Oncotarget

Research Papers:

Integrated analysis of chromosome copy number variation and gene expression in cervical carcinoma

PDF |  HTML  |  How to cite

Oncotarget. 2017; 8:108912-108922. https://doi.org/10.18632/oncotarget.22403

Metrics: PDF 1380 views  |   HTML 2292 views  |   ?  

Deng Yan, Song Yi, Wang Chi Chiu, Liu Gui Qin, Wong Hoi Kin, Chung Tony Kwok Hung, Han Linxiao, Choy Kwong Wai, Sui Yi, Yang Tao and Tang Tao _

Abstract

Deng Yan1,2,*, Song Yi1,*, Wang Chi Chiu1,2, Liu Gui Qin3, Wong Hoi Kin1, Chung Tony Kwok Hung1, Han Linxiao4, Choy Kwong Wai1,2, Sui Yi5, Yang Tao6 and Tang Tao1,2

1Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Hong Kong, China

2CUHK Shenzhen Research Institute, Shenzhen, China

3Shenzhen Laboratory of Ophthalmology, Shenzhen Eye Hospital, Affiliated Shenzhen Eye Hospital of Shenzhen University, Shenzhen, China

4Dongguan Third People’s Hospital, Dongguan, China

5Department of Nutrition, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China

6Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China

*These authors have contributed equally to this work

Correspondence to:

Tang Tao, email: [email protected]

Yang Tao, email: [email protected]

Sui Yi, email: [email protected]

Keywords: cervical cancer; chromosome copy number variation; gene expression; cluster analysis; cell cycle pathways

Received: April 19, 2017    Accepted: September 21, 2017    Published: November 11, 2017

ABSTRACT

Objective: This study was conducted to explore chromosomal copy number variations (CNV) and transcript expression and to examine pathways in cervical pathogenesis using genome-wide high resolution microarrays.

Methods: Genome-wide chromosomal CNVs were investigated in 6 cervical cancer cell lines by Human Genome CGH Microarray Kit (4x44K). Gene expression profiles in cervical cancer cell lines, primary cervical carcinoma and normal cervical epithelium tissues were also studied using the Whole Human Genome Microarray Kit (4x44K).

Results: Fifty common chromosomal CNVs were identified in the cervical cancer cell lines. Correlation analysis revealed that gene up-regulation or down-regulation is significantly correlated with genomic amplification (P=0.009) or deletion (P=0.006) events. Expression profiles were identified through cluster analysis. Gene annotation analysis pinpointed cell cycle pathways was significantly (P=1.15E-08) affected in cervical cancer. Common CNVs were associated with cervical cancer.

Conclusion: Chromosomal CNVs may contribute to their transcript expression in cervical cancer.


INTRODUCTION

Although the most important etiological agent in cervical cancer is human papillomavirus (HPV) infection, only a small proportion of infected women developed cervical cancer. HPV infection alone is insufficient to induce malignant changes. Copy number variation (CNV) is a very common phenomenon in cervical cancer and may be important in its pathogenesis [1]. Comparative genomic hybridization (CGH) studies for cervical cancer progression have shown that chromosome 3q gain was associated with the transition from pre-invasive to invasive cervical carcinoma. Subsequently, array-based CGH (aCGH), where arrays of genomic sequences replaced metaphase chromosomes as hybridization targets, was established. More detailed and precise genomic variations had been found in cervical cancer by using aGCH [2]. Lando et al. reported several potential driver genes for cervical carcinogenesis aCGH [3]. However, a recent study reported a limited correlation between chromosomal CNV and gene expression by single nucleotide polymorphisms array platform [4].

In this study, a high resolution Human Genome CGH Microarray Kit was used to detect genome-wide chromosomal CNV in 6 cervical cancer cell lines. We also performed gene expression studies in the cervical cancer cell lines, cervical carcinoma tissues and normal cervical epithelia by using Whole Human Genome Microarray Kit. Using appropriate bioinformatics software, we identified several chromosomal CNV regions and aberrantly expressed genes in cervical cancer. Statistical analysis and gene annotation analysis were also performed for the array data.

RESULTS

Fifty common chromosomal CNVs were identified in cervical cancer cell lines

Using aCGH at a genome-wide resolution of 250 kb, a total of 50 common chromosomal CNV regions were identified, ranging from 0.5 Mb to 80 Mb. Of these, 21 common amplification regions (including 13 significant amplification regions) and 29 deletion regions (including 2 significant deletion regions) were identified (Table 1).

