding, additional prognostic markers, with a clear understanding of their underlying roles, are still required. Through bioinformatics, a graph-clustering method by DPClus was used to detect co-expressed modules. The aim was to identify a set of discriminating genes that could be used for predicting ESCC through graph-clustering and GO-term analysis. The results showed that CXCL12, CYP2C9, TGM3, MAL, S100A9, EMP-1 and SPRR3 were highly associated with ESCC development. In our study, all their predicted roles were in line with previous reports, whereby the assumption that a combination of meta-analysis, graph-clustering and GO-term analysis is effective for both identifying differentially expressed genes, and reflecting on their functions in ESCC.

esophageal squamous cell carcinoma; meta-analysis; graph clustering


Combination of meta-analysis and graph clustering to identify prognostic markers of ESCC

Hongyun GaoI, * Send correspondence to: Shitao Cui Department of Cardiothoracic Surgery Shanghai 10th People's Hospital Tongji University, n. 301 Yanchang Middle Road, 200072 Shanghai, China E-mail: cuishitaodr@gmail.com. ; Lishan WangII, * Send correspondence to: Shitao Cui Department of Cardiothoracic Surgery Shanghai 10th People's Hospital Tongji University, n. 301 Yanchang Middle Road, 200072 Shanghai, China E-mail: cuishitaodr@gmail.com. ; Shitao CuiIII; Mingsong WangIII

IDepartment of Emergency, Shanghai 10th People's Hospital, Tongji University, Shanghai, China

IIDepartment of Molecular Biology, Shanghai Jiao Tong University, Shanghai, China

IIIDepartment of Cardiothoracic Surgery, Shanghai 10th People's Hospital, Tongji University, Shanghai, China

Send correspondence to Send correspondence to: Shitao Cui Department of Cardiothoracic Surgery Shanghai 10th People's Hospital Tongji University, n. 301 Yanchang Middle Road, 200072 Shanghai, China E-mail: cuishitaodr@gmail.com.

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is one of the most malignant gastrointestinal cancers and occurs at a high frequency rate in China and other Asian countries. Recently, several molecular markers were identified for predicting ESCC. Notwithstanding, additional prognostic markers, with a clear understanding of their underlying roles, are still required. Through bioinformatics, a graph-clustering method by DPClus was used to detect co-expressed modules. The aim was to identify a set of discriminating genes that could be used for predicting ESCC through graph-clustering and GO-term analysis. The results showed that CXCL12, CYP2C9, TGM3, MAL, S100A9, EMP-1 and SPRR3 were highly associated with ESCC development. In our study, all their predicted roles were in line with previous reports, whereby the assumption that a combination of meta-analysis, graph-clustering and GO-term analysis is effective for both identifying differentially expressed genes, and reflecting on their functions in ESCC.

Key words: esophageal squamous cell carcinoma, meta-analysis, graph clustering.

Introduction

Esophageal squamous cell carcinoma (ESCC) is one of the six most common cancers worldwide, especially in China and other Asian countries (Lehrbach et al., 2003; Zhi et al., 2003). Despite improvements in detection, surgical techniques and chemoradiotherapy, the five-year survival rate remains low (Kato et al., 2002). Prediction is usually according to tumor, node and metastasis system (TNM) classification. However, TNM classification merely reflects the status of cancer progression at the time of diagnosis. In contrast, molecular biological analysis clarifies biological behavior during cancer progression. Thus, the combination of the two could be more accurate in reflecting the clinical outcome (Takeno et al., 2007).

