Skip to main content

The database of chromosome imbalance regions and genes resided in lung cancer from Asian and Caucasian identified by array-comparative genomic hybridization

Abstract

Background

Cancer-related genes show racial differences. Therefore, identification and characterization of DNA copy number alteration regions in different racial groups helps to dissect the mechanism of tumorigenesis.

Methods

Array-comparative genomic hybridization (array-CGH) was analyzed for DNA copy number profile in 40 Asian and 20 Caucasian lung cancer patients. Three methods including MetaCore analysis for disease and pathway correlations, concordance analysis between array-CGH database and the expression array database, and literature search for copy number variation genes were performed to select novel lung cancer candidate genes. Four candidate oncogenes were validated for DNA copy number and mRNA and protein expression by quantitative polymerase chain reaction (qPCR), chromogenic in situ hybridization (CISH), reverse transcriptase-qPCR (RT-qPCR), and immunohistochemistry (IHC) in more patients.

Results

We identified 20 chromosomal imbalance regions harboring 459 genes for Caucasian and 17 regions containing 476 genes for Asian lung cancer patients. Seven common chromosomal imbalance regions harboring 117 genes, included gain on 3p13-14, 6p22.1, 9q21.13, 13q14.1, and 17p13.3; and loss on 3p22.2-22.3 and 13q13.3 were found both in Asian and Caucasian patients. Gene validation for four genes including ARHGAP19 (10q24.1) functioning in Rho activity control, FRAT2 (10q24.1) involved in Wnt signaling, PAFAH1B1 (17p13.3) functioning in motility control, and ZNF322A (6p22.1) involved in MAPK signaling was performed using qPCR and RT-qPCR. Mean gene dosage and mRNA expression level of the four candidate genes in tumor tissues were significantly higher than the corresponding normal tissues (P<0.001~P=0.06). In addition, CISH analysis of patients indicated that copy number amplification indeed occurred for ARHGAP19 and ZNF322A genes in lung cancer patients. IHC analysis of paraffin blocks from Asian Caucasian patients demonstrated that the frequency of PAFAH1B1 protein overexpression was 68% in Asian and 70% in Caucasian.

Conclusions

Our study provides an invaluable database revealing common and differential imbalance regions at specific chromosomes among Asian and Caucasian lung cancer patients. Four validation methods confirmed our database, which would help in further studies on the mechanism of lung tumorigenesis.

Peer Review reports

Background

Lung cancer is the leading cause of cancer death worldwide [1, 2]. There is mounting evidence of difference between Asian and Caucasian lung cancer patients in clinical and epidemiological aspects. For example, Lee et al. [3] reported that lung cancer in Taiwan and South China is characterized with a high percentage of female adenocarcinoma (ADC) and nonsmokers. In addition, response to certain anticancer treatments such as gefitinib is much greater in Asian patients than Caucasians [4]. However, these are rare large-scale attempts to identify the molecular characteristics of lung cancer between Asian and Caucasian. Therefore, identification and characterization of the chromosome regions with DNA copy number alteration in Western and Asian countries is important to unravel the mechanism underlying lung tumorigenesis.

Genomic DNA copy number variation is a hallmark of cancer and can lead to alteration in the expression and functions of genes residing within the affected chromosomal region [5]. Such a segment in the tumor genome may harbor either oncogenes or tumor suppressor genes depending on whether they are present in increased or decreased copy number, respectively. Identification of regions with copy number aberrations and the genes involved thus offers a basis for better understanding of cancer development [6]. Array-comparative genomic hybridization (array-CGH) can measure relative copy number at specific loci, which are represented by arrays of mapped bacterial artificial chromosomes (BAC) clones, cDNA clones, or various oligonucleotide-based formats [79]. The resolution of array-CGH is determined by the genomic spacing of the array elements thus is much more improved compared to previous conventional CGH [1012].

To identify the chromosome regions with DNA copy number alteration in patients from Western and Asian countries, we constructed a non-gapped array containing 3604 BAC clones covering 18 lung cancer-related chromosome imbalance hotspot regions identified in our previous study [13]. DNA from 40 Asian and 20 Caucasian non-small cell lung cancer (NSCLC) patients was analyzed using this specialized array to identify lung cancer-related genes either common or differential in these two ethnic groups. The Bayes statistical and comparative genomic approaches were used to identify genes with DNA copy number alterations. Our study provides a comprehensive database for chromosomal regions and genes with copy number imbalance in both Asian and Caucasian lung cancer patients. In addition, gene validation was performed using quantitative polymerase chain reaction (qPCR), reverse transcriptase-qPCR (RT-qPCR), chromogenic in situ hybridization (CISH), and immunohistochemistry (IHC) on specimens from the Asian and Caucasian lung cancer patients.

