Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 May 1.
Published in final edited form as: Clin Cancer Res. 2011 Mar 8;17(9):2955–2966. doi: 10.1158/1078-0432.CCR-10-2724

Global gene expression profiling and validation in esophageal squamous cell carcinoma (ESCC) and its association with clinical phenotypes

Hua Su 1,*, Nan Hu 1,*, Howard H Yang 2, Chaoyu Wang 1, Mikiko Takikita 3, Quan-Hong Wang 4, Carol Giffen 5, Robert Clifford 2, Stephen M Hewitt 3, Jian-Zhong Shou 6, Alisa M Goldstein 1, Maxwell P Lee 2,**, Philip R Taylor 1,**
PMCID: PMC3086948  NIHMSID: NIHMS278650  PMID: 21385931

Abstract

Purpose

Esophageal squamous cell carcinoma (ESCC) is an aggressive tumor with poor prognosis. Understanding molecular changes in ESCC will enable identification of molecular subtypes and provide potential targets for early detection and therapy.

Experimental Design

We followed up a previous array study with additional discovery and confirmatory studies in new ESCC cases using alternative methods. We profiled global gene expression for discovery and confirmation, and validated selected dysregulated genes with additional RNA and protein studies.

Results

A total of 159 genes showed differences with extreme statistical significance (P<E-15) and ≥ 2-fold differences in magnitude (tumor/normal RNA expression ratio, N=53 cases), including 116 up-regulated and 43 down-regulated genes. Of 41 genes dysregulated in our prior array study, all but one showed the same fold change directional pattern in new array studies, including 29 with ≥ 2-fold changes. Alternative RNA expression methods validated array results: more than two-thirds of 51 new cases examined by RT-PCR showed ≥ 2-fold differences for all seven genes assessed. Immunohistochemical protein expression results in 275 cases were concordant with RNA for five of six genes.

Conclusion

We identified an expanded panel of genes dysregulated in ESCC and confirmed previously identified differentially-expressed genes. Microarray-based gene expression results were confirmed by RT-PCR and protein expression studies. These dysregulated genes will facilitate molecular categorization of tumor subtypes and identification of their risk factors, and serve as potential targets for early detection, outcome prediction, and therapy.

Keywords: esophageal squamous cell carcinoma (ESCC), Affymetrix oligomicroarray, RT-PCR, tissue microarray (TMA)

Background

Esophageal cancer is the sixth most common fatal human cancer in the world (1) and the fourth most common new cancer in China (2). Shanxi Province, a region in north central China, has among the highest esophageal cancer rates in China and nearly all of these cases are esophageal squamous cell carcinoma (ESCC). ESCC is an aggressive tumor which is typically diagnosed only after the onset of symptoms when prognosis is very poor. The 19% 5-year survival rate is fourth worst among all cancers in the USA (3). One promising strategy to reduce ESCC mortality is early detection, and a better understanding of the molecular mechanisms underlying esophageal carcinogenesis and its molecular pathology will facilitate the development of biomarkers for early detection.

The application of microarray analysis is a promising method for finding clinical biomarkers in various cancers and has been successful in identifying subsets of tumors (including ESCC) that correlate with clinical parameters such as survival, histological grade, invasive status, and response to therapy (4-12). Gene expression changes that distinguish patient outcomes are subtle or variable and it is unlikely that individual genes will successfully predict clinical behavior. Taken together, however, gene expression profiles can be used to generate accurate predictors and could give us a better understanding of the molecular alterations during carcinogenesis.

In earlier studies, we documented genomic changes in ESCC, including widespread allelic loss and frequent mutations in certain putative tumor suppressor genes (13-17). Using a cDNA microarray with 7689 human cDNA clones, we previously tested expression in 19 ESCC patients and found 41 significant differentially-expressed genes. Patients with and without a positive family history for upper gastrointestinal (UGI) tract cancers were also distinguishable by their gene expression patterns (18). To confirm these original results, we expanded our expression studies of ESCC to evaluate more cases using alternative methods, including examination of more genes as well as validation/replication of selected dysregulated genes in additional patients using different methods. This confirmatory study was primarily based on global gene expression profiling of 53 ESCC patients with the Affymetrix Human Genome U133 Set (U133A and 133B). To further validate/replicate our findings we compared these data with: (i) RNA expression in 51 additional ESCC cases for seven differentially-expressed genes using quantitative real time RT-PCR; (ii) RNA expression from micro-dissected tumor and normal tissues in 17 ESCC cases for 41 dysregulated genes using the Affymetrix Human Genome U133A v2.0; and (iii) protein expression of six genes using immunohistochemistry (IHC) in 275 ESCCs on a tumor tissue microarray (TMA).

Methods

Patient selection

Four different groups of patients with ESCC were evaluated in this study, and all were enrolled in our UGI cancer genetic studies project, a single institution study using a common research protocol. Patients enrolled in the project included consecutive cases of ESCC who presented to the Thoracic Surgery Department of the Shanxi Cancer Hospital in Taiyuan, Shanxi Province, People's Republic of China, between 1998-2001, who had no prior therapy for their cancer, and who underwent surgical resection of their tumor at the time of their hospitalization. Selection of patients for RNA studies was based solely on the availability of appropriate tissues for RNA testing (ie, consecutive testing of cases with available frozen tissue, tumor samples that were predominantly (>50%) tumor, and tissue RNA quality/quantity adequate for testing); patients without frozen tissues were included in the protein studies. After obtaining informed consent, patients were interviewed to obtain information on demographic and lifestyle cancer risk factors, and clinical data were collected. Selected demographic and clinical-pathologic features of the four different ESCC patient groups studied are shown in Table 1. In total, 396 different ESCC cases were evaluated. All cases were histologically confirmed as ESCC by pathologists at both the Shanxi Cancer Hospital and the NCI. This study was approved by the Institutional Review Boards of the Shanxi Cancer Hospital and the NCI.

Table 1. Summary of characteristics of patients in the 4 groups studied.

Study group by lab method applied

Cases studied with Affymetrix U133A/B set
(N=53)
Cases studied with Affymetrix U133A V2
(N=17)
Cases studied with real time qRT-PCR
(N=51)
Cases studied with tumor tissue microarray
(N=275)
Gender (% male) 0.64 0.35 0.61 0.66
Age (years, median) 58 53 54 57
Tobacco use (% yes) 0.59 0.13 0.53 0.6
Alcohol use (% daily or weekly) 0.53 0.12 0.14 0.21
Family history of upper gastrointestinal cancer (% yes) 0.32 0.47 0.33 0.26
Tumor grade (%)
 1 0.09 0.00 0.06 0.17
 2 0.72 0.88* 0.69 0.59
 3 0.19 0.12 0.25 0.23
 4 0.00 0.00 0.00 <0.01
Tumor stage (%)
 I 0.00 0.00 0.02 <0.01
 II 0.25 0.24 0.24 0.12
 III 0.74 0.76 0.75 0.87
 IV 0.02 0.00 0.00 0.01
Lymph node metastasis (% yes) 0.43 0.47 0.51 0.44
*

One case missing information

Tissue collection

Paired esophageal cancer and normal tissue distant to the tumor were collected during surgery. Tissues for RNA analyses were snap-frozen in liquid nitrogen and stored at -130°C until used, while tissues for IHC analyses were fixed in 70% alcohol and processed to paraffin.

