Microarray technology to investigate genes associated with papillary thyroid carcinoma

  • Authors:
    • Xinyong Zhu
    • Jing Yao
    • Wen Tian
  • View Affiliations

  • Published online on: January 9, 2015     https://doi.org/10.3892/mmr.2015.3180
  • Pages: 3729-3733
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

DNA microarray data on thyroid tissue from patients with papillary thyroid carcinoma (PTC) and from healthy controls were compared in order to investigate the regulatory genes and uncover the underlying regulatory network in PTC. The DNA microarray data set, GSE3678, was downloaded from Gene Expression Omnibus database. This included seven thyroid tissue samples from patients with PTC and seven samples from healthy controls. Raw data were processed and differentially expressed genes (DEGs) were identified using corresponding R packages. Gene regulation analysis was conducted using TRANSFAC® and TRED. A total of 171 DEGs were obtained. A regulatory network was then established, using 104 of the DEGs. Subsequently, pathway enrichment analyses of the genes were conducted using Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool. Three differentially expressed transcription factors were identified: Trefoil factor 3, cut‑like homeobox 2 and forkhead box protein A2. The most significant pathways involving the 104 DEGs were pathways involved in cancer. Biological process analysis using DAVID, suggested that these genes were associated with the positive regulation of gene expression, gene transcription and metabolic processes. The present study identified a range of genes associated with the development of PTC. The results of the present study were beneficial for understanding the regulatory mechanisms involved in PTC, and for developing clinical diagnostic and therapeutic approaches for this disease.

Introduction

Thyroid cancer is the most common type of malignancy in the head and neck. It can be divided into four pathological types: Papillary thyroid carcinoma (PTC), follicular carcinoma, medullary carcinoma and undifferentiated carcinoma. PTC is the most common of these and accounts for ~80% of cases (1). The incidence of PTC in the USA has increased in recent years (2). Nevertheless, the prognosis for PTC is good and overall survival is ~90% (3).

Alterations in the RET/PTC-RAS-BRAF signaling pathway are characteristic of PTC (4,5). Activating BRAF and ret/PTC1 mutations may be an important step in the development of this disease (610). The etiology of PTC remains poorly understood. Therefore the identification of diagnostic and prognostic biomarkers is required. The detection of thyroid nodules is facilitated by high-resolution sonography. However, the detection of the nodules as either benign or malignant using fine-needle aspiration biopsy, real-time sonographic elastography, ultrasound elastography or Micro-Pure imaging, is problematic (11).

Microarray technology may be an effective tool for investigating the underlying regulatory network involved in PTC (1215). In addition, He et al (16) have elucidated the role of microRNAs in the predisposition to, and development of PTC. Despite this, a greater understanding of PTC is required in order to improve clinical diagnosis and treatment, for example by identifying early biomarkers.

In the present study, gene expression profiles of thyroid tissue from patients with PTC were compared with those from healthy controls, in order to identify differentially expressed genes (DEGs). The regulatory associations for the DEGs were retrieved and potential key genes were identified. Pathway enrichment and biological process analyses were conducted to investigate the roles of these genes in the development of PTC.

Materials and methods

Microarray data

Microarray data set, GSE3678, consisting of 14 thyroid tissue samples, was downloaded from Gene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/). This included seven thyroid tissue samples from patients with PTC and seven from healthy people. Raw data were obtained from Affymetrix Human Genome U133 Plus 2.0 Array (Affymetrix, Inc., Santa Clara, CA, USA).

Screening of DEGs

Raw data were processed using software package GEOquery in R and differential expression analysis was conducted using limma (17) in R. The Beyer-Hardwick (BH) method was adopted for multiple corrections (18) in order to identify DEGs between the disease and the control groups. A fold change (FC) of >2 (| logFC |> 2) and P<0.05 were set as the cutoff values.

Establishment of a gene regulatory network

Gene regulation analysis was conducted using TRANSFAC® and TRED (19). A total of 5,558 regulatory associations were identified in TRANSFAC and TRED and extracted to a single database. The 171 DEGs were entered into this database in order to set up a regulatory network, and the corresponding schematic diagram of 104 DEGs was generated using Cytoscape version 2.8 (20).

Pathway enrichment analysis, biological process analysis and gene ontology (GO) annotation

Pathway enrichment analysis and biological process analysis with GO annotation were conducted for genes included in the regulatory network, using the Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool (21) in order to determine their involvement in the development of PTC.