Methods

Clinical samples preparation and DNA/RNA extraction

Tissues were collected after obtaining appropriate institutional review board permission (VGHIRB No. 201001019IA) and informed consent from the recruited patients. Surgically resected tumor tissue and corresponding normal tissue were collected from 40 patients diagnosed with primary non-small cell lung cancer (NSCLC) admitted to Taipei Veterans General Hospital, Taiwan. Twenty NSCLC tumor tissues from Caucasian patients were obtained from the University of Chicago, USA. The list of these 60 patients along with the patients used for further validations is described in the Additional file 1: Table S1. Histological classification was determined according to the WHO classification system and the tumor-node-metastasis system. Information on the age, sex, tumor type, tumor stage and smoking history of the patients was obtained from hospital records. The genomic DNA was prepared using proteinase K digestion and phenol-chloroform extraction. Total RNA was prepared from tumors and normal lung tissues, using TRIzol reagent (Invitrogen, Carlsbad, CA). cDNA was generated using SuperScript reverse transcriptase (Invitrogen).

Array-CGH

The genomic microarray used contained 3604 BAC clones representing the human genome at 18 non-gap chromosome regions ( Additional file 2: Table S2). Clone mapping was analyzed using the BAC end pairs database of UCSC Genome browser (http://genome.ucsc.edu/, build 36 - hg18). The BAC library used was RP-11. The extracted BAC DNA was amplified to 10 μg using Phi29 DNA polymerase reaction (TempliPhi DNA Sequencing Template Amplification Kit, Amersham, Princeton, NJ), then digested with DNaseI (New England Biolabs, Ipswich, MA), and purified with MultiScreen-PCR Plates (Millipore, Danvers, MA). After digestion, the DNA fragments were resolved by gel electrophoresis and the fragment sizes were optimized to within 0.5–3 kb. Finally, the purified BAC DNA fragments were spotted in triplicate of 72 blocks onto the glass slides (Corning, NY) with spotting solution (50% DMSO, 50% dH2O). Representative array images are shown in the Additional file 3: Figure S1.

Approximately 500 ng of tumor genomic DNA and reference DNA [for Asian samples: 28 matched-normal genomic DNA pool; for Caucasian samples: male human genomic DNA (Promega, Leiden, the Netherlands)] were labeled by random priming using BioPrime DNA Labeling System (Invitrogen) with Cy3-dCTP (reference) or Cy5-dCTP (tumor). Labeled tumor and reference DNA were combined together with 100 μg human Cot-1 DNA (Invitrogen). Hybridization was performed using MAUI hybridization system (BioMcro Systems, Salt Lake City, UT) by agitating the hybridization solution for 50 hours at 50°C. The arrays were scanned using scanner GenePix4000B (Axon Instruments, Foster City, CA) and the images were segmented and transformed to Cy3-Cy5 log ratio using GenePix software (Axon Instruments, Inc). Further data processing, including normalization and data analysis was performed and is described in detail in the Result section.

Biological software analysis for candidate gene selection

The array results were further analyzed by MetaCore™ analysis software (GeneGo, Inc., St. Joseph, MI) to identify genes involved in cellular pathways, disease-specific biomarker genes, and genes annotated in the gene ontology databases. Calculation of statistical significance throughout MetaCore was based on P value, calculated based on hypergeometric distribution [14]. P value of 0.05 was used for the cutoff of candidate gene selection.

Real-time quantitative polymerase chain reaction (qPCR)

qPCR was used to confirm the array-CGH results and to determine the DNA copy number variations of identified candidate genes in 30 NSCLC tumor and corresponding normal samples from Asian, and 30 NSCLC tumor and 12 autopsy samples from Caucasian. DNA copy number was quantified by qPCR using the ABI 7900 Sequence Detection System (PE Applied Biosystems, Foster City, CA). Genomic DNA (100 ng) was amplified using the SYBR Green PCR Master Mix (Roche, Palo Alto, CA) and specific primers for each candidate gene located in exon and adjacent intron, and the GAPDH internal control gene located within the promoter region. The primer sequences and annealing temperature are listed in the Additional file 4: Table S3. The mean DNA dosage ratio between each candidate gene and the GAPDH internal control gene was calculated in all normal and all tumor samples analyzed [15].

Quantitative reverse transcription polymerase chain reaction (RT-qPCR)

RT-qPCR was used to measure the mRNA expression of four candidate genes in 45 NSCLC tumor and the corresponding normal samples from Asian, and 24 tumor and corresponding normal samples from Caucasian (TissueScan Lung Cancer Panel IV: HLRT304; OriGene, Rockville, MD). RT-qPCR was conducted using the ABI 7900 Sequence Detection System (PE Applied Biosystems). Total RNA (4 μg) from tumor and the corresponding normal tissues were reverse transcribed into cDNA and amplified using the SYBR Green PCR Master Mix (Roche) and specific primers for each candidate gene and the ß-actin internal control gene. The primer sequences and annealing temperature are listed in the Additional file 4: Table S3. The mean mRNA expression ratio between each candidate gene and the ß-actin internal control gene was calculated in all normal and all tumor samples analyzed.

Chromogenic in situhybridization (CISH)

CISH was done on 4 μm thick formalin-fixed paraffin embedded tissue section slides following the standard protocol described in Chang et al. [16]. The probes were digoxigenin-labeled with BAC DNA (RP11-375N9 and RP11-170O19 for ARHGAP19 gene probe; RP11-391k6 for ZNF322A gene probe), and then ethanol precipitated with 10 μg Human Cot-1 DNA. At least 100 non-overlapping and intact tumor nuclei were evaluated. The tumor was considered as CISH amplification positive if the copy number was ≥4 signals per nucleus in more than 40% of tumor cells evaluated [16].