Total RNA preparation

RNA was extracted by two methods. For the confirmatory analysis of ESCC cases with the Affymetrix U133A/B chip set and the validation/replication in cases using real time RT-PCR, total RNA was extracted by the Trizol method following the protocol of the manufacturer. Only tumor samples with high purity (≥ 50% tumor cells) were selected for this extraction and subsequent analyses. A second method of RNA extraction was used for the micro-dissected tissue samples. For these samples, five to ten consecutive 8-micron sections were cut from frozen tumor tissues and the normal counterpart tissues, and tumor and/or normal cells were manually micro-dissected under light microscopy. RNA from tumor and matched normal tissue was extracted using the protocol from PureLink RNA mini kit (Catalog number 12183-018A, Invitrogen, Carlsbad, CA, 92008, USA). For both extraction methods, the quality and quantity of total RNA were determined on the RNA 6000 Labchip/Agilent 2100 Bioanalyzer (Agilent Technology, Inc, Germantown, MD).

Probe preparation and hybridization

Each microarray experiment was performed using eight micrograms of total RNA obtained from either the Trizol or PureLink extraction methods. Probes were prepared according to the protocol provided by the manufacturer (19). Procedures included first strand synthesis, second strand synthesis, double-strand cDNA clean up, in vitro transcription, cRNA purification, and fragmentation. Twenty micrograms of biotinylated cRNA were finally applied to each hybridization array, either onto the Affymetrix GeneChip Human Genome U133 Set (HG_U133A and HG_U133B, Affymetrix, Santa Clara, CA) or the Affymetrix GeneChip Human Genome U133A 2.0 After hybridization at 45°C overnight, arrays were developed with phycoerythrin-conjugated streptavidin using a fluidics station (Genechip Fluidics Station 450, Santa Clara, CA) and scanned (Genechip Scanner 3000, Santa Clara, CA) to obtain quantitative gene expression levels. Paired tumor and normal tissue specimens from each patient were processed simultaneously during the RNA extractions and hybridizations.

Quantitative RT-PCR

Confirmation by real-time RT-PCR analysis for seven genes was performed on an ABI 7000 Sequence Detection System using paired tumor/normal ESCC samples from 51 cases as previously described (20). Briefly, one to five micrograms of total RNA were first converted to cDNA using Superscript II (Invitrogen Corporation) in the presence of an oligo (dT)12-18 primer, and 100 ng of cDNA was applied for the subsequent PCR reaction (94°C ×10 min; 95°C ×15 seconds, 60°C ×1 min; 40 cycles). Results of the real-time RT-PCR data are presented as CT values, where CT is defined as the threshold PCR cycle number at which an amplified product is first detected. The average CT was calculated for each gene evaluated and GAPDH, and the ΔCT was determined as the mean of the triplicate CT values for the evaluated gene minus the mean of the triplicate CT values for GAPDH. The ΔΔCT represents the difference between the paired tissue samples, as calculated by the formula ΔΔCT = (ΔCT of tumor - ΔCT of normal). The N-fold differential expression of the evaluated gene for a tumor sample compared with its normal epithelial counterpart was expressed as 2- ΔΔCT, which represents the fold change in the target gene expression in tumor normalized to an internal control gene (GAPDH) and relative to the normal control.

Immunohistochemistry (IHC) analysis of ESCC tumor microarray (TMA)

The details of patient selection and TMA construction were previously described (20). Six genes that were significantly over- or under-expressed on our previous 8K cDNA array were selected for IHC evaluation on the ESCC tumor TMA. These included CDC25B, LAMC2, FADD, KRT14, FSCN1 (all over-expressed) and KRT4 (under-expressed).

Slides were stained according to manufacturer's protocols for each of the seven gene proteins (for details, see Supplemental Table 1). In brief, five μm thickness deparaffinized sections were pretreated with 3% H2O2 in methanol for 10 minutes. Antigen retrieval included pressure cooker treatment for 5 or 25 minutes and 10% normal goat serum for one hour to block endogenous peroxidase activity, followed by incubation with primary antibodies at an appreciated dilution of 1:40 or 1:50 for overnight at 4°C. The next day the slides were treated using the secondary antibody (anti-mouse IgG (H+L), Vector Laboratories, Burlingame, CA, 1:500 dilution) for one hour at room temperature, followed by the ABC (Vector Laboratories, Burlingame, CA) solution for one hour at room temperature. Slides were developed with 0.02% 3′, 3′-diaminobenzidine solution (DAB, Sigma), counterstained with hematoxylin, dehydrated in ethanol, and cleared in xylene. These procedures were performed for all antibodies studied.

Immunohistochemical assessment

For assessment of gene proteins, two scores were assigned to each core: (i) the cytoplasmic staining intensity [categorized as 0 (absent), 1 (weak), 2 (moderate), or 3 (strong)]; and (ii) the percentage of positively stained epithelial cells [scored as 0 (0%), 1 (1-25%), 2 (26-50%), 3 (51-75%), or 4 (>75%)]. An overall protein expression score was calculated by multiplying the intensity and positivity scores (overall score range, 0-12). This overall score for each patient was further simplified by dichotomizing it to negative (overall score of ≤ 3) or positive (score of ≥ 4). Stains were reviewed by two pathologists (MK and SMH) and discussed to determine an appropriate analytic approach. Following the establishment of criteria, all cores on both arrays were read by a single pathologist (MT) using the described criteria.

Statistical analyses

Formal statistical analyses were applied only to cases studied with the Affymetrix U133A/B set. Data from the other three groups studied here were limited to descriptive statistics. For all the Affymetrix U133A/B array data, raw data sets (CEL files on all samples) after scanning were normalized using RMA, implemented in Bioconductor in R (21). The GEO accession numbers for these array data are GSE23400. Hierarchical clustering was performed to characterize RNA array expression patterns and distinguish differences between tumor and normal samples. Paired t-tests were used to identify differences in matched tumor/normal sample expression. Paired t-tests were all performed using the R package.

Results

Patient information

Characteristics of the 396 total patients evaluated here are shown in Table 1. The four separate study groups included cases evaluated by the Affymetrix U133A/B chip set (n=53), RT-PCR (n=51), the Affymetrix U133 V2 chip (n=17), and the tumor TMA (n=275). The median age for cases in the four study groups ranged from 53 to 58 years, males predominated in all but one of the groups, tobacco use (medians 13 to 60%) and alcohol use (medians 12 to 53%) were common as was a family history of UGI cancer (medians 26 to 47%). The vast majority of the tumors were grade 2, over three-fourths were stage III, and metastatic disease was evident for nearly half the cases.