Table 1: Physical location of the chromosome CNV regions identified by array CGH in cervical cancer cell lines

Region

Region length

Cytoband location

Event

Genes

Frequency %

P-value

Reference

chr1:10001-3752828

3,742,828

p36.33 - p36.32

Gain

70

83.33333333

0.003

Gopeshwar Narayan, et al., 2007; Connie P. Matthews, et al., 2000; Y.W. CHOI, et al., 2007

chr2:75161025-85137285

9,976,261

p12 - p11.2

Loss

17

83.33333333

0.006

Y.W. CHOI*, et al., 2007

chr2:137,395,646-170,227,851

32,832,206

q21.3 - q31.1

Loss

84

100

>0.05

F.Y. Huang et al.2005; Y.W. CHOI, et al., 2007

chr2:178,374,598-197,908,813

19,534,216

q31.2 - q33.1

Loss

67

83.33333333

>0.05

F.Y. Huang et al.2005; Y.W. CHOI, et al., 2007

chr2:209,391,516-216,053,786

6,662,271

q34 - q35

Loss

15

83.33333333

>0.05

Lockwood WW, et al., 2007; F.Y. Huang et al.2005; G Ng, et al., 2007

chr3:60,001-8,582,632

8,522,632

p26.3 - p25.3

Loss

15

83.33333333

>0.05

F.Y. Huang et al.2005; Connie P. Matthews, et al., 2000; Y.W. CHOI, et al., 2007

chr3:16,297,340-37,460,259

21,162,920

p25.1 - p22.2

Loss

60

83.33333333

>0.05

Lockwood WW, et al., 2007; F.Y. Huang et al.2005; Connie P. Matthews, et al., 2000

chr3:58,690,297-90,181,487

31,491,191

p14.2 - p11.1

Loss

52

83.33333333

0.028

Lockwood WW, et al., 2007; F.Y. Huang et al.2005; Connie P. Matthews, et al., 2000; Y.W. CHOI, et al., 2007; G Ng, et al., 2007

chr3:93,605,515-101,219,924

7,614,410

q11.2 - q12.3

Loss

38

83.33333333

>0.05

N/A

chr4:10,479,679-39,591,168

29,111,490

p16.1 - p14

Loss

62

83.33333333

>0.05

Lockwood WW, et al., 2007; F.Y. Huang et al.2005; Y.W. CHOI, et al., 2007; G Ng, et al., 2007

chr4:41,596,003-44,365,208

2,769,206

p13

Loss

10

83.33333333

>0.05

Lockwood WW, et al., 2007; F.Y. Huang et al.2005; G Ng, et al., 2007

chr4:58,367,789-139,871,504

81,503,716

q12 - q31.1

Loss

300

83.33333333

>0.05

Lockwood WW, et al., 2007; G Ng, et al., 2007

chr4:141,329,535-182,773,601

41,444,067

q31.1 - q34.3

Loss

127

83.33333333

>0.05

Lockwood WW, et al., 2007; Y.W. CHOI, et al., 2007; G Ng, et al., 2007

chr5:49,690,172-58,526,175

8,836,004

q11.1 - q11.2

Loss

35

83.33333333

>0.05

Connie P. Matthews, et al., 2000

chr6:44,372,753-58,614,061

14,241,309

p21.1 - p11.2

Loss

69

83.33333333

>0.05

Connie P. Matthews, et al., 2000; Y.W. CHOI, et al., 2007

chr6:61,982,931-73,725,450

11,742,520

q11.1 - q13

Loss

16

83.33333333

>0.05

Connie P. Matthews, et al., 2000

chr6:75,067,155-105,246,238

30,179,084

q13 - q21

Loss

83

83.33333333

>0.05

Connie P. Matthews, et al., 2000

chr6:112,398,800-148,256,165

35,857,366

q21 - q24.3

Loss

143

83.33333333

>0.05

Connie P. Matthews, et al., 2000; Y.W. CHOI, et al., 2007

chr7:76,075,269-97,170,202

21,094,934

q11.23 - q21.3

Loss

77

83.33333333

>0.05

N/A

chr7:105,159,121-110,522,522

5,363,402

q22.3 - q31.1

Loss

24

83.33333333

>0.05

N/A

chr8:12,830,587-20,081,624

7,251,038

p22 - p21.3

Loss

27

83.33333333

>0.05

Lockwood WW, et al., 2007; G Ng, et al., 2007

chr8:75,336,800-85,510,468

10,173,669

q21.11 - q21.2

Loss

27

83.33333333

>0.05

N/A

chr8:142,141,881-146,304,022

4,162,142

q24.3

Gain

92

83.33333333

0

Gopeshwar Narayan, et al., 2007; F.Y. Huang et al.2005; Connie P. Matthews, et al., 2000; Y.W. CHOI, et al., 2007; G Ng, et al., 2007