Immunohistochemistry (IHC)

The protein expression level of the candidate gene, PAFAH1B1, was evaluated by IHC in 28 NSCLC samples from Asian and 37 lung cancer samples from Caucasian tissue array. Paraffin blocks of tumors were cut into 5-μm slices and then processed using standard deparaffinization and rehydration techniques. Polyclonal antibody against PAFAH1B1 (1:800; Novus, Littleton, CO) was used to detect the protein expression. The evaluation of IHC was conducted blindly without prior knowledge of the clinical and pathologic characteristics of the cases. The samples were graded high expression when >50% tumor cells were stained positive using adequate staining in surrounding normal stromal and epithelial cells.

Statistical analysis

The Bayes regression approach was used to determine the chromosomal breaking points [17]. Candidate gene selection by MetaCore (GeneGo, Inc.) was calculated based on hypergeometric distribution [14] and a P value of 0.05 was used for the cutoff. A two-tailed t test was used to determine the statistical significance of difference in DNA dosage and mRNA expression level of candidate genes in clinical samples. Chi-square test was conducted to examine the association between overexpression of candidate genes and clinical pathological parameters, including sex, age, tumor type, tumor stage, and smoking. A P < 0.05 was considered to be statistically significant.

Results

Generation of a specialized non-gapped CGH microarray representing 18 human chromosome imbalance hotspot regions

This study constructed a non-gapped BAC array covering 18 human chromosome regions including 2p22.1-p25.1, 2q34-q36.1, 3p11.2-p26.3, 4p15.1-p15.31, 4q33-q34.2, 6p12.3-p21.2, 6p22.1-p22.3, 9q21.1-q21.3, 10q21.3-25.1, 12p12.31-13.33, 12q14-q15, 13q12.3-14.2, 17p12-13.3, 17q24.2-25.3, Xp22.11, Xp22.13-22.31, Xq21.31-21.33, and Xq25-26.1 (see Additional file 2: Table S2). The chromosomal regions examined in the current array-CGH showed a high loss of heterozygosity (LOH) frequency in our previous genome-wide LOH study [13]. To construct this specialized array, genomic segments fully covering these 18 chromosome regions with 3604 BAC genomic clones without gaps were mapped. BAC clones were purified, amplified, and arrayed onto glass slides. The homogenous self-self hybridization image of reference DNA shown in the Additional file 3: Figure S1 (upper left panel) indicates the feasibility of our specialized array-CGH.

Block-wise normalization was conducted and a Bayes regression approach was used to identify the regions with chromosomal copy number alterations

Tumor DNA from 40 Asian NSCLC patients and 20 Caucasian NSCLC patients were labeled with Cy5 (red dye) and co-hybridized with reference DNA (for Asian samples: 28 matched-normal genomic DNA pool; for Caucasian samples: ten male genomic DNA purchased from Promega) labeled with Cy3 (green dye) for array-CGH. Clinical information for the patients is shown in the Additional file 1: Table S1. A “block-wise normalization” method, which normalized the array-CGH data by the log ratio of the self-self hybridization of reference DNA according to the mean and the standard deviations of each array block, was established (lower panel, Additional file 3: Figure S1). The Bayes regression approach was used to determine the chromosomal breakpoints [17]. A region was assigned +1, 0, or −1 when its gene dosage is higher, uncertain, or lower than those of its neighbors to define the chromosome alteration regions ( Additional file 3: Figure S1), respectively. Our array-CGH data has been submitted to Gene Expression Omnibus (GEO) database of NCBI with accession number GSE21276.

The chromosome regions show high frequency of chromosomal imbalance and significant correlation with specific cancer sub-type in Asian and Caucasian lung cancer patients

The chromosome regions with copy number alteration in 40 Asian and 20 Caucasian NSCLC patients were determined. A region with a frequency of gain or loss greater than 50% was defined as a significant chromosomal imbalance region [18, 19]. In samples from Asian lung cancer patients, we have identified eight regions of chromosomal gain and nine regions of chromosomal loss (Table 1A) harboring 476 genes (UCSC Genome browser). In Caucasian lung cancer patients, eleven chromosome regions with frequent gain and nine regions with frequent loss harboring a total of 459 genes were identified (Table 1B). Interestingly, seven common chromosomal imbalance regions harboring 117 genes, included gain on 3p13-14, 6p22.1, 9q21.13, 13q14.1, and 17p13.3; and loss on 3p22.2-22.3 and 13q13.3 (Table 1A-B) were found in both Asian and Caucasian patients. In addition, when tumors were grouped into two major NSCLC tumor subtypes, adenocarcinoma (ADC) and squamous cell carcinoma (SCC), six chromosomal imbalance regions showed difference of more than 50% in alteration frequency between ADC and SCC in Asian lung cancer patients (Table 2A). In Caucasian lung cancer patients, 14 chromosome regions showed significant difference between ADC and SCC patients (Table 2B). When tumors were grouped based on the tumor stage, three chromosomal imbalance regions with difference of more than 50% in alteration frequency between early stage (stages I and II) and late stage (stages III and IV) in Asian lung cancer patients (Table 3A) were found. In Caucasian lung cancer patients, five chromosome regions showed higher alteration frequency in late stage patients than early stage patients (Table 3B).