Affymetrix U133A/B experimental quality control

In the present study, we used the Affymetrix Human U133 set (Chip A and Chip B) which contain 39,000 transcripts and variants, including approximately 33,000 well-substantiated human genes in greater than 45,000 probesets. We assayed hybridization quality using the Affymetrix GCOS software. The average MAS5 Present call of the 106 HG_U133A chips from the 53 ESCC patients was 50% (range 41 – 59%), average scale factor was 3.0 (range 1.7–8.7), average background was 58.5 (range 36.5-81.2), average noise was 2.5 (range 1.3-3.56), and ratio of 3′/5′ signal of house keeping gene GAPDH was 0.9 (range 0.7-1.3). Averages for the 102 HG_U133B chips from 51 ESCC patients (two cases had no total RNA left) were: Present call 34% (range 22 – 42%), scale factor 7.7 (range 4.4 –15.6), background 65.5 (range 36.8-145.3), noise 2.8 (range 1.5-5.9), and ratio of 3′/5′ signal of housekeeping gene GAPDH 1.0 (range 0.8-1.6). Other sample quality control parameters built into the chips by Affymetrix were also consistent with high quality data. Expression signals for all probesets were used for the analysis.

Hierarchical clustering analysis of gene expression data

We used hierarchical clustering to characterize gene expression for all tumor/normal tissue pairs that had both U133 A and B array data (n=51 pairs). First, we selected the 10% of probesets (n=4498) that had the highest variation across all 102 samples examined (variance > 0.31). An unsupervised 2-way hierarchical clustering analysis with the 4498 probesets clearly separated tumors from normal samples (Supplemental Figure 1). Only two normal samples and three tumors were misclassified based on this structure of two clusters. Tumors were further separated into several sub-clusters, although no clinical data, such as grade, stage, and metastasis, were associated with these sub-clusters.

Identification of genes differentially expressed between tumors and normal samples

To identify genes whose expression levels were altered in tumors, we performed paired t-tests for 53 cases with the Affymetrix U133A/B chip data. We found 642 genes (854 probesets) that showed significant differences in gene expression between tumor and normal tissues; these genes showed 2-fold or greater changes and were statistically significant after Bonferroni correction (ie, P-values less than 1.12E-6) (Supplemental Table 2). To highlight a shorter list of target genes, we also applied a more extreme p-value criterion (P-value <E-15) in conjunction with at least a 2-fold change, which identified 159 genes – 116 up-regulated genes (Table 2A) and 43 down-regulated genes (Table 2B).

Table 2.

Table 2A: Summary of over-expressed genes (P<E-15)*,

No. Symbol Gene name and related function Locus ID P-value Fold-change
Extracellular matrix
1 MMP1 matrix metalloproteinase 1 4312 8.61E-22 21.74
2 CTHRC1 collagen triple helix repeat containing 1 115908 8.63E-26 14.71
3 SPP1 secreted phosphoprotein 1 6696 5.56E-21 9.11
4 COL1A1 collagen, type I, alpha 1 1277 3.07E-25 7.94
5 COL1A2 collagen, type I, alpha 2 1278 6.15E-21 6.49
6 COL11A1 collagen, type XI, alpha 1 1301 3.40E-16 5.30
7 COL3A1 collagen, type III, alpha 1 1281 1.16E-19 4.48
8 COL5A2 collagen, type V, alpha 2 1290 1.34E-16 4.25
9 ANLN anillin, actin binding protein 54443 8.78E-17 4.18
10 PLAU plasminogen activator, urokinase 5328 5.52E-18 3.98
11 SPARC secreted protein, acidic, cysteine-rich 6678 7.78E-17 3.45
12 MFAP2 microfibrillar-associated protein 2 4237 1.73E-21 3.23
13 COL7A1 collagen, type VII, alpha 1 1294 2.34E-18 2.65

Cell adhesion
14 POSTIN periostin, osteoblast specific factor 10631 2.01E-18 8.37
15 CDH11 cadherin 11, type 2, OB-cadherin 1009 1.33E-18 5.17
16 CSPG2 chondroitin sulfate proteoglycan 2 1462 1.30E-17 4.61
17 LAMB3 laminin, beta 3 3914 4.81E-17 3.49
18 THBS2 thrombospondin 2 7058 8.35E-17 2.86
19 PTK7 PTK7 protein tyrosine kinase 7 5754 2.02E-18 2.02

DNA replication/repair/transcription
20 SNAI2 snail homolog 2 6591 1.35E-19 4.60
21 TOP2A topoisomerase (DNA) II alpha 170kDa 7153 1.14E-17 4.01
22 SOX4 SRY (sex determining region Y)-box 4 6659 6.78E-19 3.45
23 RFC4 replication factor C (activator 1) 4, 37kDa 5984 1.92E-23 3.29
24 GINS1 GINS complex subunit 1 9837 7.69E-20 2.94
25 HMGB3 high-mobility group box 3 3149 1.73E-16 2.94
26 MCM2 MCM2 minichromosome maintenance deficient 2, mitotin 4171 2.89E-20 2.70
27 GMNN geminin, DNA replication inhibitor 51053 4.69E-17 2.49
28 UHRF1 ubiquitin-like, containing PHD and RING finger domains, 1 29128 1.32E-16 2.42
29 MCM6 MCM6 minichromosome maintenance deficient 6 4175 1.58E-20 2.41
30 PCNA proliferating cell nuclear antigen 5111 4.42E-17 2.39
31 MCM5 MCM5 minichromosome maintenance deficient 5 4174 5.36E-16 2.34
32 MCM4 MCM4 minichromosome maintenance deficient 4 4173 4.32E-16 2.20
33 FEN1 flap structure-specific endonuclease 1 2237 2.96E-17 2.11
34 MSH6 mutS homolog 6 (E. coli) 2956 1.83E-16 2.10
35 TOPBP1 topoisomerase (DNA) II binding protein 1 11073 8.45E-18 2.10
36 FANCI Fanconi anemia, complementation group I 55215 3.80E-18 2.10
37 RAD51AP1 RAD51 associated protein 1 10635 1.70E-16 2.09
38 ZNF281 zinc finger protein 281 23528 3.47E-16 2.08

Cell growth/proliferation/differentiation factor
39 ECT2 epithelial cell transforming sequence 2 oncogene 1894 2.17E-22 4.32
40 ASPM asp (abnormal spindle)-like, microcephaly associated 259266 5.15E-18 3.23
41 PRC1 protein regulator of cytokinesis 1 9055 3.25E-16 3.12
42 FSCN1 fasin homolog 1, actin-bundling protein 6624 4.16E-19 3.06
43 NUSAP1 nucleolar and spindle associated protein 1 51203 3.14E-16 2.92
44 MET met proto-oncogene (hepatocyte growth factor receptor) 4233 5.76E-18 2.76
45 CDKN3 cyclin-dependent kinase inhibitor 3 1033 1.98E-17 2.47
46 TPX2 TPX2, microtubule-associated protein homolog 22974 5.17E-17 2.40
47 KIF20A kinesin family member 20A 10112 6.32E-18 2.19
48 AURKB aurora kinase B 9212 3.93E-16 2.09