chr9:128,223,213-139,309,447

11,086,235

q33.3 - q34.3

Gain

211

83.33333333

0.017

Lockwood WW, et al., 2007; Y.W. CHOI, et al., 2007

chr11:60,001-3,696,670

3,636,670

p15.5 - p15.4

Gain

85

83.33333333

0

Y.W. CHOI, et al., 2007; G Ng, et al., 2007

chr11:20,658,997-31,795,373

11,136,377

p15.1 - p13

Loss

27

83.33333333

>0.05

F.Y. Huang et al.2005

chr11:65,627,563-67,839,841

2,212,279

q13.1 - q13.2

Gain

77

83.33333333

0.003

Y.W. CHOI, et al., 2007

chr12:38,766,104-42,827,028

4,060,925

q12

Loss

12

83.33333333

>0.05

N/A

chr13:45,697,630-55,761,181

10,063,552

q14.12 - q21.1

Loss

56

83.33333333

>0.05

Lockwood WW, et al., 2007; G Ng, et al., 2007

chr14:23,251,874-24,898,017

1,646,144

q11.2- q12

Gain

64

83.33333333

0.001

N/A

chr16:60,001-3,153,334

3,093,334

p13.3

Gain

137

83.33333333

0.048

Y.W. CHOI, et al., 2007

chr16:3,970,244-5,071,063

1,100,820

p13.3

Gain

23

83.33333333

0.048

Y.W. CHOI, et al., 2007

chr16:28,276,920-31,195,342

2,918,423

p11.2

Gain

100

83.33333333

0.048

Y.W. CHOI, et al., 2007

chr16:66,279,668-70,782,910

4,503,243

q21 - q22.1

Gain

103

83.33333333

0.021

Lockwood WW, et al., 2007; Connie P. Matthews, et al., 2000

chr16:83,959,097-90,294,753

6,335,657

q23.3 - q24.3

Gain

78

83.33333333

0.021

Lockwood WW, et al., 2007; Connie P. Matthews, et al., 2000; Y.W. CHOI, et al., 2007

chr17:7,175,150-8,229,647

1,054,498

p13.1

Gain

59

83.33333333

0.009

N/A

chr17:72,693,870-81,060,000

8,366,131

q25.1 - q25.3

Gain

166

83.33333333

>0.05

Lockwood WW, et al., 2007; Y.W. CHOI, et al., 2007

chr18:18,510,899-43,242,321

24,731,423

q11.1 - q12.3

Loss

70

83.33333333

>0.05

Lockwood WW, et al., 2007; Y.W. CHOI, et al., 2007

chr18:62,450,433-71,009,737

8,559,305

q22.1 - q22.3

Loss

11

83.33333333

>0.05

Lockwood WW, et al., 2007; G Ng, et al., 2007

chr19:1,271,138-4,752,741

3,481,604

p13.3

Gain

105

83.33333333

>0.05

Y.W. CHOI, et al., 2007

chr19:12,747,550-14,740,086

1,992,537

p13.13 - p13.12

Gain

56

83.33333333

>0.05

Gopeshwar Narayan, et al., 2007

chr19:16,170,761-19,780,245

3,609,485

p13.12 - p13.11

Gain

103

83.33333333

>0.05

N/A

chr19:45,216,651-51,316,691

6,100,041

q13.32 - q13.33

Gain

201

83.33333333

>0.05

Lockwood WW, et al., 2007

chr19:55,542,540-56,189,743

647,204

q13.42

Gain

31

83.33333333

>0.05

N/A

chr19:58,530,030-59,114,839

584,810

q13.43

Gain

23

83.33333333

>0.05

N/A

chr20:60,195,293-62,965,520

2,770,228

q13.33

Gain

63

83.33333333

>0.05

Connie P. Matthews, et al., 2000; Y.W. CHOI, et al., 2007; G Ng, et al., 2007

chr21:14,417,523-32,339,619

17,922,097

q11.2 - q22.11

Loss

61

83.33333333

>0.05

N/A

chrX:77,966,491-93,063,726

15,097,236

q21.1 - q21.32

Loss

25

83.33333333

>0.05

Connie P. Matthews, et al., 2000

chrX:120,138,580-127,769,411

7,630,832

q24 - q25

Loss

10

83.33333333

>0.05

Connie P. Matthews, et al., 2000

chrX:152,449,419-153,711,912

1,262,494

q28

Gain

47

83.33333333

0

Gopeshwar Narayan, et al., 2007; Connie P. Matthews, et al., 2000

Eleven of these (5 amplification regions and 6 deletion regions) have not been previously described in cervical cancer (Table 1). A total of 3514 genes were identified, and many tumor related genes, such as ABL1, BCL3, CDH1, CDKN1C, EPHA3, ERBB4, FOSL1, JUNB, MLH1, MYB, p53, RB1, ROS1, SKI, TGFBR1 and THRB, were located in these 50 chromosomal CNV regions.