Table 1 Chromosome imbalance regions in lung cancer (alteration frequency > 50%)
Table 2 Chromosome imbalance regions with different alteration frequency in relation to tumor subtypes in lung cancer
Table 3 Chromosome imbalance regions with different alteration frequency in relation to tumor stages in lung cancer

The significance of altered biological processes in lung cancer

We then used Database for Annotation, Visualization and Integrated Discovery (DAVID) bioinformatics resources [20] to identify the difference of significantly altered biological process between Asian and Caucasian lung cancer patients. The results showed that there were seven common altered biological processes of gene ontology between Asian and Caucasian, including cation transport, metal ion transport, cellular chemical homeostasis, chromatin assembly or disassembly, protein-DNA complex assembly, nucleosome assembly, and nucleosome organization (Figure1A-B, indicated by * symbol). In addition, unique altered biological processes including monovalent inorganic cation transport, protein oligomerization, Wnt receptor signaling pathway, regulation of membrane potential, and response to estrogen stimulus were found in Asian lung cancer patients (Figure1A). Unique biological processes in Caucasian included cell surface receptor linked signal transduction, G-protein coupled receptor protein signaling pathway, ion homeostasis, cellular cation homeostasis, and chemotaxis (Figure1B).

Figure 1
figure 1

Pie chart of significantly altered biological processes in Asian (A) and Caucasian (B) lung cancer patients. The data were analyzed by DAVID bioinformatics resources. The common biological processes in both Asian and Caucasian marked with asterisk (*).

A total of 134 candidate NSCLC-related genes are selected by MetaCore analysis and integration analysis with the expression array database

To select potential NSCLC-related genes for further studies, the following strategy of candidate gene decision was performed (Figure2). First, a total of 818 genes from Asian and Caucasian databases with chromosome alteration frequency more than 50% were further analyzed by MetaCore™ analysis software. Calculation of statistical significance throughout MetaCore for cellular pathways, disease-specific biomarker genes, and genes annotated in the gene ontology databases was based on P value, calculated using the hypergeometric distribution [14]. P value of 0.05 was used for the cutoff of candidate gene selection. Collectively, 214 genes were selected as NSCLC-related candidate genes, which are involved in cell cycle, apoptosis, cell migration, signal transduction, DNA repair and other cell growth and motility control pathways (see Additional file 5: Table S4). Second, the candidate genes analyzed with MetaCore were further integrated with the expression array for Asian lung cancer patients (GEO database with accession number GSE21933) and the gene expression database of the National Cancer Institute's Cancer Genome Anatomy Project (http://cgap.nci.nih.gov) for Caucasian lung cancer patients (see Additional file 5: Table S4). We found 134 genes showing correlation between DNA copy number and mRNA expression patterns by integrating our array-CGH database with the expression array databases (Table 4). Finally, we performed literature search and selected 54 novel lung cancer candidate genes which have not been reported to be altered in lung cancer. In the current study, we validated four genes based on their novelty and antibody availability. These four genes were ARHGAP19 on 10q24.1 functioning in Rho activity control, FRAT2 on 10q24.1 involved in Wnt signaling, PAFAH1B1 on 17p13.3 functioning in motility control, and ZNF322A on 6p22.1 involved in MAPK pathway for following validation assays at the DNA, and RNA levels.

Figure 2
figure 2

Flowchart summarizing the strategy of candidate genes selection. First, a total of 818 genes with more than 50% frequency of chromosome alteration were analyzed by MetaCore software. Second, the candidate genes analyzed with MetaCore were further integrated with the gene expression database. Integrating our array-CGH database with the expression array databases, 134 genes showing correlation between DNA copy number and mRNA expression patterns were found. Finally, literature search was performed to select 54 novel lung cancer candidate genes for following validation assays at the DNA, and RNA levels.

Table 4 Candidate genes in lung cancer

A good correlation is found between array-CGH data and various validation methods including CISH, qPCR, RT-qPCR, and IHC

Four methods were employed for gene validation in 147 lung cancer patients. The clinical characteristics and the validation method conducted for each patient are shown in the Additional file 1: Table S1. qPCR for DNA copy number was performed for four candidate genes in tumors from 30 Asian and 30 Caucasian lung cancer patients, whose samples were analyzed in array-CGH. The mean gene dosage of lung cancer candidate genes ARHGAP19, FRAT2, PAFAH1B1, and ZNF322A in tumor tissues was significantly higher than the corresponding normal tissues in Asian lung cancer. Gene dosage of PAFAH1B1 and ZNF322A was significantly higher in tumor tissues than autopsy tissues from Caucasian (Figure3A). RT-qPCR of the four genes was conducted with cDNA from tumor and corresponding normal tissues of 45 Asian lung cancer patients. Only two candidate genes, PAFAH1B1 and ZNF322A, were analyzed in tumor and corresponding normal lung tissues from 24 Caucasian lung cancer patients due to the mRNA inavailability. The mean mRNA expression level of all genes analyzed in the tumor tissues was significantly higher than in the corresponding normal tissues in Asian and Caucasian patients (Figure3B). In addition, the validation data showed concordant results with array-CGH.