Results

DEGs

Differential expression analysis identified 171 genes as DEGs.

Gene regulatory network

The regulatory associations for the 171 DEGs were identified from the pre-established database. In total, 111 associations between transcription factors (TFs) and target genes among 104 DEGs were determined by the comparison between 5,558 regulatory associations and 171 DEGs, using TRANSFAC and TRED (Fig. 1).

Among the 104 DEGs, three TFs were identified: Trefoil factor 3 (TFF3), cut-like homeobox 2 (CUX2) and forkhead box protein A2 (FOXA2). The target genes of these DEGs are listed in Table I.

Table I

Differentially expressed transcription factors and their target genes.

Table I

Differentially expressed transcription factors and their target genes.

TF geneTarget gene
TFF3IFNB1
CUX2CYBB
FOXA2APOB
FOXA2F2
FOXA2IGFBP1
FOXA2PROC
FOXA2TTF1
FOXA2MSH2

[i] TF, transcription factor; CUX2, cut-like homeobox 2; FOXA2, forkhead box protein A2; IFNB1, interferon β 1; CYBB, cytochrome b-245, β polypeptide; APOB, apolipoprotein B; IGFBP1, insulin-like growth factor binding protein 1; TTF1, thyroid transcription factor 1; MSH2, mutS homolog 2.

Pathway enrichment analysis, biological process analysis and GO annotation

Pathway enrichment analysis was conducted for the 104 DEGs. It identified six pathways (Table II); the most significant of these was in cancer. These results suggest that certain DEGs participate in pathways involved in PTC.

Table II

Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis results for the 104 DEGs.

Table II

Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis results for the 104 DEGs.

Term IDPathway nameCountP-valueFDR
hsa05200Pathways in cancer264.72E-155.04E-12
hsa05221Acute myeloid leukemia101.62E-081.71E-05
hsa05220Chronic myeloid leukemia92.12E-060.002235
hsa05222Small cell lung cancer95.04E-060.005318
hsa05212Pancreatic cancer81.80E-050.019025
hsa05216Thyroid cancer53.34E-040.351572

[i] DEGs, differentially expressed genes; FDR, false discovery rate.

Biological process analysis was also conducted for the 104 DEGs. Certain DEGs were associated with the positive regulation of gene expression, gene transcription and metabolic processes (Table III).

Table III

GO biological process analysis results for the 104 DEGs.

Table III

GO biological process analysis results for the 104 DEGs.

GO TermDescriptionCountP-valueFDR
GO:0010628Positive regulation of gene expression431.43E-312.40E-28
GO:0045944Positive regulation of transcription from RNA polymerase II promoter372.78E-314.67E-28
GO:0045893Positive regulation of transcription, DNA-dependent403.57E-316.00E-28
GO:0051254Positive regulation of RNA metabolic process404.92E-318.27E-28
GO:0045941Positive regulation of transcription427.54E-311.27E-27
GO:0010557Positive regulation of macromolecule biosynthetic process431.77E-292.98E-26
GO:0045935Positive regulation of nucleobase, nucleoside, nucleotide and nucleic acid metabolic process424.21E-297.07E-26
GO:0051173Positive regulation of nitrogen compound metabolic process421.46E-282.46E-25
GO:0009891Positive regulation of biosynthetic process432.06E-283.46E-25
GO:0006357Regulation of transcription from RNA polymerase II promoter431.25E-272.10E-24

[i] DEGs, differentially expressed genes; GO, gene ontology; FDR, false discovery rate.

Discussion

In the present study, microarray data for thyroid tissue samples from PTC patients and from healthy controls were compared. A total of 171 DEGs were identified in these samples. Gene regulation analysis revealed 111 associations among 104 of these DEGs. Three differentially expressed TFs were identified: TFF3, CUX2 and FOXA2. Pathway enrichment and biological process analyses were conducted for the 104 DEGs in order to investigate their involvement in the development of PTC. Pathway enrichment analysis indicated that these genes are associated with colorectal, pancreatic, thyroid and lung cancers, and leukemia (Fig. 2). Biological process analysis suggested that these genes exhibit corresponding functions through positive regulation of gene expression, transcription and metabolic processes.