Recently, it was discovered that certain genes could be associated with inadequate ESCC prognosis. For example, the high expression of CCR7 (Ding et al., 2003), COX2 (Takatori et al., 2008), Beclin-1 (Chen et al., 2009), TLR9 (Takala et al., 2011), and FOXC2 (Nishida et al., 2011), could be significantly correlated with invasion, stage, tumor depth, lymph node metastasis, and poor survival. Overexpression of steroid receptor coactivatior-3(SRC-3) was more frequently observed in primary ESCC in the late T stages (T3/T4) than in the earlier T1/T2 (Xu et al., 2007). Although over-expression of cysteine-rich 61 (Cyr61) was related to less overall survival of patients in stage I/II, there was no affect on the overall survival of patients in stage III/IV (Xie et al., 2011). Low claudin-4 expression was found to be significantly associated with histological differentiation, invasion depth, and lymph node metastasis. Low claudin-4 expression revealed an unfavorable influence on disease-free and overall survival (Sung et al., 2011). PRL-1 protein expression significantly correlated with the stage of ESCC, 79.4% of the cases (27/34) in stage III ESCC, and 33.3% of the cases (1/3) in stage 1 ESCC (Yuqiong et al., 2011).

However, additional molecular prognostic markers are essential as aids in developing more effective therapeutic strategies for better prognosis. In this study, the aim was to identify more differentially expressed genes in ESCC, and predict their underlying functions. Meta-analysis provides a powerful tool for analyzing microarray experiments by combining data from multiple studies, besides presenting unique computational challenges. The Bioconductor package RankProd provides a novel and intuitive tool for this, by detecting differentially expressed genes by means of the non-parametric rank product method (Hong et al., 2006). The graphclustering approach was used for identifying gene expression profiles that distinguish ESCC from normal samples. Furthermore, the relevant pathways in the cluster were also analyzed by GO term analysis, to so explain potential mechanisms in response to ESCC.

Data and Methods

Meta-analysis for expression profile and differentially expressed gene (DEG) analysis

Two ESCC related expression profiles, GSE23400 and GSE20347, were obtained separately from a public functional genomics data repository GEO, based on the Affymetrix Human Genome U133A Array and Affymetrix Human Genome U133A 2.0 Array, respectively.

In the GSE23400 dataset, 53 ESCC and 53 matched-normal samples were analyzed. Contributors chose not to include clinical phenotypes in their GEO submission (Su et al., 2011). In the GSE20347 dataset, 17 ESCC and 17 matched-normal samples were approved by the Institutional Review Boards of the Shanxi Cancer Hospital, and the US National Cancer Institute (NCI). Cases diagnosed with ESCC between 1998 and 2001 in the Shanxi Cancer Hospital in Taiyuan, Shanxi Province, PR China, and considered candidates for curative surgical resection, were identified and recruited for participation in the study. None of the cases had undergone prior therapy. Shanxi was the ancestral home of all of them. After obtaining informed consent, cases were interviewed, as a means of obtaining information on demographics, cancer risk factors (e.g., smoking, alcohol consumption, and a detailed family history of cancer), and clinical information (Yoo et al., 2008; Hu et al., 2010).

Statistical analysis

DEGs for the GSE23400 and GSE20347 dataset were first independently identified with the limma method, whereupon the RankProd package was then applied to overcome heterogeneity. Only those with a percentage of false-positives (PFP) (Hong et al., 2006) = 1% were considered differentially expressed between treatments and controls.

The Spearman rank correlation (r) was used for assaying comparative target-gene correlations, to thus demonstrate the potential connection between DEGs. This coefficient (Cureton 1965), which is conceptually similar to the Pearson correlation, measures the strength of the associations between two variables. The significance level was set at r >0.9, which is a more stringent threshold than the empirical value (Fukushima et al., 2011). Detection of this level was with DPClus. All statistical tests were carried out with the R program. A detailed workflow is shown in Figure 1.


Co-expression Network analysis and graph-clustering

DPClus (Altaf-Ul-Amin et al., 2006), a graph-clustering algorithm that can extract densely connected nodes as a cluster, was used to identify co-expressed groups. It is based on the density-and-periphery tracking of clusters. In this study, the overlapping-mode with DPClus settings was used. The parameter settings of cluster property cp; density values were set to 0.5 (Fukushima et al., 2011).