Figure 3
figure 3

DNA copy number validation by qPCR (A) and mRNA expression validation by RT-qPCR (B) of four candidate genes in clinical samples. The Y-axis is the mean DNA dosage ratio (A) or mean mRNA expression ratio (B) between candidate gene and the internal control gene in all samples analyzed. The P values of comparison between normal (N) and tumor (T) samples for each gene is as indicated.

CISH analysis of patients indicated that copy number amplification indeed occurred for ARHGAP19 and ZNF322A genes in lung cancer patients (Figure4A). To determine protein expression level of the selected candidate genes in lung cancer, IHC was performed for PAFAH1B1 protein on paraffin blocks from 28 Asian patients and tissue arrays of Caucasian patients including 37 lung cancer patients (Figure4B). The frequency of PAFAH1B1 protein overexpression was 68% in Asian and 70% in Caucasian.

Figure 4
figure 4

DNA copy number and protein expression level validation in specimens from lung cancer patients. (A) Representative CISH images of ARHGAP19 and ZNF322A genes in paraffin-embedded lung tumor tissue. Images taken from tumor without gene copy number amplification (left) and with gene copy number amplification (right). The enlarged image of one nucleus is shown on the lower right corner. CISH images: 400X magnification. (B) Representative IHC images of PAFAH1B1 protein in tissue array. Patients with protein overexpression are indicated by + symbol. Patients with normal expression are indicated by – symbol. IHC images: 200X magnification.

Discussion

Here, we generated a non-gapped specialized CGH microarray representing 18 human chromosome imbalance hotspot regions in lung cancer. This specialized CGH array identified the chromosomal regions whose structural alteration may play critical roles in lung tumorigenesis. Our array-CGH study provided an invaluable database of chromosomal imbalance regions and candidate genes in Asian and Caucasian lung cancer patients. CISH, qPCR, RT-qPCR, and IHC were employed for gene validation of four novel genes with amplification. The putative lung cancer-related genes identified in our array-CGH database would help in further studies on the mechanism of lung tumorigenesis.

Using the UCSC Genome browser, we have annotated a total of 476 genes for Asian patients and 459 genes for Caucasian patients showing high frequency of chromosomal imbalance to be our candidate gene database (Table 1). To improve the validity for identifying potential cancer-related gene, the genes residing within the chromosomal imbalance regions were integrated with gene expression databases. Among the genes selected, the ion transport and chromatin remodeling are the two main common biological processes altered in both Asia and Caucasian populations, suggesting that the candidate genes involved in these processes may play important roles in lung tumorigenesis. For example, irregular choline transport and metabolism have been involved in growth arrest and apoptosis in human lung ADC [21]. Dysfunction of histone methylation was also shown to contribute to human carcinogenesis [22]. Interestingly, genes involved in Wnt receptor signaling pathway were unique in Asian lung cancer, whereas Caucasian lung cancer was addictive to cell surface receptor linked signal transduction and G-protein coupled receptor protein signaling pathways (Figure1A-B). Genes involved in these pathways warrant further studies.

This study found several candidate genes which are located in the common chromosome alteration regions in both Asian and Caucasian, such as PAFAH1B1 gene on 17p13.3. In array-CGH data, chromosome region 17p13.3 harboring PAFAH1B1 showed gain of copy number in 60% of Asian and 70% of Caucasian patients (Table 1). In addition, PAFAH1B1 DNA copy number and mRNA expression level in tumor tissues was significantly higher than that of the matched-normal tissues in both Asian and Caucasian lung cancer patients (Figure3). The frequency of PAFAH1B1 protein overexpression was 68% in Asian and 70% in Caucasian (Figure4). Interestingly, chromosome region 17p13.3 harboring PAFAH1B1 gene also demonstrated association with advanced tumor stage in array-CGH data (Table 3A). Overexpression of PAFAH1B1 mRNA was found in 59.1% of early-stage and 78.3% of late-stage lung cancer patients of Asian descent ( Additional file 6: Table S5), suggesting that the alteration of PAFAH1B1 gene may be involved in both tumor initiation and progression. More patients will be analyzed to clarify its tumorigenic role.

We also found several candidate genes showing different alteration frequency between Asian and Caucasian patients, such as ARHGAP19 and FRAT2. These two closely located (within a 39.8 kb region at 10q24.1) candidate genes, showed alteration frequency of 57.5% with gain of gene copy number in Asian (Table 1A), whereas the alteration frequency was only 20% in Caucasian patients. In addition, both qPCR and RT-qPCR confirmed significant gene amplification and mRNA overexpression in tumors from Asian but not from Caucasian lung cancer patients (Figure3A-B). Interestingly, in relation to clinical pathological parameters of Asian lung cancer patients, mRNA overexpression of ARHGAP19 and FRAT2 was significantly associated with lung SCC ( Additional file 6: Table S5). ARHGAP19 encodes a Rho GTPase-activating protein that stimulates the intrinsic GTP hydrolysis activity of Rho family proteins [23]. ARHGAP family genes are considered to be cancer-associated genes because their genetic alterations lead to carcinogenesis through the dysregulation of Rho/Rac/Cdc42-like GTPases [24]. The up-regulation of the FRAT2 gene has been reported in human gastric cancer [25] and has been implicated in carcinogenesis through activation of the Wnt signaling pathway [26]. Our previous study showed that more than 50% of Taiwanese lung cancer patients had alterations in proteins involved in Wnt/β-catenin signaling [27]. It is important to investigate the role of ARHGAP19 and FRAT2 alterations in SCC lung tumorigenesis and their correlations with Rho and Wnt signaling pathways.