Underexpression of TFF3 in PTC has been confirmed by previous studies (22,23) and it is proposed as a biomarker at the RNA level (24,25). Its target gene is interferon β 1 (Table I), which exhibits anticancer activity (26). CUX2 (Table I) contains three CUT domains, which are three internal CUT repeats, and a homeodomain, which are DNA-binding motifs. CUX2 is important in the negative regulation of transcription and has been linked to tumor progression (27). FOXA2 is a member of the forkhead box-O family (Table I), which is involved in the regulation of a series of cellular events, such as differentiation, DNA repair, cell cycle arrest and apoptosis (28,29). It is a potential oncogene in anaplastic thyroid carcinoma (30). Kim et al (31) confirmed the decreased expression of FOXA2 in PTC cells using reverse transcription-quantitative polymerase chain reaction. Akagi et al (32) also found a reduced expression of FOXA2 in PTC cells compared with that in healthy thyroid cells. The enforced expression of a range of genes, including FOXA2, inhibits PTC cell growth (32). Furthermore, Akagi et al (32) reported that the CpG island in the promoter region of FOXA2 was aberrantly methylated and that treatment with 5-aza-2-deoxycytidine induced the expression of FOXA2 in PTC cells. Insulin-like growth factor binding protein 1 (IGFBP1) is a target gene of FOXA2 (Table I). Yashiro et al (33) demonstrated that IGFBP1 activity is significantly higher in cells from patients with PTC, therefore it may be involved in PTC cell growth regulation.

Pathway enrichment analysis indicated that the top pathway that the DEGs were found to be associated with was pathways in cancer, involving 26 DEGs. Fibronectin 1 is a glycoprotein present in a dimeric or multimeric form, which is present at the cell surface and in the extracellular matrix. It is involved in cell adhesion and migration, and is therefore a potential drug target with which to treat cancer, by blocking the processes involved in metastasis (34). Its overexpression in PTC at the mRNA level has been demonstrated by Takano et al (35). Cyclin-dependent kinase inhibitor 2B forms a complex with cyclin-dependent kinase 4 or cyclin-dependent kinase 6, and functions as a cell growth regulator (36). Arnaldi et al (37) revealed that it is underexpressed in malignant thyroid tissue. Peroxisome proliferator-activated receptor γ 1 (PPARG1) fuses with paired box 8 and has been confirmed as an oncogene in human thyroid carcinoma (38). In the present study, peroxisome proliferator-activated receptor γ coactivator 1-α, a target gene of PPARG1, was found to be differentially expressed, which was in accordance with the findings of Antico Arciuch et al (39).

Biological process analysis suggested that the DEGs identified in the present study were linked to gene expression, gene transcription, and the positive regulation of RNA and nitrogen compound metabolic processes (Table III). Therefore, a variety of regulatory mechanisms were involved in the development of PTC. Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminal domain 1 (CITED1) and CITED2 were transcriptional coactivators and were identified as DEGs. Prasad et al (14) indicated that CITED1 was upregulated in PTC cells compared with cells from healthy thyroids. Therefore it may serve as a diagnostic marker for PTC.

In conclusion, 171 DEGs in PTC were identified and the molecular mechanisms of 104 DEGs were investigated following the creation of a regulatory network. Three TFs associated with PTC were identified. Pathway enrichment and biological process analyses of the genes in the network confirmed their involvement in the development of PTC. The results of the present study were beneficial for future research into the genes and regulatory mechanisms associated with the pathogenesis of PTC.

Acknowledgements

This study was supported by a grant from Capital Clinical Feature Application Research (Z141107002514102).

References

1 

Kitamura Y, Minobe K, Nakata T, et al: Ret/PTC3 is the most frequent form of gene rearrangement in papillary thyroid carcinomas in Japan. J Hum Genet. 44:96–102. 1999. View Article : Google Scholar : PubMed/NCBI

2 

Jemal A, Tiwari RC, Murray T, et al: Cancer statistics, 2004. CA Cancer J Clin. 54:8–29. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Lang BH, Chow SM, Lo CY, et al: Staging systems for papillary thyroid carcinoma: a study of 2 tertiary referral centers. Ann Surg. 246:114–121. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Kimura ET, Nikiforova MN, Zhu Z, Knauf JA, Nikiforov YE and Fagin JA: High prevalence of BRAF mutations in thyroid cancer: genetic evidence for constitutive activation of the RET/PTC-RAS-BRAF signaling pathway in papillary thyroid carcinoma. Cancer Res. 63:1454–1457. 2003.PubMed/NCBI