GO Term enrichment analysis

The use of Gene Ontology (GO) terms by collaborating databases facilitates uniform queries. The controlled vocabularies are structured for queries at different levels, thereby also facilitating the assignment of properties to genes or gene products, also at different levels, depending on the depth of knowledge involved.

DAVID (Huang da et al., 2009) was used to identify which GO terms were significantly over-represented in the biological process. The terms with p-value <0.05 and count numbers >2 were considered as significant (Boyle et al., 2004; Guo et al., 2006).

Results

Differently expressed gene selection and correlation network construction

The publicly available microarray data sets, GSE23400 and GSE20347, were obtained from GEO. Differentially expressed genes (DEGs) with fold change >2 and p-value <0.05 were selected by microarray analysis. Following limma method analysis, 519 genes from GSE23400 and 1360 from GSE20347 were selected as DEGs. On applying RankProd packages for meta-analysis, 9 up-regulated genes and 1876 down-regulated ones, with a percentage of false-positives (PFP) 1% and fold change value >2, were considered differentially expressed. 1885 DEGs were finally collected after meta-analysis (Figure 2).


To obtain the relationships among DEGs, the co-expressed value r = 0.9 and corrected p-value = 0.01 were considered as threshold. Finally, a correlation network was constructed from 724 relationships among 202 DEGs (Figure 3).


Graph-clustering identifies modules significantly enriched in biochemical pathways

At r >0.9, DPClus (Altaf-Ul-Amin et al., 2006) was used for identifying 6 clusters, ranging in size from 8 to 22 genes, in the correlation network of ESCC (Figure 3). Clusters 1, 4, 5 and 6, in particular, have mutual connections since they share the same genes. The more genes shared, the greater mutual connectivity (corresponding to thicker lines). Graph-clustering results are presented in Figure 4. The over-represented GO terms in the clusters were used to assess their significance. The results of graph-clustering by GO term enrichment analysis appear in Table 1.


Significant GO Terms (p-value <0.05, using the hypergeometric test) were listed, as to fucose catabolic process, cytoskeleton organization, epidermis development, response to injury, etc. (Table 1). Ectoderm development (GO:0007398), epidermis development (GO:0008544) and epidermal cell differentiation (GO:0009913) were commonly enriched in clusters 4 and 5. Keratinization (GO:0031424) was enriched in clusters 1 and 4.

Discussion

In this study, 1885 DEGs were first identified through meta-analysis. Among these, a correlation network was constructed with 202 DEGs producing 724 relationships. By applying graph-clustering, these 202 DEGs were then clustered into six clusters. Clusters 1, 4, 5 and 6 seemed to be mutually connected. Details of these connections were confirmed by GO-term enrichment analysis, whereby it was shown that the genes of cluster 1 may be involved in the fucose catabolic process, whereas those in clusters 4 and 5 were mainly associated with epidermis development and differentiation. Although there was no connection between cluster 2 and the others, it was also significantly effective in ESCC invasion and metastasis. The genes in this cluster were related to cytoskeleton and extracellular matrix organization. Therefore, the proposal is to speculate on certain genes in these clusters, identifiably involved in ESCC.

Cluster 2: CXCL12

CXCL12, also known as stromal cell-derived factor-1, was first discovered among the chemokines secreted by mouse bone-marrow stromal cells, with CXCR4 as its specific receptor. The two interact to form a coupled molecular pair, which plays a prominent role in regulating directional migration and proliferation in ESCC, where both are positively located within the membrane and cytoplasm (Wang et al., 2009). Expression of the two is significantly correlated with lymph node metastasis, in the tumor stage, gender and lymphatic invasion. The overall and disease-free survival rate is significantly lower in patients with positive CXCL12 expression than in those with negative (Sasaki et al., 2009). Furthermore, the CXCL12-CXCR4 combination possibly induces up-regulated expression of matrix metalloproteinases, whereby their further involvement in extracellular matrix modeling and the mediation of metastasis in ESCC, (Zhang et al., 2005a; Bartolomé et al., 2006; Lu et al., 2010). Apparently, MMP-7, MMP-9, and MT1-MMP are also closely associated with invasion depth and venous invasion in ESCC (Samantaray et al., 2004). This predicted mechanism was in accordance with our GO-term analysis.