Interestingly, ZNF322A on 6p22.1 showed significant gene amplification and mRNA overexpression in Caucasian, and borderline significant in Asian lung cancer patients (Figure3). The DNA copy number validation for ZNF322A gene was lower than the array CGH result because the BAC may harbor some other genes in addition to ZNF322A. ZNF322A has been reported to be a novel human C2H2 Krüppel-like zinc-finger protein, which regulates transcriptional activation in the MAPK signaling pathways. The Krüppel zinc-finger protein is involved in the regulation of normal cell growth, differentiation, embryogenesis, and tumorigenesis [28]. Identification of the role of ZNF322A in lung tumorigenesis is worthy of further study.

Note that no common tumor subtype-specific regions for ADC and SCC were found in Asian and Caucasian lung cancer patients when the genomic alteration profiles were compared (Table 2A). Even in the same racial group, extreme difference of chromosome alterations could occur between ADC and SCC. These data implicated that distinct etiological mechanism in addition to tobacco and environmental exposures are involved in lung cancer of Asian from Caucasian, supporting the epidemiologic observation that lung cancer in Asian shows relatively high percentage of non-smoking female ADC. Notably, Broët et al. [29] identified molecular differences between NSCLCs from East-Asian and Western European patients by using single nucleotide polymorphism microarray platform, and revealed several copy number aberrations significantly associated with ethnicity (gain on 1p36, 16p13, 16p12, and 16p11 in East-Asian; loss on 19p13 in Western European patients), EGFR mutation (gain on 1p36, 1p35, 7p22-21, 7p15-12, 14q31-32, 16p13, and 16p12 in EGFR mutant tumors; loss on 21q21-22 in EGFR wild-type tumors), but not to K-ras or p53 mutations. Correlation of the mutational spectrum of EGFR K-ras p53, and ALK genes to our array-CGH data warrants further investigations.

Conclusions

In conclusion, our specialized array-CGH results provide an invaluable database of specific chromosome regions about the gene copy number alterations common or differential in Caucasian and Asian lung cancer patients. Functional studies using cell, animal, and clinical models will be conducted to identify their cellular functions and etiological roles in lung tumorigenesis.

Abbreviations

array-CGH:

Array-comparative genomic hybridization

qPCR:

Quantitative polymerase chain reaction

ADC:

Adenocarcinoma

BAC:

Bacterial artificial chromosomes

NSCLC:

Non-small cell lung cancer

RT-qPCR:

Reverse transcriptase-qPCR

LOH:

Loss of heterozygosity

DAVID:

Database for Annotation, Visualization and Integrated Discovery.

References

  1. Govindan R, Page N, Morgensztern D, Read W, Tierney R, Vlahiotis A, Spitznagel EL, Piccirillo J: Changing epidemiology of small-cell lung cancer in the United States over the last 30 years: analysis of the surveillance, epidemiologic, and end results database. J Clin Oncol. 2006, 24: 4539-4544. 10.1200/JCO.2005.04.4859.

    Article  PubMed  Google Scholar 

  2. Parkin DM, Bray FI, Devesa SS: Cancer burden in the year 2000. The global picture. Eur J Cancer. 2001, 37 (Suppl 8): S4-S66.

    Article  PubMed  Google Scholar 

  3. Lee LN, Shew JY, Sheu JC, Lee YC, Lee WC, Fang MT, Chang HF, Yu CJ, Yang PC, Luh KT: Exon 8 mutation of p53 gene associated with nodal metastasis in non-small-cell lung cancer. Am J Respir Crit Care Med. 1994, 150: 1667-1671.

    Article  CAS  PubMed  Google Scholar 

  4. Phan VH, Moore MM, McLachlan AJ, Piquette-Miller M, Xu H, Clarke SJ: Ethnic differences in drug metabolism and toxicity from chemotherapy. Expert Opin Drug Metab Toxicol. 2009, 5: 243-257. 10.1517/17425250902800153.

    Article  CAS  PubMed  Google Scholar 

  5. Davies JJ, Wilson IM, Lam WL: Array CGH technologies and their applications to cancer genomes. Chromosome Res. 2005, 13: 237-248. 10.1007/s10577-005-2168-x.

    Article  CAS  PubMed  Google Scholar 

  6. Kallioniemi A: CGH microarrays and cancer. Curr Opin Biotechnol. 2008, 19: 36-40. 10.1016/j.copbio.2007.11.004.

    Article  CAS  PubMed  Google Scholar 

  7. Hou Q, Wu YH, Grabsch H, Zhu Y, Leong SH, Ganesan K, Cross D, Tan LK, Tao J, Gopalakrishnan V, Tang BL, Kon OL, Tan P: Integrative genomics identifies RAB23 as an invasion mediator gene in diffuse-type gastric cancer. Cancer Res. 2008, 68: 4623-4630. 10.1158/0008-5472.CAN-07-5870.