5 

Melillo RM, Castellone MD, Guarino V, et al: The RET/PTC-RAS-BRAF linear signaling cascade mediates the motile and mitogenic phenotype of thyroid cancer cells. J Clin Invest. 115:1068–1081. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Cohen Y, Xing M, Mambo E, et al: BRAF mutation in papillary thyroid carcinoma. J Natl Cancer Inst. 95:625–627. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Fusco A, Viglietto G and Santoro M: A new mechanism of BRAF activation in human thyroid papillary carcinomas. J Clin Invest. 115:20–23. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Nikiforova MN, Kimura ET, Gandhi M, et al: BRAF mutations in thyroid tumors are restricted to papillary carcinomas and anaplastic or poorly differentiated carcinomas arising from papillary carcinomas. J Clin Endocrinol Metab. 88:5399–5404. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Jhiang SM, Sagartz JE, Tong Q, et al: Targeted expression of the ret/PTC1 oncogene induces papillary thyroid carcinomas. Endocrinology. 137:375–378. 1996.PubMed/NCBI

10 

Tallini G, Santoro M, Helie M, et al: RET/PTC oncogene activation defines a subset of papillary thyroid carcinomas lacking evidence of progression to poorly differentiated or undifferentiated tumor phenotypes. Clin Cancer Res. 4:287–294. 1998.PubMed/NCBI

11 

Ciledag N, Arda K, Aribas BK, Aktas E and Köse SK: The utility of ultrasound elastography and MicroPure imaging in the differentiation of benign and malignant thyroid nodules. AJR Am J Roentgenol. 198:W244–W249. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Giordano TJ, Kuick R, Thomas DG, et al: Molecular classification of papillary thyroid carcinoma: distinct BRAF, RAS, and RET/PTC mutation-specific gene expression profiles discovered by DNA microarray analysis. Oncogene. 24:6646–6656. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Jarzab B, Wiench M, Fujarewicz K, et al: Gene expression profile of papillary thyroid cancer: sources of variability and diagnostic implications. Cancer Res. 65:1587–1597. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Prasad ML, Pellegata NS, Kloos RT, Barbacioru C, Huang Y and de la Chapelle A: CITED1 protein expression suggests papillary thyroid carcinoma in high throughput tissue microarray-based study. Thyroid. 14:169–175. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Vasko V, Espinosa AV, Scouten W, et al: Gene expression and functional evidence of epithelial-to-mesenchymal transition in papillary thyroid carcinoma invasion. Proc Natl Acad Sci USA. 104:2803–2808. 2007. View Article : Google Scholar : PubMed/NCBI

16 

He H, Jazdzewski K, Li W, et al: The role of microRNA genes in papillary thyroid carcinoma. Proc Natl Acad Sci USA. 102:19075–19080. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Smyth GK: Limma: linear models for microarray data. Bioinformatics and Computational Biology Solutions using R and Bioconductor. Gentleman R, et al: Springer; New York: pp. 397–420. 2005, View Article : Google Scholar

18 

Benjamini Y and Hochberg Y: Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B Stat Methodol. 57:289–300. 1995.

19 

Wingender E, Kel AE, Kel OV, et al: TRANSFAC, TRRD and COMPEL: towards a federated database system on transcriptional regulation. Nucleic Acids Res. 25:265–268. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Smoot ME, Ono K, Ruscheinski J, Wang PL and Ideker T: Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics. 27:431–432. 2011. View Article : Google Scholar :

21 

Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Huang Y, Prasad M, Lemon WJ, et al: Gene expression in papillary thyroid carcinoma reveals highly consistent profiles. Proc Natl Acad Sci USA. 98:15044–15049. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Hawthorn L, Stein L, Varma R, Wiseman S, Loree T and Tan D: TIMP1 and SERPIN-A overexpression and TFF3 and CRABP1 underexpression as biomarkers for papillary thyroid carcinoma. Head Neck. 26:1069–1083. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Finley DJ, Arora N, Zhu B, Gallagher L and Fahey TJ III: Molecular profiling distinguishes papillary carcinoma from benign thyroid nodules. J Clin Endocrinol Metab. 89:3214–3223. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Takano T, Miyauchi A, Yoshida H, Kuma K and Amino N: High-throughput differential screening of mRNAs by serial analysis of gene expression: decreased expression of trefoil factor 3 mRNA in thyroid follicular carcinomas. Br J Cancer. 90:1600–1605. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Qin XQ, Beckham C, Brown JL, et al: Human and mouse IFN-beta gene therapy exhibits different anti-tumor mechanisms in mouse models. Mol Ther. 4:356–364. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Mccarthy N: Tumorigenesis: Cut here for differentiation. Nat Rev Cancer. 12:3202012. View Article : Google Scholar : PubMed/NCBI