Cluster 3: CYP2C9

In ESCC, higher CYP2C9 expression levels occur in the early tumor stages (pT1/pT2), compared to more advanced local tumors (pT3/pT4). Its selective inhibition decreases tumor-cell proliferation and G0/G1 phase cell-cycle arrest (Schmelzle et al., 2010). Although, even with GO-term analysis, the role of these genes in cluster 3 remained unknown, the inference is their possible involvement in the tumor-cell cycle, in accordance with their function.

Cluster 4: TGM3 and MAL genes

TGM3 is a member of a family of Ca2+-dependent enzymes, thought to be critically involved in the cross-linking of structural proteins and formation of the cell envelope (CE), thereby contributing to the rigid structures that play vital roles in shape determination and barrier functions. The down-regulation of TGM3 genes in human ESCC tissues may lead to incapacity to form CEs and sustain toxic material (Chen et al. 2000; Luo et al. 2004). Although TGM3 expression is significantly inversely correlated with the histological grade of esophageal carcinoma, there is no obvious correlation with lymph node metastasis and depth of invasion. So, there is every indication that this gene may be an important adhesion molecule expressed by epithelial cells, and thus regarded as an invasion suppressor molecule in ESCC (Liu et al., 2006). Although details of its role in ESCC were not clear, it is proposed that it may be involved in esophageal epithelial cell differentiation and keratinocyte differentiation, as previous described (Zhang et al., 2005b). The MAL gene, a T-cell differentiation antigen, was found to be down-regulated in 10 ESCC-patients bearing tumors in different stages of development (Kazemi-Noureini et al., 2004). It has already been shown to express in four alternatively spliced forms of transcripts during the intermediate and late stages of T-cell differentiation, and in that of epithelial cells (Alonso and Weissman, 1987). Its ectopic expression in carcinoma TE3 cells could lead to the repressed formation of tumors in nude mice, the inhibition of cell motility, and the production of apoptosis by the Fas pathway (Mimori et al., 2003), whereby the proposal that Mal may be a tumor suppressor gene in ESCC development.

Cluster 5: S100A9 gene

S100A9, a calcium-binding protein belonging to the S100 family, was detected as significantly down-regulated in ESCC (Ji et al., 2004). Moreover, S100A9 staining is decreased in poorly and moderately differentiated ESCCs, when compared with those well-differentiated, hence the inferrence that loss of S100A9 expression in ESCC generally occurs along with worsening esophageal epithelial differentiation in histological grades (Kong et al., 2004). These genes, together with the other 11 S100 genes and epidermal differentiation complex (EDC) genes, have been mapped to human chromosome 1q21, which is a region of structural and numerical aberration, involved in esophageal carcinogenesis and progression (Luo et al., 2004). In brief, down-regulation of S100A9 is a common event in esophageal carcinogenesis and progression through affecting epithelial cell differentiation, proliferation and apoptosis, as well as the expression of genes encoding epidermal structural proteins in a calcium-dependent manner (Luo et al. 2003; Zhi et al., 2003; Zhou et al., 2005).