    Article  CAS  PubMed  Google Scholar 

  8. Kim YH, Kwei KA, Girard L, Salari K, Kao J, Pacyna-Gengelbach M, Wang P, Hernandez-Boussard T, Gazdar AF, Petersen I, Minna JD, Pollack JR: Genomic and functional analysis identifies CRKL as an oncogene amplified in lung cancer. Oncogene. 2010, 29: 1421-1430. 10.1038/onc.2009.437.

    Article  CAS  PubMed  Google Scholar 

  9. Lockwood WW, Chari R, Chi B, Lam WL: Recent advances in array comparative genomic hybridization technologies and their applications in human genetics. Eur J Hum Genet. 2006, 14: 139-148. 10.1038/sj.ejhg.5201531.

    Article  CAS  PubMed  Google Scholar 

  10. Albertson DG: Gene amplification in cancer. Trends Genet. 2006, 22 (8): 447-455. 10.1016/j.tig.2006.06.007.

    Article  CAS  PubMed  Google Scholar 

  11. Mantripragada KK, Buckley PG, de Stahl TD, Dumanski JP: Genomic microarrays in the spotlight. Trends Genet. 2004, 20: 87-94. 10.1016/j.tig.2003.12.008.

    Article  CAS  PubMed  Google Scholar 

  12. Albertson DG, Pinkel D: Genomic microarrays in human genetic disease and cancer. Hum Mol Genet. 2003, 12 (Spec No 2): R145-R152.

    Article  CAS  PubMed  Google Scholar 

  13. Tseng RC, Chang JW, Hsien FJ, Chang YH, Hsiao CF, Chen JT, Chen CY, Jou YS, Wang YC: Genomewide loss of heterozygosity and its clinical associations in non small cell lung cancer. Int J Cancer. 2005, 117: 241-247. 10.1002/ijc.21178.

    Article  CAS  PubMed  Google Scholar 

  14. Sharma RK, Rogojina AT, Chalam KV: Multiplex immunoassay analysis of biomarkers in clinically accessible quantities of human aqueous humor. Mol Vis. 2009, 15: 60-69.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Kresse SH, Skarn M, Ohnstad HO, Namlos HM, Bjerkehagen B, Myklebost O, Meza-Zepeda LA: DNA copy number changes in high-grade malignant peripheral nerve sheath tumors by array CGH. Mol Cancer. 2008, 7: 48-

    Article  PubMed  PubMed Central  Google Scholar 

  16. Chang JW, Liu HP, Hsieh MH, Fang YF, Hsieh MS, Hsieh JJ, Chiu YT, Tsai HY, Chen YH, Chen YT, Hsu HY, Chen YT, Tsai SF, Chen YR, Hsi BL, Huang SF: Increased epidermal growth factor receptor (EGFR) gene copy number is strongly associated with EGFR mutations and adenocarcinoma in non-small cell lung cancers: a chromogenic in situ hybridization study of 182 patients. Lung Cancer. 2008, 61: 328-339. 10.1016/j.lungcan.2008.01.009.

    Article  PubMed  Google Scholar 

  17. Wen CC, Wu YJ, Huang YH, Chen WC, Liu SC, Jiang SS, Juang JL, Lin CY, Fang WT, Hsiung CA, Chang IS: A Bayes regression approach to array-CGH data. Stat Appl Genet Mol Biol. 2006, 5: Article3-

    PubMed  Google Scholar 

  18. Kang JU, Koo SH, Kwon KC, Park JW, Kim JM: Gain at chromosomal region 5p15.33, containing TERT, is the most frequent genetic event in early stages of non-small cell lung cancer. Cancer Genet Cytogenet. 2008, 182: 1-11. 10.1016/j.cancergencyto.2007.12.004.

    Article  CAS  PubMed  Google Scholar 

  19. Uchida M, Tsukamoto Y, Uchida T, Ishikawa Y, Nagai T, Hijiya N, Nguyen LT, Nakada C, Kuroda A, Okimoto T, Kodama M, Murakami K, Noguchi T, Matsuura K, Tanigawa M, Seto M, Ito H, Fujioka T, Takeuchi I, Moriyama M: Genomic profiling of gastric carcinoma in situ and adenomas by array-based comparative genomic hybridization. J Pathol. 2010, 221: 96-105. 10.1002/path.2686.

    Article  CAS  PubMed  Google Scholar 

  20. Dennis G, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA: DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 2003, 4: P3-10.1186/gb-2003-4-5-p3.

    Article  PubMed  Google Scholar 

  21. Wang T, Li J, Chen F, Zhao Y, He X, Wan D, Gu J: Choline transporters in human lung adenocarcinoma: expression and functional implications. Acta Biochim Biophys Sin. 2007, 39: 668-674. 10.1111/j.1745-7270.2007.00323.x.

