Microarray expression profiling of long non‑coding RNAs in epithelial ovarian cancer

  • Authors:
    • Ye Ding
    • Da‑Zheng Yang
    • Yong‑Ning Zhai
    • Kai Xue
    • Feng Xu
    • Xiao‑Yan Gu
    • Su‑Min Wang
  • View Affiliations

  • Published online on: June 21, 2017     https://doi.org/10.3892/ol.2017.6448
  • Pages: 2523-2530
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Although numerous long non-coding RNAs (lncRNAs) have been identified to be important in human cancer, their potential regulatory roles in epithelial tumorigenesis and tumor progression in ovarian cancer remain unclear. The purpose of the present study was to investigate lncRNAs that were differentially expressed (DE) in epithelial ovarian cancer and to explore their potential functions. The lncRNA profiles in five pairs of human epithelial ovarian cancer tissues and their adjacent normal tissues were described using microarrays. The results of the microarray analysis revealed that 672 upregulated and 549 downregulated (fold‑change ≥2.0) lncRNAs were DE between the cancerous and normal tissues. Reverse transcription‑quantitative polymerase chain reaction was used to validate the microarray results using four upregulated (RP11‑1C1.7, XLOC_003286, growth arrest‑specific 5 and ZNF295‑AS1) and four downregulated (protein tyrosine kinase 7, maternally expressed gene 3, AC079776.2 and ribosomal protein lateral stalk subunit P0 pseudogene 2) lncRNAs. Furthermore, gene ontology and pathway analyses were used to carry out functional analyses of the candidate genes of DE lncRNAs. The results identified lncRNAs with significantly altered expression profiles in human epithelial ovarian cancer cells compared with those in adjacent normal cells. These data offer new insights into the occurrence and development of epithelial ovarian cancer, and these lncRNAs may provide novel molecular biomarkers for further research on epithelial ovarian cancer.

Introduction

Ovarian cancer is the most common cause of mortality from gynecological tumors in women worldwide (1). The 5-year survival rate for patients with advanced ovarian cancer has been reported to be ~30% (2). The incidence of ovarian cancer in Asian countries is considerably lower than that in developed countries, but the difference is reducing (3). In China, the estimated incidence of ovarian cancer during 1999–2010 was 7.91 per 100,000 people (4). Epithelial ovarian cancer accounts for nearly 90% of all ovarian tumors (5). The high mortality of epithelial ovarian cancer is attributed to late-stage diagnosis in >70% of the patients (6). Constant damage and repair of ovarian surface epithelial cells, use of gonadotropin-releasing hormone and steroid hormones, inflammation, genetic factors, and environmental factors have been previously shown to be associated with epithelial ovarian cancer (79); however, the exact molecular mechanisms of its occurrence and development remain to be fully identified.

For more than half a century, the concept of gene was limited to the messenger RNA (mRNA) coding region of the genome. With progress in life science research in the post-genome era, numerous studies have demonstrated the involvement of non-coding RNAs (ncRNAs) at various levels in the cell, including transcription, and post-transcriptional regulation of nuclear internal and external signal communication (10). In addition, these RNAs have been demonstrated to be closely associated with the pathological processes of numerous serious diseases (11). Long ncRNAs (lncRNAs) are non-coding RNAs >200 nt in length. Accumulating evidence indicates that lncRNAs serve an important role in various biological processes such as genomic imprinting, transcription activation and inhibition, chromosome recombination, intranuclear transportation, and organ development (12,13). Certain studies have indicated that aberrant regulation of lncRNAs is associated with various types of human cancer (14). Furthermore, lncRNAs are often used as a potential biomarker in the diagnosis and prognosis of tumors (15). Although a few lncRNAs have been implicated in the progression of epithelial ovarian cancer, the functions of the majority of lncRNAs remain to be investigated.