Common genes in clusters 4 and 5: EMP-1 and SPRR3

Clusters 4 and 5 were closely connected by several genes (Figure 4), such as EMP-1 and SPRR3. The expression of EMP-1, a member of the PMP22 family, is found to be down-regulated in esophageal cancers (Zinovyeva et al., 2010). Over-expression of this gene in the EC9706 cell-line leads to inhibited cell proliferation, S-phase arrest, and G1-phase prolongation, thus indicating its participation in the cell-cycle. In addition, EMP-1 transfection could induce different forms of gene expression, such as integrin beta 7 (ITGB7), integrin beta 8 (ITGB8) and cadherin 5 (CDH5), involved in cell signaling, cell adhesion and cell-cell communication. Retinoic acid receptor beta (RAR-β) is another up-regulated gene induced by EMP-1. Retinoids can modulate epithelial cell-growth, differentiation, and apoptosis in vitro and in vivo by binding to specific nuclear retinoid receptors, such as RAR-β. They can also prevent abnormal squamous cell differentiation in non-keratinizing tissues. Therefore, the EMP-1 and RAR-β interaction might also play important roles in epithelial cell and keratinocyte differentiation (Wang et al., 2003).

SPRR3, one of the substrates for TGM3, is a small proline-rich protein, abundantly expressed in oral and esophageal epithelia. Its expression, although less concomitant with TGM3 loss in ESCC (Luo et al. 2004), is highly induced during human epidermal keratinocyte differentiation, thus being considered a differentiation marker of squamous epithelium (Abraham et al., 1996). Recent reviews imply that SPRR3 is frequently down-regulated in ESCC, when compared to adjacent paired mucosa (Chen et al., 2000a; de A Sim o et al., 2011). Further studies have shown that exogenous expression of SPRR3 significantly suppresses ESCC cell formation by inducing CDK11p46 protein expression and apoptosis (Zhang et al., 2008). Therefore, SPRR3 might play a crucial role in the maintenance of normal esophageal epithelial homeostasis.

Finally, the combination meta-analysis, graph-clustering and GO term analysis is presumedly effective for identifying differentially expressed genes, and speculating on their respective roles in ESCC. Hence, other unidentified genes will be the focus of our future studies.

Internet Resources

GEO public functional genomics data repository, http://www.ncbi.nlm.nih.gov/geo/ (September 27, 2011).

R program, http://www.r-project.org/ (September 27, 2011)

DPClus graph clustering program, http://kanaya.naist.jp/DPClus (September 27, 2011)

Gene Ontology, http://www.geneontology.org.

Received: October 16, 2011

Accepted: February 6, 2012.

Associate Editor: Jeremy A. Squire

License information: This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

*These authors contributed equally to this paper.