    Article  CAS  PubMed  Google Scholar 

  22. Hayami S, Kelly JD, Cho HS, Yoshimatsu M, Unoki M, Tsunoda T, Field HI, Neal DE, Yamaue H, Ponder BA, Nakamura Y, Hamamoto R: Overexpression of LSD1 contributes to human carcinogenesis through chromatin regulation in various cancers. Int J Cancer. 2010, 128: 574-586.

    Article  Google Scholar 

  23. Lv L, Xu J, Zhao S, Chen C, Zhao X, Gu S, Ji C, Xie Y, Mao Y: Sequence analysis of a human RhoGAP domain-containing gene and characterization of its expression in human multiple tissues. DNA Seq. 2007, 18: 184-189.

    Article  CAS  PubMed  Google Scholar 

  24. Katoh Y, Katoh M: Identification and characterization of ARHGAP27 gene in silico. Int J Mol Med. 2004, 14: 943-947.

    CAS  PubMed  Google Scholar 

  25. Saitoh T, Katoh M: FRAT1 and FRAT2, clustered in human chromosome 10q24.1 region, are up-regulated in gastric cancer. Int J Oncol. 2001, 19: 311-315.

    CAS  PubMed  Google Scholar 

  26. Saitoh T, Moriwaki J, Koike J, Takagi A, Miwa T, Shiokawa K, Katoh M: Molecular cloning and characterization of FRAT2, encoding a positive regulator of the WNT signaling pathway. Biochem Biophys Res Commun. 2001, 281: 815-820. 10.1006/bbrc.2001.4421.

    Article  CAS  PubMed  Google Scholar 

  27. Tseng RC, Lin RK, Wen CK, Tseng C, Hsu HS, Hsu WH, Wang YC: Epigenetic silencing of AXIN2/betaTrCP and deregulation of p53-mediated control lead to wild-type beta-catenin nuclear accumulation in lung tumorigenesis. Oncogene. 2008, 27: 4488-4496. 10.1038/onc.2008.83.

    Article  CAS  PubMed  Google Scholar 

  28. Li Y, Wang Y, Zhang C, Yuan W, Wang J, Zhu C, Chen L, Huang W, Zeng W, Wu X, Liu M: ZNF322, a novel human C2H2 Kruppel-like zinc-finger protein, regulates transcriptional activation in MAPK signaling pathways. Biochem Biophys Res Commun. 2004, 325: 1383-1392. 10.1016/j.bbrc.2004.10.183.

    Article  CAS  PubMed  Google Scholar 

  29. Broet P, Dalmasso C, Tan EH, Alifano M, Zhang S, Wu J, Lee MH, Regnard JF, Lim D, Koong HN, Agasthian T, Miller LD, Lim E, Camilleri-Broet S, Tan P: Genomic profiles specific to patient ethnicity in lung adenocarcinoma. Clin Cancer Res. 2011, 17: 3542-3550. 10.1158/1078-0432.CCR-10-2185.

    Article  CAS  PubMed  Google Scholar 

Pre-publication history

Download references

Acknowledgments

This work was supported in part by grant NSC99-2628-B-006-004-MY3 from the National Science Council and landmark project A0012 from the National Cheng Kung University (YCW), by grant NSC100-2319-B-004-001 (ISC) and also supported by NCI 5R01CA125541-04 and 5R01CA129501-03 (RS). Study sponsors had no involvement on study design, analysis and interpretation of data, and decision on the submitted manuscript. We thank the Bioinformatics Center of National Cheng-Kung University for providing the MetaCore™ analysis software. We also thank Dr. Jing-Jou Yan (Pathology Department, National Cheng-Kung University) for IHC data interpretation.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yi-Ching Wang.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

YCW and FYL designed this study. FYL and JWC performed research. ISC, FUT, and SSJ analyzed array data. HSH, RK, SN, and RS provided clinical samples. YCW ,YJC, and SFH provided experimental materials. YCW and FYL wrote the manuscript. All authors read and approved the final manuscript.

Electronic supplementary material

12885_2011_3311_MOESM1_ESM.pdf

Additional file 1: Table S1.Overall list of lung cancer patient in array-CGH, and candidate gene validations. (PDF 46 KB)

Additional file 2: Table S2.The selected 18 chromosome regions investigated in the current array-CGH study. (PDF 9 KB)

12885_2011_3311_MOESM3_ESM.pdf

Additional file 3: Figure S1.Array image and schematic illustrations of the Bayes regression approach for identifying the regions with chromosomal copy number changes. (PDF 552 KB)

Additional file 4: Table S3.Primers and PCR conditions used in the current study. (PDF 39 KB)

Additional file 5: Table S4.List of candidate genes in Asian and Caucasian lung cancer patients. (PDF 121 KB)

12885_2011_3311_MOESM6_ESM.pdf

Additional file 6: Table S5.Association between mRNA expression level and lung cancer clinicopathological parameters of four candidate genes in Asian. (PDF 47 KB)

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Lo, FY., Chang, JW., Chang, IS. et al. The database of chromosome imbalance regions and genes resided in lung cancer from Asian and Caucasian identified by array-comparative genomic hybridization. BMC Cancer 12, 235 (2012). https://doi.org/10.1186/1471-2407-12-235

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2407-12-235

Keywords