Therefore, the present study used an lncRNA microarray to identify lncRNAs that are differentially expressed (DE) in epithelial ovarian cancer. The microarray results were verified by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) for specific DE lncRNAs. The present data may provide a molecular basis for understanding the pathogenesis of epithelial ovarian cancer.

Materials and methods

Tissue collection

For tissue collection, five patients with epithelial ovarian cancer were recruited between May and July 2014 at the Department of Gynecology, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University (Nanjing, China). The patients were pathologically confirmed as having epithelial ovarian cancer. Epithelial ovarian cancer tissues and surrounding normal tissues were collected following surgery, snap frozen in liquid nitrogen, and stored at −80°C. Written informed consent was obtained from all patients and the study was approved by the ethics committee of Nanjing Medical University.

RNA extraction

Total RNA was extracted from five pairs of epithelial ovarian cancer and adjacent normal tissues using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's protocol, and quantified using a NanoDrop ND-1000 spectrophotometer (NanoDrop; Thermo Fisher Scientific, Inc., Wilmington, DE, USA). The RNA integrity of each sample was assessed using standard denaturing gel electrophoresis, as previously described (16).

Microarray and data analysis

Microarray analysis was performed by Kangchen Biotech Co., Ltd. (Shanghai, China). Arraystar Human LncRNA Microarray V3.0 (Arraystar Inc., Rockville, MD, USA) is designed for the global profiling of human lncRNAs and protein-coding transcripts. This software is capable of detecting ~30,586 lncRNAs and 26,109 coding transcripts (17). Briefly, mRNA was purified from total RNA upon removal of ribosomal RNA using the mRNA-ONLY™ Eukaryotic mRNA Isolation kit (Epicentre, Madison, WI, USA). Then, each sample was amplified and transcribed into fluorescent complementary RNA (cRNA) along the entire length of the transcripts without 3′-bias using the Quick Amp Labeling kit, One-Color (Agilent Technologies, Inc., Santa Clara, CA, USA) according to the manufacturer's protocol. The labeled cRNAs were purified using the RNeasy Mini kit (Qiagen Inc., Valencia, CA, USA). The concentration and specific activity of the labeled cRNAs (pmol cyanine 3/µg cRNA) were measured by the NanoDrop ND-1000. First, 1 µg of each labeled cRNA was fragmented by adding 5 µl of 10X blocking agent and 1 µl of 25X fragmentation buffer (both Agilent Technologies, Inc.). The mixture was then heated at 60°C for 30 min, and subsequently, 25 µl of 2X hybridization buffer (GE Healthcare Life Sciences, Little Chalfont, UK) was added to dilute the labeled cRNA. For microarray analysis, 50 µl of the hybridization solution was dispensed into the gasket slide and assembled to the lncRNA expression microarray slide. The slides were incubated for 17 h at 65°C in a Microarray Hybridization Oven (Agilent Technologies, Inc.). The hybridized arrays were washed with Gene Expression Wash Buffer (Agilent Technologies, Inc.) and scanned with using the G2505C Microarray Scanner System (Agilent Technologies, Inc.). Feature Extraction software version 11.0.1.1 (Agilent Technologies, Inc.) was used to analyze the acquired array images. Quantile normalization and subsequent data processing were performed using the GeneSpring GX v12.1 software package (Agilent Technologies, Inc.).

Gene ontology (GO) and pathway analyses

GO and pathway analyses were used to determine the roles of DE mRNAs in biological pathways or GO terms. Differentially regulated mRNAs were uploaded into the Database for Annotation, Visualization and Integrated Discovery (http://david.abcc.ncifcrf.gov/), which utilized GO terms to identify the molecular function represented in the gene profile. Pathway analysis was carried out based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (http://www.genome.ad.jp/kegg/).