Cell cycle regulators
49 CKS2 CDC28 protein kinase regulatory subunit 2 1164 2.78E-18 3.91
50 CEP55 centrosomal protein 55KDa 55165 1.53E-19 3.59
51 CDC20 CDC20 cell division cycle 20 homolog 991 2.79E-17 2.92
52 CDC2 cell division cycle 2 983 1.42E-19 2.91
53 FOXM1 forkhead box M1 2305 4.02E-18 2.90
54 CKS1B CDC28 protein kinase regulatory subunit 1B BUB1 budding uninhibited by benzimidazoles 1 homolog 1163 6.99E-21 2.89
55 BUB1B beta 701 5.45E-20 2.82
56 CCNB1 cyclin B1 891 1.45E-16 2.71
57 NUF2 cell division cycle associated 1 83540 6.23E-16 2.67
58 DLGAP5 discs, large homolog-associated 5 9787 7.79E-19 2.65
59 BIRC5 baculoviral IAP repeat-containing 5 332 3.86E-16 2.62
60 MARCKSL1 MARCKS-like 1 65108 2.86E-17 2.51
61 NEK2 NIMA (never in mitosis gene a)-related kinase 2 4751 3.88E-17 2.44
62 CENPF centromere protein F, 350/400ka (mitosin) 1063 1.57E-18 2.41
63 AURKA serine/threonine kinase 6 6790 3.67E-17 2.40
64 MAD2L1 MAD2 mitotic arrest deficient-like 1 4085 5.76E-16 2.36
65 CENPA centromere protein A 1058 5.93E-18 2.17
66 BUB1 BUB1 budding uninhibited by benzimidazoles 1 homolog 699 2.34E-17 2.09
67 ATR ataxia telangiectasia and Rad3 related 545 2.69E-18 2.06

Cell membrane protein/signal tranduction
68 FZD6 frizzled homolog 6 (Drosophila) 8323 2.29E-18 2.99
69 NETO2 neuropilin (NRP) and tolloid (TLL)-like 2 81831 1.06E-17 2.68
70 LAPTM4B lysosomal associated protein transmembrane 4 beta 55353 1.26E-17 2.55
71 PLXNA1 plexin A1 5361 3.73E-19 2.50
72 TBL1XR1 transducin (beta)-like 1X-linked receptor 1 79718 8.86E-17 2.17
73 EFNA1 ephrin-A1 1942 5.74E-18 2.15
74 PPIL5 peptidylprolyl isomerase (cyclophilin)-like 5 122769 9.88E-17 2.10
75 RANBP1 RAN binding protein 1 5902 1.61E-18 2.10
76 DPY19L4 dpy-19-like 4 286148 5.74E-16 2.09
77 LRRC8D leucine rich repeat containing 8 family, member D 55144 6.80E-16 2.09
78 ATP6V1C1 ATPase, H+ transporting 528 3.35E-16 2.08
79 TMEM185B transmembrane protein 185 B 79134 4.69E-16 2.07

Protein binding/modification/transportation/protein chaperon
80 DTL denticleless homolog (Drosophila) 51514 1.78E-18 2.86
81 IGF2BP2 insulin-like growth factor 2 mRNA binding protein 2 10644 4.70E-16 2.84
82 SERPINH1 serpin peptidase inhibitor, clade H, member 1, 871 1.88E-16 2.81
83 SMC2 structural maintenance of chromosomes 2 10592 5.63E-16 2.55
84 SLC12A4 solute carrier family 12, member 4 6560 1.39E-16 2.54
85 TFRC transferrin receptor (p90, CD71) 7037 6.63E-18 2.53
86 ATAD2 ATPase family, AAA domain containing 2 29028 6.46E-16 2.33
87 SLC16A1 solute carrier family 16, member 1 6566 6.79E-16 2.28
88 HSPH1 heat shock 105KDa/110KDa protein 10808 4.07E-16 2.28
89 UBE2T ubiquitin-conjugating enzyme E2T 29089 5.30E-16 2.24
90 HOMER3 homer homolog 3 (Drosophila) 9454 3.13E-16 2.31
91 AGRIN agrin 375790 9.27E-21 2.13
92 SLC33A1 solute carrier family 33 (acetyl-CoA transporter), member 1 9197 2.82E-16 2.12
93 NUP107 nucleoporin 107kDa 57122 7.19E-16 2.12
94 HSP90AA1 heat shock 90kDa protein 1, alpha 3320 1.97E-16 2.12
95 HSPE1 heat shock 10kDa protein 1 (chaperonin 10) 3336 1.13E-16 2.10

Biochemical enzymes activity
96 SULF1 sulfatase 1 23213 9.88E-20 4.36
97 MEST mesoderm specific transcript homolog (mouse) 4232 3.27E-18 2.92
98 MTHFD1L formyltetrahydrofolate synthetase domain containing 1 25902 2.66E-17 2.78
99 TTK TTK protein kinase 7272 4.79E-17 2.77
100 MTHFD2 methylene tetrahydrofolate dehydrogenase 10797 7.75E-16 2.68
101 PRKDC protein kinase, DNA-activated, catalytic polypeptide 5591 5.67E-18 2.40
102 MELK maternal embryonic leucine zipper kinase 9833 5.94E-16 2.39
103 HPRT1 hypoxanthine phosphoribosyltransferase 1 3251 6.76E-18 2.24
104 C20orf3 chromsome 20 oopen reading frame 3 57136 6.35E-20 2.16
105 DNMT1 DNA (cytosine-5-)-methyltransferase 1 1786 2.09E-18 2.15
106 GMPS guanine monphosphate synthetase 8833 3.14E-16 2.09
107 PTDSS1 phosphatidylserine synthase 1 9791 3.30E-17 2.07

Calcium ion binding/transport
108 ITPR3 inositol 1,4,5-triphosphate receptor, type 3 3710 7.09E-19 2.19

Others
109 KIF4A kinesin family member 4A 24137 2.87E-21 2.62
110 KIF14 kinesin family member 14 9928 9.05E-20 2.46
111 SR140 U2-associated SR140 protein 23350 1.64E-18 2.38
112 VOPP1 vesicular, overexpressed in cancer, prosurvival protein 1 81552 7.83E-16 2.36
113 ACTL6A actin-like 6A 86 4.17E-19 2.36
114 TRIP13 thyroid hormone receptor interactor 13 9319 2.16E-16 2.28
115 CBX3 chromobox homolog 3 (HP1 gamma homolog, Drosophila) 11335 1.47E-22 2.18
116 SLC20A1 solute carrier family 20 (phosphate transporter), member 1 6574 3.75E-17 2.13
Table 2B: Summary of under-expressed genes (P<E-15)*,

No. Symbol Genes name and related function Locus ID P-value Fold-change
DNA replication/transcription
1 KLF4 Kruppel-like factor 4 (gut) 9314 5.08E-16 0.43

Cell growth/proliferation/differentiation factor
2 NDRG2 NDRG family member 2 57447 2.93E-19 0.38