Chromosomal CNVs could contribute to their transcript expression in cervical cancer

To evaluate if there is any association between chromosomal CNVs and gene expression changes in cervical cancer, we analyzed the gene expression profiles of the cervical cancer cell lines and normal cervical epithelium samples. 17.52% of transcripts (7,211 out of 41,152) exhibited a 2-fold over-expression and 9.02% of transcripts (3,712 out of 41,152) displayed a 2-fold down-regulation in 6 cervical cancer cell lines compared with 3 normal cervical epithelium tissues. Within the 21 common genomic amplification regions, 27.94% of the transcripts (772 out of 2,794) showed 2-fold over-expression. In the 13 significant amplification regions, the percentage was 29.56% (459 out of 1,553). In the 29 deletion regions, 10.46% (287 out of 2744) revealed 2-fold down-regulation. In the 2 significant deletion regions, the percentage was 11.67% (7 out of 60) (Figure 1A and 1B). Statistical analysis showed that gene up-regulation or down-regulation was significantly correlated with genomic amplification (P < 0.01, Spearsman correlation test) or deletion (P < 0.01) events. Thus, chromosomal CNVs can contribute to their transcript expression in cervical cancer. Two tumor related genes, ABL1 and p53, which were located in the genomic amplification regions, were found to be over-expressed by at least 2-fold in cervical cancer.

Gene expression variation in different genomic regions.

Figure 1: Gene expression variation in different genomic regions. (A) Percentage of up-regulated transcripts in whole genomic regions, common amplification regions and significant amplification regions. (B) Percentage of down-regulated transcripts in whole genomic regions, common deletion regions and significant deletion regions.

Profiles differed between cervical cancer cell lines, primary cervical carcinoma and normal cervical epithelium tissues

Expression profiles of transcripts across 6 different cervical cancer cell lines and 2 cervical carcinoma tissues and 3 normal, age-matched, cervical epithelium samples were analyzed using a hierarchical clustering algorithm (unsupervised K-means clustering). Cervical cancer cell lines, cervical carcinoma and normal cervical epithelium were divided two main groups (Figure 2A): cervical cancer cell lines for one group, and clinical cervical carcinoma and normal cervical epithelium for the other group. Interestingly, when gene tree clustering analysis was used to analyze genes with aberrant expression within the 50 common chromosome CNV regions and 15 significant chromosome CNV regions, similar results were obtained (Figure 2B and 2C)

Gene tree clustering analysis.

Figure 2: Gene tree clustering analysis. (A) Gene tree clustering analysis for the gene expression profiles of the cervical cancer cell lines, cervical carcinomas and normal cervical epithelium tissues; (B) gene tree clustering analysis for the aberrantly expressed genes in 50 common chromosomal CNV regions in cervical carcinoma; (C) gene tree clustering analysis for the aberrantly expressed genes in 15 significant chromosomal CNV regions in cervical carcinoma.

Gene ontology analysis for aberrantly expressed genes

By using a volcano plot in GeneSpring, 9,446 transcripts (27.4%) were found to be changed over two fold in the “Cancer group” (including 6 cervical cancer cell lines and 2 clinical cervical carcinomas) compared with the “Normal group” (including 3 cervical epithelium tissues). Among these genes, 6,001 transcripts were up-regulated by over 2 fold, and 3,445 transcripts were down-regulated by over 2 fold.

Pathway analysis showed that cell cycle pathways, cell communication pathways and DNA polymerase pathways were significantly affected pathways in cervical cancer

To further investigate the biological significance of these aberrantly expressed genes, pathway analysis was performed. The analysis results showed that cycle cycle pathways (P=1.15e-8), cell communication pathways (P=1.15e-8) and DNA polymerase pathways (P=1.15e-8) were significantly were affected in the cervical cancer (Table 2). Pathway analysis for the 446 differentially expressed transcripts in the 15 significant chromosomal CNV regions also showed that the cell cycle pathway involved the highest number of transcripts (including ABL1, DUSP9, E2F4, TP53, PKMYT1 and PPP1CA) (Figure 3), and the P value is 0.00062 (Table 3).

Cell cycle pathway analysis in cervical cancer.

Figure 3: Cell cycle pathway analysis in cervical cancer. (A) Differently expressed genes involved in the cell cycle pathway in cervical carcinoma; (B) differently expressed genes within the significant chromosomal CNV regions involved in the cell cycle pathway in cervical carcinoma. Each rectangle represents one gene. The rectangle covered by gray color indicates that this gene is differently expressed in cervical carcinoma compared with normal cervix.

Table 2: Pathway analysis for the differentially expressed genes in cervical cancer

Pathway

Number of genes with each pathway

Genelist vs pathway random overlap p-value

Cell cycle - Homo sapiens (human)

111

1.15E-08

Proteasome - Homo sapiens (human)

27

2.66E-08

One carbon pool by folate - Homo sapiens (human)

22

5.11E-07

Cell Communication - Homo sapiens (human)

60

5.85E-07

Pyrimidine metabolism - Homo sapiens (human)

52

6.05E-06

Purine metabolism - Homo sapiens (human)

77

2.27E-05

DNA polymerase - Homo sapiens (human)

19

5.80E-05

Arginine and proline metabolism - Homo sapiens (human)

35

0.000103

Riboflavin metabolism - Homo sapiens (human)

14

0.00036

gamma-Hexachlorocyclohexane degradation - Homo sapiens (human)