  • Abraham JM, Wang S, Suzuki H, Jiang HY, Rosenblum-Vos LS, Yin J and Meltzer SJ (1996) Esophagin cDNA cloning and characterization: A tissue-specific member of the small proline-rich protein family that is not expressed in esophageal tumors. Cell Growth Diff 7:855-860.
  • Alonso MA and Weissman SM (1987) cDNA cloning and sequence of MAL, a hydrophobic protein associated with human T-cell differentiation. Proc Natl Acad Sci USA 84:1997-2001.
  • Altaf-Ul-Amin M, Shinbo Y, Mihara K, Kurokawa K and Kanaya S (2006) Development and implementation of an algorithm for detection of protein complexes in large interaction networks. BMC Bioinformatics 7:e207.
  • Bartolomé RA, Molina-Ortiz I, Samaniego R, Sánchez-Mateos P, Bustelo XR and Teixidó J (2006) Activation of Vav/Rho GTPase signaling by CXCL12 controls membrane-type matrix metalloproteinase-dependent melanoma cell invasion. Cancer Res 66:248-258.
  • Boyle EI, Weng S, Gollub J, Jin H, Botstein D, Cherry JM and Sherlock G (2004) GO: TermFinder - Open source software for accessing Gene Ontology information and finding significantly enriched Gene Ontology terms associated with a list of genes. Bioinformatics 20:3710-3715.
  • Chen BS, Wang MR, Cai Y, Xu X, Xu ZX, Han YL and Wu M (2000a) Decreased expression of SPRR3 in Chinese human oesophageal cancer. Carcinogenesis 21:2147-2150.
  • Chen BS, Wang MR, Xu X, Cai Y, Xu ZX, Han YL and Wu M (2000b). Transglutaminase 3, an esophageal cancer-related gene. Int J Cancer 88:862-865.
  • Chen Y, Lu Y, Lu C and Zhang L (2009) Beclin-1 expression is a predictor of clinical outcome in patients with esophageal squamous cell carcinoma and correlated to Hypoxia-Inducible Factor (HIF)-1A expression. Pathol Oncol Res 15:487-493.
  • Cureton EE (1965) The average Spearman rank correlation when ties are present: A correction. Psychometrika 30:377.
  • de A Simão T, Souza-Santos PT, de Oliveira DS, Bernardo V, Lima SC, Rapozo DC, Kruel CD, Faria PA, Ribeiro Pinto LF and Albano RM (2011) Quantitative evaluation of SPRR3 expression in esophageal squamous cell carcinoma by qPCR and its potential use as a biomarker. Exp Mol Pathol. 91:584-589.
  • Ding Y, Shimada Y, Maeda M, Kawabe A, Kaganoi J, Komoto I, Hashimoto Y, Miyake M, Hashida H and Imamura M (2003) Association of CC chemokine receptor 7 with lymph node metastasis of esophageal squamous cell carcinoma. Clin Cancer Res 9:3406-3412.
  • Fukushima A, Kusano M, Redestig H, Arita M and Saito K (2011) Metabolomic correlation-network modules in Arabidopsis based on a graph-clustering approach. BMC Syst Biol 5:e1.
  • Guo L, Lobenhofer EK, Wang C, Shippy R, Harris SC, Zhang L, Mei N, Chen T, Herman D, Goodsaid FM, et al. (2006) Rat toxicogenomic study reveals analytical consistency across microarray platforms. Nat Biotechnol 24:1162-1169.
  • Hong F, Breitling R, McEntee CW, Wittner BS, Nemhauser JL and Chory J (2006) RankProd: A bioconductor package for detecting differentially expressed genes in meta-analysis. Bioinformatics 22:2825-2827.
  • Hu N, Clifford RJ, Yang HH, Wang C, Goldstein AM, Ding T, Taylor PR and Lee MP (2010) Genome wide analysis of DNA copy number neutral loss of heterozygosity (CNNLOH) and its relation to gene expression in esophageal squamous cell carcinoma. BMC Genomics 11:e576.
  • Huang da W, Sherman BT and Lempicki RA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Prot 4:44-57.
  • Ji J, Zhao L, Wang X, Zhou C, Ding F, Su L, Zhang C, Mao X, Wu M and Liu Z (2004) Differential expression of S100 gene family in human esophageal squamous cell carcinoma. J Cancer Res Clin Oncol 130:480-486.