Cell membrane protein/signal transduction
3 MAL mal, T-cell differentiation protein 4118 2.78E-16 0.06
4 SIM2 single-minded homolog 2 (Drosophila) 6493 7.36E-18 0.35
5 EPS8L2 EPS8-like 2 64787 3.91E-16 0.38
6 UBL3 ubiquitin-like 3 5412 9.89E-19 0.40

Protein transportation/protein binding
7 SHROOM3 Shroom family member 3 57619 1.43E-19 0.29
8 SASH1 SAM and SH3 domain containing 1 23328 2.48E-16 0.38
9 SORBS2 Arg/Abl-interacting protein ArgBP2 8470 6.01E-19 0.40
10 CAB39L calcium binding protein 39-like 81617 1.50E-17 0.45
11 SH3GLB2 SH3-domain GRB2-like endophilin B2 56904 2.99E-16 0.46
12 MPP7 membrane protein, palmitoylated 7 143098 8.38E-18 0.47
13 SORT1 sortilin 1 6272 4.98E-16 0.48

Biochemical enzymes activity
14 PPP1R3C protein phosphatase 1, regulatory (inhibitor) subunit 3C 5507 2.88E-17 0.15
15 HPGD hydroxyprostaglandin dehydrogenase 15-(NAD) 3248 2.96E-18 0.16
16 ADH1B alcohol dehydrogenase IB (class I), beta polypeptide 125 1.44E-18 0.20
17 GPX3 glutathione peroxidase 3 (plasma) 2878 7.40E-16 0.23
18 FUT6 fucosyltransferase 6 (alpha (1,3) fucosyltransferase) 2528 2.93E-16 0.23
19 CFD complement factor D 1675 2.13E-18 0.23
20 MGLL monoglyceride lipase 11343 1.06E-16 0.28
21 PCAF p300/CBP-associated factor glycerophosphodiester phosphodiesterase domain containing 8850 7.04E-18 0.29
22 GDPD3 3 79153 3.31E-16 0.39
23 GPD1L glycerol-3-phosphate dehydrogenase 1-like 23171 4.34E-19 0.41
24 THSD4 thrombospondin, type I 79875 8.17E-16 0.41
25 HLCS holocarboxylase synthetase 3141 1.18E-17 0.42
26 ECHDC2 enoyl CoA hydratase domain containing 2 55268 6.38E-16 0.43
27 PADI1 peptidyl arginine deiminase, type I 29943 6.21E-16 0.45

Calcium ion binding
28 NUCB2 nucleobindin 2 4925 9.48E-17 0.25

Others
29 CRISP3 cysteine-rich secretory protein 3 10321 2.92E-17 0.05
30 ENDOU endonuclease, ployU-specific 8909 6.17E-16 0.15
31 GCOM1 GRINL1A complex upstream protein 145781 1.16E-18 0.23
32 LPIN1 lipin 1 23175 7.90E-17 0.28
33 SH3BGRL2 SH3 domain binding glutamic acid-rich protein like 2 83699 1.76E-19 0.30
34 CGNL1 cingulin-like 1 84952 4.74E-17 0.30
35 TP53INP2 tumor protein p53 inducible nuclear protein 2 58476 1.47E-16 0.37
36 KIAA0232 KIAA0232 gene product 9778 3.54E-18 0.42
37 UACA uveal autoantigen with coiled-coil domains 55075 8.51E-16 0.48

Unknown
38 C10orf116 chromosome 10 open reading frame 116 10974 1.98E-16 0.27
39 FAM46B family with sequence similarity 46, member B 115572 1.66E-17 0.28
40 C21orf81 chromosome 21 open reading frame 81 114035 2.46E-16 0.37
41 RMND5B required for meiotic nuclear division 5 homolog B 64777 4.82E-16 0.40
42 C15orf52 chromosome 15 open reading frame 52 388115 6.21E-17 0.48
43 COBL cordon-bleu homolog (mouse) 23242 6.89E-16 0.50
*

Genes ordered by magnitude of fold change within each sub-category

For genes with more than one probeset, only the most significant probset in the gene is shown.

Affymetrix U133 v2.0 micro-dissected tissue validation

In our initial RNA expression study (ie, 8K cDNA study) [18], we identified 41 differentially-expressed genes. As part of our validation efforts here, we also compared RNA expression for these 41 dysregulated genes by alternative methods, including different microarray platforms as well as different methods for RNA extraction and tissue procurement. These comparisons included results from three sets of analyses involving independent samples, consisting of the 8K cDNA study (N=19), the Affymetrix U133A/B chip set (N=53), and the Affymetrix U133 V2 chip (N=17). Both the 8K cDNA and the Affymetrix U133A/B chip studies used total RNA extracted with the Trizol method but without micro-dissection. The Affymetrix U133 V2 chip study employed micro-dissected tissues from which RNA was extracted with the PureLink protocol.

A cross-platform comparison between the 8K cDNA and Human U133A/B set showed that, of the 41 dysregulated genes from our previous study, 40 were evaluable on both platforms (one gene was not found in the Affymetrix probeset). Of these 40 genes, all but one (CD3EAP) showed the same gene expression pattern on both platforms (ie, both up- or both down-regulated) (Table 3). In addition to the directionality of the changes, the magnitude of the changes was also very similar: changes were 2-fold or greater for 10 of 13 (77%) up-regulated genes on both platforms, while 19 of 28 (68%) down-regulated genes showed fold changes of 0.50 or less on both platforms.

Table 3. Comparison of 41 dysregulated genes from previous 8K cDNA microarray with 2 Affymetrix microarrays.

No. Gene Fold change by microarray type*,

8K cDNA array
(N=19)
Affymetrix U133A/B array
(N=53)
Affymetrix U133A v2.0 array
(N=17)
Over-expressed genes

1 COL3A1 2.91 4.48 6.96
2 COL7A1 2.31 2.65 2.36
3 KRT14 2.31 2.50 2.60
4 FSCN1 2.22 3.06 2.96
5 SPARC 2.08 3.45 4.40
6 LAMC2 2.06 3.28 5.67
7 TAGLN2 2.06 1.54 1.30
8 FADD 2.04 2.30 5.44
9 CST1 2.04 3.47 7.40
10 HLA-B 2.03 1.43 1.56
11 CXCL10 2.03 1.88 6.69
12 CDC25B 2.01 2.22 3.49
13 COL1A2 2.01 6.49 11.82

Under-expressed genes

14 KRT4 0.16 0.17 0.13
15 TGM3 0.21 0.10 0.07
16 CSTA 0.26 0.53 0.53
17 CRCT1 0.27 0.11 0.06
18 SPRR1A 0.28 0.38 0.35
19 FOSL2 0.29 NA 0.64
20 UPK1A 0.29 0.32 0.17
21 EMP1 0.31 0.24 0.15
22 SPINK5 0.31 0.14 0.08
23 CSTB 0.34 0.28 0.39
24 C10orf116 0.34 0.27 NA
25 PRR4 0.35 0.42 0.92
26 SLURP1 0.36 0.13 0.06
27 KLK13 0.38 0.19 0.21
28 S100A9 0.38 0.45 0.35
29 CNN3 0.38 0.47 0.29
30 BTC 0.38 0.56 0.80
31 HEMGN 0.40 0.97 NA
32 PPL 0.41 0.24 0.17
33 EGR1 0.43 0.56 1.19
34 APC2 0.44 0.92 0.99
35 KLK11 0.45 0.33 0.24
36 HPGD 0.45 0.16 0.19
37 CD3EAP 0.47 1.33 1.61
38 DUSP5 0.47 0.27 0.21
39 EVPL 0.48 0.37 0.21
40 RARB 0.48 0.86 NA
41 CD48 0.50 0.91 0.78
*

NA = not available

Gene order based on 8K cDNA array values

A comparison of different RNA extraction methods applied to the Affymetrix platforms showed that, of 38 genes examined on both Affymetrix platforms, only one had a different expression pattern (Table 3). EGR1 was down-regulated on the Affymetrix U133A/B chip set (fold change 0.56), but up-regulated on the Affymetrix U133 v2 chip (fold change 1.19).