17

0.000515

Hematopoietic cell lineage - Homo sapiens (human)

45

0.000654

Valine, leucine and isoleucine biosynthesis - Homo sapiens (human)

10

0.000804

Glycosphingolipid biosynthesis - ganglioseries - Homo sapiens (human)

13

0.000894

Pathogenic Escherichia coli infection - EHEC - Homo sapiens (human)

33

0.000941

Pathogenic Escherichia coli infection - EPEC - Homo sapiens (human)

33

0.000941

Selenoamino acid metabolism - Homo sapiens (human)

23

0.00105

Valine, leucine and isoleucine degradation - Homo sapiens (human)

31

0.00139

Urea cycle and metabolism of amino groups - Homo sapiens (human)

17

0.00172

ECM-receptor interaction - Homo sapiens (human)

46

0.00179

Glycan structures - biosynthesis 2 - Homo sapiens (human)

36

0.00261

2,4-Dichlorobenzoate degradation - Homo sapiens (human)

6

0.00264

Butanoate metabolism - Homo sapiens (human)

24

0.00302

Lysine degradation - Homo sapiens (human)

30

0.00449

Aminoacyl-tRNA biosynthesis - Homo sapiens (human)

17

0.0047

Cell adhesion molecules (CAMs) - Homo sapiens (human)

63

0.0062

Folate biosynthesis - Homo sapiens (human)

22

0.00732

Ascorbate and aldarate metabolism - Homo sapiens (human)

10

0.00742

Histidine metabolism - Homo sapiens (human)

22

0.00918

Alkaloid biosynthesis II - Homo sapiens (human)

14

0.0101

Limonene and pinene degradation - Homo sapiens (human)

18

0.0102

Focal adhesion - Homo sapiens (human)

91

0.0108

Methionine metabolism - Homo sapiens (human)

11

0.0112

Oxidative phosphorylation - Homo sapiens (human)

49

0.012

Citrate cycle (TCA cycle) - Homo sapiens (human)

16

0.0153

Glycosaminoglycan degradation - Homo sapiens (human)

11

0.016

Nitrobenzene degradation - Homo sapiens (human)

10

0.0165

Olfactory transduction - Homo sapiens (human)

17

0.0179

Glycolysis Gluconeogenesis - Homo sapiens (human)

31

0.0197

Ethylbenzene degradation - Homo sapiens (human)

11

0.0221

Ubiquitin mediated proteolysis - Homo sapiens (human)

26

0.0236

Glyoxylate and dicarboxylate metabolism - Homo sapiens (human)

9

0.0243

Arachidonic acid metabolism - Homo sapiens (human)

25

0.0306

N-Glycan biosynthesis - Homo sapiens (human)

21

0.033

Chondroitin sulfate biosynthesis - Homo sapiens (human)

7

0.037

Linoleic acid metabolism - Homo sapiens (human)

17

0.0415

Pentose phosphate pathway - Homo sapiens (human)

14

0.0465

Apoptosis - Homo sapiens (human)

38

0.0467

Protein export - Homo sapiens (human)

8

0.0498

Table 3: Pathway analysis for the differentially expressed genes within the 15 significant chromosome CNV regions in cervical cancer

Pathway

Number of common genes with each pathway

Genelist vs pathway random overlap p-value

Cell cycle - Homo sapiens (human)

11

0.000622

Purine metabolism - Homo sapiens (human)

7

0.0126

Axon guidance - Homo sapiens (human)

6

0.0365

Insulin signaling pathway - Homo sapiens (human)

6

0.0386

Selenoamino acid metabolism - Homo sapiens (human)

5

0.000365

Tyrosine metabolism - Homo sapiens (human)

5

0.00382

Glycerophospholipid metabolism - Homo sapiens (human)

5

0.00419

Tryptophan metabolism - Homo sapiens (human)

5

0.0137

Nitrobenzene degradation - Homo sapiens (human)

4

0.000101

Aminophosphonate metabolism - Homo sapiens (human)

4

0.000199

Histidine metabolism - Homo sapiens (human)

4

0.00452

Androgen and estrogen metabolism - Homo sapiens (human)

4

0.00747

Glycan structures - biosynthesis 2 - Homo sapiens (human)

4

0.0281

Sulfur metabolism - Homo sapiens (human)

3

0.000724

Ethylbenzene degradation - Homo sapiens (human)

3

0.00309

RNA polymerase - Homo sapiens (human)

3

0.00781

1- and 2-Methylnaphthalene degradation - Homo sapiens (human)

3

0.0136

Benzoate degradation via CoA ligation - Homo sapiens (human)

3

0.0153

Limonene and pinene degradation - Homo sapiens (human)

3

0.0153

Pyruvate metabolism - Homo sapiens (human)

3

0.0362

Bisphenol A degradation - Homo sapiens (human)

2

0.0233

Glycosphingolipid biosynthesis - ganglioseries - Homo sapiens (human)

2

0.03

Dentatorubropallidoluysian atrophy (DRPLA) - Homo sapiens (human)

2

0.0323

Parkinson's disease - Homo sapiens (human)

2

0.0323

DISCUSSION

In our genome-wide CNV analysis, we identified 50 frequently altered genomic regions (ranging from 0.5 Mb to 80 Mb), of which 11 have not been previously described in cervical cancer (Table 1). These differences in our results may be due to the different platforms of assay, different settings of analysis or the different cervical cancer cells. In these 50 commonly altered genomic regions, 3514 genes are included. Some of these, especially oncogenic or tumor suppressor genes, may be associated with the development of cervical cancer.