  • Kato K, Hida Y, Miyamoto M, Hashida H, Shinohara T, Itoh T, Okushiba S, Kondo S and Katoh H (2002) Overexpression of caveolin-1 in esophageal squamous cell carcinoma correlates with lymph node metastasis and pathologic stage. Cancer 94:929-933.
  • Kazemi-Noureini S, Colonna-Romano S, Ziaee AA, Malboobi MA, Yazdanbod M, Setayeshgar P and Maresca B (2004) Differential gene expression between squamous cell carcinoma of esophagus and its normal epithelium; altered pattern of mal, akr1c2, and rab11a expression. World J Gastroenterol 10:1716-1721.
  • Kong JP, Ding F, Zhou CN, Wang XQ, Miao XP, Wu M and Liu ZH (2004) Loss of myeloid-related proteins 8 and myeloid-related proteins 14 expression in human esophageal squamous cell carcinoma correlates with poor differentiation. World J Gastroenterol 10:1093-1097.
  • Lehrbach DM, Nita ME and Cecconello I (2003) Molecular aspects of esophageal squamous cell carcinoma carcinogenesis. Arq Gastroenterol 40:256-261.
  • Liu W, Yu ZC, Cao WF, Ding F and Liu ZH (2006) Functional studies of a novel oncogene TGM3 in human esophageal squamous cell carcinoma. World J Gastroenterol 12:3929-3932.
  • Lu C, Ji Y, Ge D, Guo J and Ding J (2010) The expression of CXCR4 and its relationship with matrix metalloproteinase9/vascular endothelial growth factor in esophageal squamous cell cancer. Dis Esophagus 24:283-290.
  • Luo A, Kong J, Hu G, Liew CC, Xiong M, Wang X, Ji J, Wang T, Zhi H, Wu M, et al. (2004) Discovery of Ca2+-relevant and differentiation-associated genes downregulated in esophageal squamous cell carcinoma using cDNA microarray. Oncogene 23:1291-1299.
  • Mimori K, Shiraishi T, Mashino K, Sonoda H, Yamashita K, Yoshinaga K, Masuda T, Utsunomiya T, Alonso MA, Inoue H, et al. (2003) MAL gene expression in esophageal cancer suppresses motility, invasion and tumorigenicity and enhances apoptosis through the Fas pathway. Oncogene 22:3463-3471.
  • Nishida N, Mimori K, Yokobori T, Sudo T, Tanaka F, Shibata K, Ishii H, Doki Y and Mori M (2011) FOXC2 is a novel prognostic factor in human esophageal squamous cell carcinoma. Ann Surg Oncol 18:535-542.
  • Samantaray S, Sharma R, Chattopadhyaya T, Gupta SD and Ralhan R (2004) Increased expression of MMP-2 and MMP-9 in esophageal squamous cell carcinoma. J Cancer Res Clin Oncol 130:37-44.
  • Sasaki K, Natsugoe S, Ishigami S, Matsumoto M, Okumura H, Setoyama T, Uchikado Y, Kita Y, Tamotsu K, Hanazono K, et al. (2009) Expression of CXCL12 and its receptor CXCR4 in esophageal squamous cell carcinoma. Oncol Rep 21:65-71.
  • Schmelzle M, Dizdar L, Matthaei H, Baldus SE, Wolters J, Lindenlauf N, Bruns I, Cadeddu RP, Kröpil F, Topp SA, et al. (2010) Esophageal cancer proliferation is mediated by cytochrome P450 2C9 (CYP2C9). Prostaglandins Other Lipid Mediators 94:25-33.
  • Su H, Hu N, Yang HH, Wang C, Takikita M, Wang QH, Giffen C, Clifford R, Hewitt SM, Shou JZ, et al. (2011) Global gene expression profiling and validation in esophageal squamous cell carcinoma and its association with clinical phenotypes. Clin Cancer Res 17:2955-2966.
  • Sung CO, Han SY and Kim SH (2011) Low expression of claudin-4 is associated with poor prognosis in esophageal squamous cell carcinoma. Ann Surg Oncol 18:273-281.
  • Takala H, Kauppila JH, Soini Y, Selander K, Vuopala K, Lehenkari P, Saarnio J and Karttunen T (2011) Toll-like receptor 9 is a novel biomarker for esophageal squamous cell dysplasia and squamous cell carcinoma progression. J Innate Immun 3:631-638.