28 

Arden KC: Multiple roles of FOXO transcription factors in mammalian cells point to multiple roles in cancer. Exp Gerontol. 41:709–717. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Reagan-Shaw S and Ahmad N: The role of Forkhead-box Class O (FoxO) transcription factors in cancer: a target for the management of cancer. Toxicol Appl Pharmacol. 224:360–368. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Nucera C, Eeckhoute J, Finn S, et al: FOXA1 is a potential oncogene in anaplastic thyroid carcinoma. Clin Cancer Res. 15:3680–3689. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Kim HS, Kim Do H, Kim JY, et al: Microarray analysis of papillary thyroid cancers in Korean. Korean J Intern Med. 25:399–407. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Akagi T, Luong QT, Gui D, et al: Induction of sodium iodide symporter gene and molecular characterisation of HNF3 beta/FoxA2, TTF-1 and C/EBP beta in thyroid carcinoma cells. Br J Cancer. 99:781–788. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Yashiro T, Arai M, Shizume K, et al: Increased activity of insulin-like growth factor-binding protein in human thyroid papillary cancer tissue. Jpn J Cancer Res. 85:46–52. 1994. View Article : Google Scholar : PubMed/NCBI

34 

Soikkeli J, Podlasz P, Yin M, et al: Metastatic outgrowth encompasses COL-I, FN1, and POSTN upregulation and assembly to fibrillar networks regulating cell adhesion, migration, and growth. Am J Pathol. 177:387–403. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Takano T, Matsuzuka F, Sumizaki H, Kuma K and Amino N: Rapid detection of specific messenger RNAs in thyroid carcinomas by reverse transcription-PCR with degenerate primers: specific expression of oncofetal fibronectin messenger RNA in papillary carcinoma. Cancer Res. 57:3792–3797. 1997.PubMed/NCBI

36 

Soto JL, Cabrera CM, Serrano S and López-Nevot MA: Mutation analysis of genes that control the G1/S cell cycle in melanoma: TP53, CDKN1A, CDKN2A, and CDKN2B. BMC Cancer. 5:362005. View Article : Google Scholar : PubMed/NCBI

37 

Arnaldi LA, Borra RC, Maciel RM and Cerutti JM: Gene expression profiles reveal that DCN, DIO1, and DIO2 are underexpressed in benign and malignant thyroid tumors. Thyroid. 15:210–221. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Kroll TG, Sarraf P, Pecciarini L, et al: PAX8-PPARgamma1 fusion oncogene in human thyroid carcinoma [corrected]. Science. 289:1357–1360. 2000. View Article : Google Scholar : PubMed/NCBI

39 

Antico Arciuch VG, Russo MA, Dima M, et al: Thyrocyte-specific inactivation of p53 and Pten results in anaplastic thyroid carcinomas faithfully recapitulating human tumors. Oncotarget. 2:1109–1126. 2011.PubMed/NCBI

Related Articles

Journal Cover

May-2015
Volume 11 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu X, Yao J and Tian W: Microarray technology to investigate genes associated with papillary thyroid carcinoma. Mol Med Rep 11: 3729-3733, 2015
APA
Zhu, X., Yao, J., & Tian, W. (2015). Microarray technology to investigate genes associated with papillary thyroid carcinoma. Molecular Medicine Reports, 11, 3729-3733. https://doi.org/10.3892/mmr.2015.3180
MLA
Zhu, X., Yao, J., Tian, W."Microarray technology to investigate genes associated with papillary thyroid carcinoma". Molecular Medicine Reports 11.5 (2015): 3729-3733.
Chicago
Zhu, X., Yao, J., Tian, W."Microarray technology to investigate genes associated with papillary thyroid carcinoma". Molecular Medicine Reports 11, no. 5 (2015): 3729-3733. https://doi.org/10.3892/mmr.2015.3180