The gene tree clustering result suggested that during the development of cervical carcinoma, gene expression significantly changes, and cervical carcinoma can be distinguished from normal cervical epithelium tissue by clustering analysis of the gene expression profile. Since the cervical cancer cell lines were separate from primary cervical carcinoma and normal cervical epithelium tissue, we assumed that extended culturing of cervical cancer cell lines may also significantly alter their gene expression profiles.

The integrated analysis of genome-wide chromosomal copy number changes and gene expression profiling indicated that the identified CNVs could contribute to the expression of some but not all genes (Figure 1). This finding is consistent with a report by Vazquez-Mena et al. which used a different array platform to detect the correlation between CNVs and gene expression variation in cervical cancer cell lines. Other factors, such as epigenetic changes or transcription factors, may also contribute to variation of gene expression in cervical cancer [4]. Pathway analysis indicated that significant changes of some pathways, especially those involving the cell cycle, may be involved in the pathogenesis of cervical cancer.

ABL1 and p53, the two cell cycle pathway tumor-related genes which were located in the significant genomic amplification regions, were found to be overexpressed at least 2-fold in cervical cancer. ABL1, which was located in the chr9:128,223,213-139,309,447 genomic amplification region, plays a role in apoptosis. p53 is a well-known tumor suppressor gene, and an increase in p53 levels plays a critical role in the induction of genes that results in cell cycle arrest [7], allowing repair of damaged DNA or activation of apoptotic pathways [8]. In cervical cancer with high risk of HPV-infection, the E6 protein from high-risk HPV can bind to tumor suppressor protein p53 for rapid degradation via a cellular ubiquitin ligase [9]. Other studies have indicated that p53 protein over-expression is not common or associated with survival in cervical carcinoma [10, 11]. However, from our array CGH and gene expression array data, p53 was located in the chr17:7,175,150-8,229,647 genomic amplification region and was over-expressed at the mRNA level (the median of the expression of p53 in cervical cancer cell lines was 1.316 [from 0.82 to 3.38); the median of the expression of p53 in normal cervical epithelium was 0.295 [from 0.153 to 0.485]). This suggests that gene dosage of p53 contributes to RNA over-expression in some cervical cancers.

Our results demonstrated that over-expression of transforming growth factor-beta 1 (TGF-β1), a gene important in cell cycle pathways, may be due to a chromosomal CNV. TGF-β1 was amplified and was also over-expressed (>2 fold) in the cancer group compared with the normal group. TGF-β1 is involved in many different critical processes, such as embryonic development, cellular maturation and differentiation, wound healing, immune regulation and inflammation. TGF-β1 is a potent inhibitor of cell proliferation at the beginning of carcinogenesis [12, 13]. When cells become resistant to TGF-β1, tumor growth may be enhanced and metastasis promoted via immune evasion and angiogenesis. An increased expression of TGF-β1 has been found in cervical cancer. Kirma et al. suggested that TGF-β1 may be a factor in inducing over-expression of an oncogene, c-fms. Blocking c-fms has been demonstrated to result in increased apoptosis and decreased motility in cervical cancer [14].

Matrix metalloproteinases (MMPs) play an important role in the enhancement of tumor-induced angiogenesis. Our aCGH data showed that 9 MMP genes (MMP1, 3, 7, 8, 10, 12, 13, 20 and 27) located within 11q22 are amplified in Caski and SiHa cell lines, consistent with Lockwood’s findings. Further analysis revealed that MMP14, 23B and 25 were located in significant genomic amplification regions and MMP1, 15, 17 and TIMP1 in all our 6 cervical cancer cell lines. This is consistent with previous work reporting over-expression of MMP12 and MMP15 [15-19].

In summary, we have identified several chromosomal CNV regions and demonstrated that chromosomal CNVs are a common phenomenon which can affect the level of RNA expression in cervical cancer. Pathway analysis for the aberrantly expressed genes suggested that significant changes of some pathways, especially those involving the cell cycle, may contribute to the pathogenesis of cervical cancer. This study provided some clinical significance for us to have a better understanding of cervical cancer pathogenesis.