It is also apparent from inspection of the data in Table 3 that the magnitude of the fold changes among up-regulated genes appears to be highest in the micro-dissected tissue samples (ie, Affymetrix U133A). For example, among the 13 up-regulated genes, none tested on the 8K cDNA array showed a fold change of three or more, while six exceeded 3-fold changes on the Affymetrix U133A/B chip set, and eight were higher than 3-fold on the Affymetrix U133 V2 chip, including five cases which reached over 5-fold changes. Although less consistent, the magnitude of the fold changes among down-regulated genes also appeared to be more extreme in the micro-dissected tissue samples.

Quantitative real time RT-PCR validation

Seven genes were selected for validation (Table 4) in a new group of 51 ESCC cases as illustrative examples of the genes which showed the most prominent differences in either the current Affymetrix or the prior cDNA array evaluations. Among the seven selected genes, four were up-regulated (COL1A2, COL3A1, MET, and KRT14) and three were down-regulated (SPINK7/ECG2, HPGD, and SASHI). Briefly, in at least two-thirds or more of the 51 patients, all four up-regulated genes showed increased mRNA expression (≥ 2-fold in tumor vs normal) while all three down-regulated genes showed decreased mRNA expression (≤ 0.5-fold in tumor vs normal). Specifically, KRT14 was increased in 67% of cases, COL1A2 in 67%, COL3A1 in 84%, and MET in 72%. Likewise, ECG2 was decreased in 84% of cases, HPGD in 80%, and SASH1 in 67%.

Table 4. Summary of 7 genes validated with quantitative RT-PCR and comparison with microarray results.

RNA expression analysis method

Quantitative RT-PCR
(N=51)
Affymetrix U133A/B chip
(N=53)
8K cDNA chip
(N=19)*

No. Gene N (frequency)
Under-expressed
(fold change ≤ 0.5)
N (frequency)
Normal expression
(fold change 0.5001- 1.9999)
N (frequency)
Over-expressed
(fold change ≥ 2.0)
Fold change
(median)
Fold change
(average)
Fold change
(average)
Up-regulated genes

1 KRT14 5 (0.10) 12 (0.24) 34 (0.67) 7.3 2.5 2.31
2 COL1A2 2 (0.04) 15 (0.29) 34 (0.67) 3.4 6.49 2.01
3 COL3A1 2 (0.04) 6 (0.12) 43 (0.84) 6.8 4.48 2.91
4 MET 5 (0.10) 9 (0.18) 37 (0.72) 5.5 2.76 NA

Down-regulated genes

5 SPINK7/ECG2 43 (0.84) 2 (0.04) 6 (0.12) 0.02 0.08 NA
6 HPGD 41 (0.80) 5 (0.10) 5 (0.10) 0.07 0.16 0.45
7 SASH1 34 (0.67) 11 (0.21) 6 (0.12) 0.27 0.38 NA
*

NA = not available

Protein expression validation

Tumor tissue samples from 313 ESCC cases were arrayed on the tumor TMA. After exclusion of cores with inadequate tissue following sectioning and tissue transfer, a total of 275 ESCC cases had IHC-based protein expression data available for at least one of the six markers evaluated as part of our validation here (Table 1). Protein expression positivity (number of evaluable ESCC cases) was: CDC25B 59% (N=275), LAMC2 82% (275), FADD 15% (248), KRT14 33% (249), FSCN1 56% (231) and KRT4 84% (171).

Discussion

In the present study we compared genome-wide gene expression in the tumors from 53 ESCC cases to their matched normal tissue samples. We found that tumors and normal tissues had intrinsically different expression patterns and were easily separated into two clusters based on unsupervised 2-way hierarchical clustering analysis. We identified 642 genes whose gene expressions differed between tumors and normal tissues using typical criteria (at least 2-fold change and P-value less than 1.12E-6).

Several recent studies analyzed gene expression profiling for ESCC (22-25). One study also used Affymetrix chips but studied only 15 ESCC cases (24), while other studies applied cDNA microarrays with Cy3 or Cy5 labeling that examined more limited numbers of genes (22;23). The largest number of ESCC cases studied was a Japanese report of 54 cases examined with the Affymetrix Human U133 A chip; however, pair-matched normal tissues were not used in that study (26). Thus, among genome-wide expression studies employing the optimal design – pair-wise matched tumor/normal tissue comparisons – the present study is the largest (53 cases) and most comprehensive (33,000 genes) ESCC evaluation to date.

With extreme statistical criteria (P-values < E-15 and at least a 2-fold change) the number of dysregulated genes was reduced to 159 (Tables 2A and 2B). The functions of these 159 genes most prominently relate to biochemical enzymes (26 genes), protein transportation or binding (23 genes), DNA replication (20 genes), cell cycle regulation (19 genes), cell membrane proteins (16 genes), extracellular matrix (13 genes), and cell growth (11 genes). Some of these genes (eg, MMP, collagen families, keratins, CDC25B, calcium-binding S100 proteins, and Annexin families) have previously been shown to function in squamous cell differentiation, invasion, or proliferation (27-29).

Examination of mRNA expression by RT-PCR for seven array-dysregulated genes in an independent series of ESCC cases showed results that were highly comparable with both our current Affymetrix U133A/B chip data and the findings from our earlier cDNA microarray study (18). Taken together, these results indicate that gene expression profiles in ESCC are consistent across different platforms and that dysregulated gene expression is a reproducible biomarker discovery tool.