  • Takatori H, Natsugoe S, Okumura H, Matsumoto M, Uchikado Y, Setoyama T, Sasaki K, Tamotsu K, Owaki T, Ishigami S, et al. (2008) Cyclooxygenase-2 expression is related to prognosis in patients with esophageal squamous cell carcinoma. Eur J Surg Oncol 34:397-402.
  • Takeno S, Noguchi T, Takahashi Y, Fumoto S, Shibata T and Kawahara K (2007) Assessment of clinical outcome in patients with esophageal squamous cell carcinoma using TNM classification score and molecular biological classification. Ann Surg Oncol 14:1431-1438.
  • Wang DF, Lou N, Zeng CG, Zhang X and Chen FJ (2009) Expression of CXCL12/CXCR4 and its correlation to prognosis in esophageal squamous cell carcinoma. Ai Zheng 28:154-158.
  • Wang HT, Kong JP, Ding F, Wang XQ, Wang MR, Liu LX, Wu M and Liu ZH (2003) Analysis of gene expression profile induced by EMP-1 in esophageal cancer cells using cDNA microarray. World J Gastroenterol 9:392-398.
  • Xie J, Xu L, Wu Z, Li L, Xu X, Wu J, Huang Q and Li E (2011) Expression of cysteine-rich 61 is correlated with poor prognosis in patients with esophageal squamous cell carcinoma. Eur J Surg Oncol 37:669-674.
  • Xu FP, Xie D, Wen JM, Wu HX, Liu YD, Bi J, Lv ZL, Zeng YX and Guan XY (2007) SRC-3/AIB1 protein and gene amplification levels in human esophageal squamous cell carcinomas. Cancer Lett 245:69-74.
  • Yoo YA, Kang MH, Kim JS and Oh SC (2008) Sonic hedgehog signaling promotes motility and invasiveness of gastric cancer cells through TGF-beta-mediated activation of the ALK5-Smad 3 pathway. Carcinogenesis 29:480-490.
  • Yuqiong L, Xi L, Ke G, Jun-Yi L and Huixiang L (2011) Expression of phosphatase regenerating Liver-1 in esophageal squamous cell carcinoma associated with metastasis. ISRN Pathology 2011:5402-5408.
  • Zhang J, Sarkar S and Yong VW (2005a) The chemokine stromal cell derived factor-1 (CXCL12) promotes glioma invasiveness through MT2-matrix metalloproteinase. Carcinogenesis 26:2069-2077.
  • Zhang J, Hui Ying Z, Fang D, Ai Ping L and Liu ZH (2005b) Transglutaminase 3 expression in C57BL/6J mouse embryo epidermis and the correlation with its differentiation. Cell Res 15:105-110.
  • Zhang Y, Feng YB, Shen XM, Chen BS, Du XL, Luo ML, Cai Y, Han YL, Xu X, Zhan QM, et al. (2008) Exogenous expression of Esophagin/SPRR3 attenuates the tumorigenicity of esophageal squamous cell carcinoma cells via promoting apoptosis. Int J Cancer 122:260-266.
  • Zhi H, Zhang J, Hu G, Lu J, Wang X, Zhou C, Wu M and Liu Z (2003) The deregulation of arachidonic acid metabolism-related genes in human esophageal squamous cell carcinoma. Int J Cancer 106:327-333.
  • Zhou G, Li H, Gong Y, Zhao Y, Cheng J and Lee P (2005) Proteomic analysis of global alteration of protein expression in squamous cell carcinoma of the esophagus. Proteomics 5:3814-3821.
  • Zinovyeva MV, Monastyrskaya GS, Kopantzev EP, Vinogradova TV, Kostina MB, Sass AV, Filyukova OB, Uspenskaya NY, Sukhikh GT, et al. (2010) Identification of some human genes oppositely regulated during esophageal squamous cell carcinoma formation and human embryonic esophagus development. Dis Esophagus 23:260-270.
  • Send correspondence to:
    Shitao Cui
    Department of Cardiothoracic Surgery
    Shanghai 10th People's Hospital
    Tongji University, n. 301
    Yanchang Middle Road, 200072 Shanghai, China
    E-mail:
  • Publication Dates

    • Publication in this collection
      19 June 2012
    • Date of issue
      2012

    History

    • Received
      16 Oct 2011
    • Accepted
      06 Feb 2012
    Sociedade Brasileira de Genética Rua Cap. Adelmio Norberto da Silva, 736, 14025-670 Ribeirão Preto SP Brazil, Tel.: (55 16) 3911-4130 / Fax.: (55 16) 3621-3552 - Ribeirão Preto - SP - Brazil
    E-mail: editor@gmb.org.br