MATERIALS AND METHODS

Cervical cancer cell lines and specimens

Six human cervical cancer cell lines (HeLa, SiHa, C33A, ME180, CC2 and CC3) were used for aCGH and gene expression array. Five clinical specimens, including three normal cervical tissues and two cervical carcinoma specimens (FIGO stage: IIA), were collected from patients (aged between 36-42 years old) at the Department of Obstetrics and Gynaecology at the Prince of Wales Hospital in Hong Kong from January 2014 to December 2014. Informed consent was obtained from all participating subjects, and Institutional Review Board approval was obtained.

Tissue micro-dissection

Micro-dissection was used as described in our previous study [5]. Briefly, the tissue specimens were frozen in OCT cryomoulds (SAKURA, Japan), sectioned (8 μm) by a cryostat at -20°C (Leica Corp., CRYOCUT 1800), and then mounted onto glass slides (SAIL BRAND, Cat No 7105) at room temperature. Sections were stained by 0.1% methyl green (Sigma) and micro-dissected using a sterile surgical blade (AESCULAP) and collected immediately for further experiments.

Microarray comparative genomic hybridization analysis

Microarray comparative genomic hybridization using Human Genome CGH Microarray Kit (4x44K) (Agilent Technologies, Santa Clara, CA, USA) was used for identifying chromosomal CNV of 6 cervical cancer cell lines and 3 normal cervical samples as per the manufacturer’s protocol. Briefly, genomic DNA of cervical cancer cell lines was extracted using a DNeasy Blood & Tissue Kit (QIAGEN, Cat No. 69506). 1 μg genomic DNA of test sample and 1 μg human sex-matched control DNA as a reference sample (Promega G1521A; Lot no. 20929604) were digested using Alu I and Rsa I. This was followed by fluorescent labeling, clean-up of labeled genomic DNA, microarray hybridization and scanning. The data were extracted using the Agilent Feature Extraction (FE) v11.0 program. After calculating the background signal, non-uniform signal and the average raw signal on each probe, the resulting data files were generated and transferred to the bioinformatics software, Nexus Copy Number version 6.1 (BioDiscovery, Inc., El Segundo, CA, USA) for analysis [6].

Gene expression analysis

The Whole Human Genome Microarray Kit, 4x44K (G4112F) was used to probe gene expression in 6 cervical cancer cell lines, 2 cervical carcinomas and 3 normal cervical epithelium tissue samples. The resulting data files were generated and transferred to GeneSpring GX version 11.5 (Agilent Technologies, Santa Clara, CA, USA) for further analysis.

Reverse transcription PCR (RT-RCR)

RT-RCR was peformed for the identified gain and loss genes both on expression level and on genome level, normalized by GAPDH and B-globin (Forward: 5’-GAAGAGCCAAGGACAGGTAC-3’, Reverse: 5’-CAACTTCATCCACGTTCACC-3’), B2M (Forward:5’-TGCTGTCTCCATGTTTGATGTATCT-3’;Reverse:5’-TCTCTGCTCCCCACCTCTAAGT-3’), respectively.

Statistical analysis

For the Microarray Comparative Genomic Hybridization Analysis, 0.37 was used as the cut off value for amplification or 0.5 for deletion of a single probe. Putative chromosome copy number changes were defined by intervals of three or more adjacent probes with log2 ratios suggestive of a deletion or duplication when compared with the log2 ratios of adjacent probes. The p value for significant difference was set to less than 0.05 to reduce the false discovery rate (FDR). For Gene Expression Analysis, the cut-off value defining an aberrant change of gene expression was set at 2 fold for data analysis. Pathway analysis for the gene expression data was performed by GeneSpring GX version 11.5, and the pathway was downloaded from the KEGG database (ftp://ftp.genome.jp/pub/kegg/).

Abbreviations

CNV Copy number variations

HPV Human papillomavirus

CGH Comparative genomic hybridization

TGF-β1 Transforming growth factor-beta 1

MMPs Matrix metalloproteinases

FDR False discovery rate

Author contributions

T.T., Y.S. and T.Y. designed the research. Y.D. and Y.S. performed the experiments and wrote the manuscript. C.C.W., H.K.W., T.K.H.C., L.X.H. and K.W.C. revised the manuscript. All authors reviewed the manuscript.

CONFLICTS OF INTEREST

None of the authors of this manuscript report conflicts of interest regarding the performance of this study or its publication.

FUNDING

This study was supported by Basic Research for 2015 Shenzhen Municipal Science and Technology Programme (JCY20150630165236963), Shenzhen Science and Technology Innovation Project (JCYJ20140414114853648), the Innovation and Technology Fund of the Hong Kong Special Administrative Region, China (Ref No. UIM/283) and grants from the National Natural Science Foundation of China (grant No. 81660150 and 81372750).

REFERENCES

1. Bodelon C, Vinokurova S, Sampson JN, den Boon JA, Walker JL, Horswill MA,  Korthauer K, Schiffman M, Sherman ME, Zuna RE, Mitchell J, Zhang X, Boland JF, et al. Chromosomal copy number alterations and HPV integration in cervical precancer and invasive cancer. Carcinogenesis. 2016; 37: 188-96.