We previously found 41 differentially-expressed genes in ESCC cases using an 8K cDNA microarray (18). All 41 of these genes were evaluated in the current Affymetrix-based study and all showed the same tumor/normal expression ratio directionality, save for one gene (FOSL2). Both the cDNA and the Affymetrix arrays used total RNA extracted by the Trizol method. To minimize the impact of normal contamination of our tumor samples, we further evaluated these 41 genes using micro-dissected RNA procured from another set of 17 ESCC cases and tested with Affymetrix U133A v2.0 chips. Results showed that the tumor:normal expression ratio directionality was the same for most of the genes (88%), however, the magnitude of the fold changes was markedly higher in the micro-dissected as opposed to the non-micro-dissected samples (8K cDNA and U133A/B set chip studies). We presume that this reflected reduced heterogeneity of the tissue samples when micro-dissection was employed, and a consequent increased signal-to-noise ratio. For example, COL1A2 just reached the 2-fold change threshold in the 8K cDNA array study, was 6.6-fold increased in the Affymetrix U133A/B set array, but was nearly 12-fold increased when micro-dissected RNA was used with the Affymetrix U133A v2.0 array (Table 3). While COL1A2 was the most extreme and clear-cut example of this increased signal-to-noise ratio, among 38 (of the 41) genes evaluated here, 28 showed their most extreme fold changes (either increased or decreased) in micro-dissected samples. Our comparison studies show that micro-dissection is a powerful approach. Results here are in agreement with other observations showing that micro-dissection provides relatively pure cell populations that are particularly useful for interrogating specific targets of interest (30).

Results from all three experiments reported here show broadly uniform findings for the expression patterns of the 41 genes emphasized, with the largest fold changes predominantly from the array that used micro-dissected RNA. To our knowledge, this is the first report confirming differential gene expression performed using two different microarray platforms and two different RNA extraction methods.

We chose six genes to evaluate at the protein level by applying IHC techniques to our ESCC tumor TMA. Three of the up-regulated genes (CDC25B 59%, LAMC2 82%, and FSCN1 56%) showed positive protein expression in the majority of ESCC cases studied, results that were highly concordant with RNA expression results. The other two up-regulated genes showed positive protein expression but in less than half of the ESCC cases studied (KRT14 33% and FADD 15%). The down-regulated gene, KRT4, was positive for protein expression in 84% of ESCCs, which did not correlate well with RNA results.

CDC25B has been shown to be a potential early biomarker as its protein expression increased with morphologic progression across the continuum of normal to dysplasia to invasive ESCC in our previous study (20). Patterns of LAMC2 protein expression showed a strong relation to survival, suggesting a potential role in prognosis (20). The expression of FSCN1 protein in epithelial neoplasms has been described (31-34), but its expression in ESCC is still unknown. The present study showed that FSCN1 protein expression was observed in most ESCC tissue cores (68%), which is in accord with RNA expression findings. While KRT14 protein expression was high in ESCCs in the current study, dysplastic and normal esophagus tissues were not evaluated. Xue et al did evaluate normal, dysplastic, and invasive ESCCs within esophagectomies from the same cases and observed that protein expression positivity increased across this morphologic progression from 13% to 41% to 62%, respectively, suggesting some discrimination between clinically and diagnostically important categories (35).

KRT4 protein expression has previously been reported in several tumors of the upper digestive tract, including esophageal adenocarcinoma (36). We found KRT4 mRNA down-regulated in both our 8K and Affymetrix microarray studies, yet protein expression was positive in 84% of ESCCs in our tumor TMA. Chung et al (37) reported that KRT4 protein expression decreased in the transition from normal to dysplasia to invasive tumor in a study of six carefully characterized ESCC cases. Of additional interest, ESCC cases with higher KRT4 mRNA in the present study had longer survival.

In summary, we identified an expanded list of 642 dysregulated genes in ESCC. These genes provide potential new targets for early detection and treatment.

Supplementary Material

1
2
3

Acknowledgments

This research was supported by National Cancer Institute contract [N02-SC-66211] with the Shanxi Cancer Hospital and Institute; and by the Intramural Research Program of the National Institutes of Health, National Cancer Institute, Division of Cancer Epidemiology and Genetics, and Center for Cancer Research.

Contributor Information

Hua Su, Email: suhu@mail.nih.gov.

Nan Hu, Email: nh38k@nih.gov.

Howard H Yang, Email: hy43h@nih.gov.

Chaoyu Wang, Email: wangc@mail.nih.gov.

Mikiko Takikita, Email: mikitaki@belle.shiga-med.ac.jp.

Quan-Hong Wang, Email: zhongmeiketi@yahoo.com.cn.

Carol Giffen, Email: giffenc@imsweb.com.

Robert Clifford, Email: clifforr@mail.nih.gov.

Stephen M Hewitt, Email: hewitts@mail.nih.gov.

Jian-Zhong Shou, Email: zhouaiping@csco.org.cn.

Alisa M Goldstein, Email: ag26o@nih.gov.