2. Lockwood WW, Coe BP, Williams AC, MacAulay C, Lam WL. Whole genome tiling path array CGH analysis of segmental copy number alterations in cervical cancer cell lines. Int J Cancer. 2007; 120: 436-43.

3. Lando M, Holden M, Bergersen LC, Svendsrud DH, Stokke T, Sundfor K, Glad IK, Kristensen GB, Lyng H. Gene dosage, expression, and ontology analysis identifies driver genes in the carcinogenesis and chemoradioresistance of cervical cancer. PLoS Genet. 2009; 5: e1000719.

4. Vazquez-Mena O, Medina-Martinez I, Juarez-Torres E, Barron V, Espinosa A, Villegas-Sepulveda N, Gómez-Laguna L, Nieto-Martínez K, Orozco L, Roman-Basaure E, Muñoz Cortez S, Borges Ibañez M, Venegas-Vega C, et al. Amplified genes may be overexpressed, unchanged, or downregulated in cervical cancer cell lines. PLoS One. 2012; 7: e32667.

5. Tang T, Wong HK, Gu W, Yu MY, To KF, Wang CC,  Wong YF, Cheung TH, Chung TK, Choy KW. MicroRNA-182 plays an onco-miRNA role in cervical cancer. Gynecol Oncol. 2013; 129: 199-208.

6. Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A. 2001; 98: 10515.

7. Orth JD, Loewer A, Lahav G, Mitchison TJ. Prolonged mitotic arrest triggers partial activation of apoptosis, resulting in DNA damage and p53 induction. Mol Biol Cell. 2012; 23: 567-76.

8. Wang X, Simpson ER, Brown KA. p53: Protection against tumor growth beyond effects on cell cycle and apoptosis. Cancer Res. 2015; 75: 5001-07.

9. Hiller T, Stubenrauch F, Iftner T. Isolation and functional analysis of five HPVE6 variants with respect to p53 degradation. J Med Virol. 2008; 80: 478-83.

10. Tommasino M, Accardi R, Caldeira S, Dong W, Malanchi I, Smet A, Zehbe I. The role of TP53 in cervical carcinogenesis. Hum Mutat. 2003; 21: 307-12.

11. Koyamatsu Y, Yokoyama M, Nakao Y, Fukuda K, Saito T, Matsukuma K, Iwasaka T.. A comparative analysis of human papillomavirus types 16 and 18 and expression of p53 gene and Ki-67 in cervical, vaginal, and vulvar carcinomas. Gynecol Oncol. 2003; 90: 547-51.

12. Mantel PY, Schmidt-Weber CB. Transforming growth factor-beta: recent advances on its role in immune tolerance. Methods Mol Biol. 2011; 677: 303-38.

13. Kloth JN, Fleuren GJ, Oosting J, de Menezes RX, Eilers PH, Kenter GG, Gorter A. Substantial changes in gene expression of Wnt, MAPK and TNFalpha pathways induced by TGF-beta1 in cervical cancer cell lines. Carcinogenesis. 2005; 26: 1493-502.

14. Kirma N, Hammes LS, Liu YG, Nair HB, Valente PT, Kumar S, Flowers LC, Tekmal RR. Elevated expression of the oncogene c-fms and its ligand, the macrophage colony-stimulating factor-1, in cervical cancer and the role of transforming growth factor-beta1 in inducing c-fms expression. Cancer Res. 2007; 67: 1918-26.

15. Luo CF, Zhu RQ, Wang H, Lu YL. Expression of COX-2 and MMP-9 in cervical carcinoma and their clinical significance. Zhonghua Zhong Liu Za Zhi. 2007; 29: 526-30.

16. Rauvala M, Aglund K, Puistola U, Turpeenniemi-Hujanen T, Horvath G, Willen R, Stendahl U. Matrix metalloproteinases-2 and -9 in cervical cancer: different roles in tumor progression. Int J Gynecol Cancer. 2006; 16: 1297-302.

17. Abraham R, Schafer J, Rothe M, Bange J, Knyazev P, Ullrich A. Identification of MMP-15 as an anti-apoptotic factor in cancer cells. J Biol Chem. 2005; 280: 34123-32.

18. Vazquez-Ortiz G, Pina-Sanchez P, Vazquez K, Duenas A, Taja L, Mendoza P, Garcia JA, Salcedo M. Overexpression of cathepsin F, matrix metalloproteinases 11 and 12 in cervical cancer. BMC Cancer. 2005; 5: 68.

19. Nasr M, Ayyad SB, El-Lamie IK, Mikhail MY. Expression of matrix metalloproteinase-2 in preinvasive and invasive carcinoma of the uterine cervix. Eur J Gynaecol Oncol. 2005; 26: 199-202.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 22403