References

  • 1.Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005;55:74–108. doi: 10.3322/canjclin.55.2.74. [DOI] [PubMed] [Google Scholar]
  • 2.Yang L, Parkin DM, Ferlay J, et al. Estimates of cancer incidence in China for 2000 and projections for 2005. Cancer Epidemiol Biomarkers Prev. 2005;14:243–50. [PubMed] [Google Scholar]
  • 3.Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;60:277–300. doi: 10.3322/caac.20073. [DOI] [PubMed] [Google Scholar]
  • 4.van d V, He YD, van't Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347:1999–2009. doi: 10.1056/NEJMoa021967. [DOI] [PubMed] [Google Scholar]
  • 5.Takahashi M, Rhodes DR, Furge KA, et al. Gene expression profiling of clear cell renal cell carcinoma: gene identification and prognostic classification. Proc Natl Acad Sci U S A. 2001;98:9754–9. doi: 10.1073/pnas.171209998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Tamoto E, Tada M, Murakawa K, et al. Gene-expression profile changes correlated with tumor progression and lymph node metastasis in esophageal cancer. Clin Cancer Res. 2004;10:3629–38. doi: 10.1158/1078-0432.CCR-04-0048. [DOI] [PubMed] [Google Scholar]
  • 7.Ishibashi Y, Hanyu N, Nakada K, et al. Profiling gene expression ratios of paired cancerous and normal tissue predicts relapse of esophageal squamous cell carcinoma. Cancer Res. 2003;63:5159–64. [PubMed] [Google Scholar]
  • 8.Bhattacharjee A, Richards WG, Staunton J, et al. Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses. Proc Natl Acad Sci U S A. 2001;98:13790–5. doi: 10.1073/pnas.191502998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Selaru FM, Zou T, Xu Y, et al. Global gene expression profiling in Barrett's esophagus and esophageal cancer: a comparative analysis using cDNA microarrays. Oncogene. 2002;21:475–8. doi: 10.1038/sj.onc.1205111. [DOI] [PubMed] [Google Scholar]
  • 10.Hu YC, Lam KY, Law S, et al. Profiling of differentially expressed cancer-related genes in esophageal squamous cell carcinoma (ESCC) using human cancer cDNA arrays: overexpression of oncogene MET correlates with tumor differentiation in ESCC. Clin Cancer Res. 2001;7:3519–25. [PubMed] [Google Scholar]
  • 11.Kan T, Shimada Y, Sato F, et al. Gene expression profiling in human esophageal cancers using cDNA microarray. Biochem Biophys Res Commun. 2001;286:792–801. doi: 10.1006/bbrc.2001.5400. [DOI] [PubMed] [Google Scholar]
  • 12.Hu YC, Lam KY, Law S, et al. Identification of differentially expressed genes in esophageal squamous cell carcinoma (ESCC) by cDNA expression array: overexpression of Fra-1, Neogenin, Id-1, and CDC25B genes in ESCC. Clin Cancer Res. 2001;7:2213–21. [PubMed] [Google Scholar]
  • 13.Wu M, Hu N, Wang XQ. Genetic factor in the etiology of esophageal cancer and the strategy of its prevention in high-incidence areas of North China. In: Lynch HT, Hirayama T, editors. Genetic Epidemiology for Cancer. CRC Press Inc; Boca Raton, FL: 1989. pp. 187–200. [Google Scholar]
  • 14.Hu N, Dawsey SM, Wu M, et al. Familial aggregation of esophageal cancer in Yangcheng County, Shanxi Province, China. Int J Epidemiol. 1992;21:877–82. doi: 10.1093/ije/21.5.877. [DOI] [PubMed] [Google Scholar]
  • 15.Hu N, Li WJ, Su H, et al. Common genetic variants of TP53 and BRCA2 in esophageal cancer patients and healthy individuals from low and high risk areas of northern China. Cancer Detect Prev. 2003;27:132–8. doi: 10.1016/s0361-090x(03)00031-x. [DOI] [PubMed] [Google Scholar]
  • 16.Li WJ, Hu N, Su H, et al. Allelic loss on chromosome 13q14 and mutation in deleted in cancer 1 gene in esophageal squamous cell carcinoma. Oncogene. 2003;22:314–8. doi: 10.1038/sj.onc.1206098. [DOI] [PubMed] [Google Scholar]
  • 17.Hu N, Wang C, Su H, et al. High frequency of CDKN2A alterations in esophageal squamous cell carcinoma from a high-risk Chinese population. Genes Chromosomes Cancer. 2004;39:205–16. doi: 10.1002/gcc.10315. [DOI] [PubMed] [Google Scholar]
  • 18.Su H, Hu N, Shih J, et al. Gene expression analysis of esophageal squamous cell carcinoma reveals consistent molecular profiles related to a family history of upper gastrointestinal cancer. Cancer Res. 2003;63:3872–6. [PubMed] [Google Scholar]
  • 19.Affymetrix. Santa Clara, CA: Affymetrix; 2001. GeneChip Expression Analysis Technical Manual. Available from: URL: http://www.affymetrix.com/support/technical/manual/expression_manual.affx. [Google Scholar]
  • 20.Shou JZ, Hu N, Takikita M, et al. Overexpression of CDC25B and LAMC2 mRNA and protein in esophageal squamous cell carcinomas and premalignant lesions in subjects from a high-risk population in China. Cancer Epidemiol Biomarkers Prev. 2008;17:1424–35. doi: 10.1158/1055-9965.EPI-06-0666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fred Hutchinson Cancer Research Center. Bioconductor - Open Source Software for Bioinformatics. 2011 Available from: URL: http://www.bioconductor.org/
  • 22.Sato T, Iizuka N, Hamamoto Y, et al. Esophageal squamous cell carcinomas with distinct invasive depth show different gene expression profiles associated with lymph node metastasis. Int J Oncol. 2006;28:1043–55. [PubMed] [Google Scholar]
  • 23.Yamabuki T, Daigo Y, Kato T, et al. Genome-wide gene expression profile analysis of esophageal squamous cell carcinomas. Int J Oncol. 2006;28:1375–84. [PubMed] [Google Scholar]
  • 24.Uchikado Y, Inoue H, Haraguchi N, et al. Gene expression profiling of lymph node metastasis by oligomicroarray analysis using laser microdissection in esophageal squamous cell carcinoma. Int J Oncol. 2006;29:1337–47. [PubMed] [Google Scholar]
  • 25.Wong FH, Huang CY, Su LJ, et al. Combination of microarray profiling and protein-protein interaction databases delineates the minimal discriminators as a metastasis network for esophageal squamous cell carcinoma. Int J Oncol. 2009;34:117–28. [PubMed] [Google Scholar]
  • 26.Kashyap MK, Marimuthu A, Kishore CJ, et al. Genomewide mRNA profiling of esophageal squamous cell carcinoma for identification of cancer biomarkers. Cancer Biol Ther. 2009;8:36–46. doi: 10.4161/cbt.8.1.7090. [DOI] [PubMed] [Google Scholar]
  • 27.Zhang X, Zhi HY, Zhang J, et al. Expression of annexin II in human esophageal squamous cell carcinoma. Zhonghua Zhong Liu Za Zhi. 2003;25:353–5. [PubMed] [Google Scholar]
  • 28.Luo A, Kong J, Hu G, et al. Discovery of Ca2+-relevant and differentiation-associated genes downregulated in esophageal squamous cell carcinoma using cDNA microarray. Oncogene. 2004;23:1291–9. doi: 10.1038/sj.onc.1207218. [DOI] [PubMed] [Google Scholar]
  • 29.Lee DG, Bell SP. ATPase switches controlling DNA replication initiation. Curr Opin Cell Biol. 2000;12:280–5. doi: 10.1016/s0955-0674(00)00089-2. [DOI] [PubMed] [Google Scholar]
  • 30.Erickson HS, Gillespie JW, Emmert-Buck MR. Tissue microdissection. Methods Mol Biol. 2008;424:433–48. doi: 10.1007/978-1-60327-064-9_34. [DOI] [PubMed] [Google Scholar]
  • 31.Jawhari AU, Buda A, Jenkins M, et al. Fascin, an actin-bundling protein, modulates colonic epithelial cell invasiveness and differentiation in vitro. Am J Pathol. 2003;162:69–80. doi: 10.1016/S0002-9440(10)63799-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Grothey A, Hashizume R, Sahin AA, McCrea PD. Fascin, an actin-bundling protein associated with cell motility, is upregulated in hormone receptor negative breast cancer. Br J Cancer. 2000;83:870–3. doi: 10.1054/bjoc.2000.1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Pelosi G, Pastorino U, Pasini F, et al. Independent prognostic value of fascin immunoreactivity in stage I nonsmall cell lung cancer. Br J Cancer. 2003;88:537–47. doi: 10.1038/sj.bjc.6600731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hashimoto Y, Shimada Y, Kawamura J, et al. The prognostic relevance of fascin expression in human gastric carcinoma. Oncology. 2004;67:262–70. doi: 10.1159/000081327. [DOI] [PubMed] [Google Scholar]
  • 35.Xue LY, Hu N, Song YM, et al. Tissue microarray analysis reveals a tight correlation between protein expression pattern and progression of esophageal squamous cell carcinoma. BMC Cancer. 2006;6:296. doi: 10.1186/1471-2407-6-296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Taniere P, Martel-Planche G, Maurici D, et al. Molecular and clinical differences between adenocarcinomas of the esophagus and of the gastric cardia. Am J Pathol. 2001;158:33–40. doi: 10.1016/S0002-9440(10)63941-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Chung JY, Braunschweig T, Hu N, et al. A multiplex tissue immunoblotting assay for proteomic profiling: a pilot study of the normal to tumor transition of esophageal squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev. 2006;15:1403–8. doi: 10.1158/1055-9965.EPI-05-0651. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2
3

RESOURCES