RT-qPCR validation

Total RNA was reverse transcribed into complementary DNA (cDNA) using the AMV Reverse Transcriptase (Promega Corporation, Madison, WI, USA) according to the manufacturer's protocol. RT-qPCR was performed using an Applied Biosystems 7300 Real-Time PCR Sequence Detection System (Thermo Fisher Scientific, Inc.). RT-qPCR was conducted using 1 µl of cDNA, 12.5 µl of 2X SYBR Green PCR Master Mix (Applied Biosystems; Thermo Fisher Scientific, Inc.), 10.5 µl of diethyl pyrocarbonate-treated water, and 0.5 µl of 10 µM forward and reverse primers, in a total volume of 25 µl. The following specific primers were used for PCR: RP11-1C1.7 forward, 5′-CTCAGGCTTGGCTCAGACAC-3′ and reverse, 5′-GCAAACAGCCTTGGAGAAGC-3′; XLOC_003286 forward, 5′-AAGGGATCTGGTCTTCAACA-3′ and reverse, 5′-TTCCACCATGTAATGGGTCC-3′; growth arrest specific 5 (GAS5) forward, 5′-TGAAGTCCTAAAGAGCAAGCC-3′ and reverse, 5′-ACCAGGAGCAGAACCATTAAG-3′; ZNF295-AS1 forward, 5′-CCCAGGAGGGAGGTGATACT-3′ and reverse, 5′-TGGGTAGCTTGTGAACCACC-3′; protein tyrosine kinase 7 (PTK7) forward, 5′-GGAAGCCACACTTCACCTAGCAG-3′ and reverse, 5′-CTGCCACAGTGAGCTGGACATGG-3′; maternally expressed gene 3 (MEG3) forward, 5′-GCTCTACTCCGTGGAAGCAC-3′ and reverse, 5′-CAAACCAGGAAGGAGACGAG-3′; AC079776.2, forward, 5′-GCCGATGGTAGAGAAGACCG-3′ and reverse, 5′-GGGGCTCAGAAGCCATCTTT-3′; and ribosomal protein lateral stalk subunit P0 pseudogene 2 (RPLP0P2) forward, 5′-AAAAACGATCAACGAACCTT-3′ and reverse, 5′-AATCGTCTCTGCTTTTCTTG-3′. The PCR conditions were as follows: Denaturation at 95°C for 10 min, followed by 40 cycles of amplification and quantification at 95°C for 15 sec and 60°C for 1 min. GAPDH (forward, 5′-CCGGGAAACTGTGGCGTGATGG-3′ and reverse, 5′-AGGTGGAGGTATGGGTGTCGCTGTT-3′) was used as the internal control. The experiments were performed in triplicate. The relative fold-change was calculated using the 2−ΔΔCq method (18).

Statistical analysis

The lncRNAs and mRNAs that exhibited significantly different expression levels between the two groups were identified through P-value/false discovery rate filtering. DE lncRNAs and mRNAs were identified by fold-change filtering and Student's t test. All data were expressed as means ± standard deviation. Statistical analysis was performed using SPSS 10.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

DE lncRNAs and mRNAs

A total of 1221 lncRNAs were significantly DE between the tumor and control groups (fold-change ≥2.0), among which, 672 were upregulated and 549 were downregulated. Among the DE mRNAs between the two groups, 525 were upregulated and 418 were downregulated. Partial results for the DE lncRNAs and mRNAs are listed in Tables I and II, respectively.

Table I.

Screening of differentially expressed lncRNAs (tumor vs. normal).

Table I.

Screening of differentially expressed lncRNAs (tumor vs. normal).

RegulationlncRNAFold-changeChromosomal localizationRNA length, bp
UpRP5-857K21.391.6369032Chr1437
Upuc001zjx.164.7598797Chr15641
UpDQ57353939.8247748Chr91,713
UpRP11-1C1.738.8887511Chr5483
UpXLOC_00413425.2266495Chr4261
UpRP11-872J21.321.4447620Chr141,512
UpLOC33881718.7987392Chr123,684
UpCDKN2B-AS115.7325039Chr91,067
UpHLA-DRB615.1244408Chr6715
UpUCA112.9894370Chr191,413
UpBX004987.511.7750069Chr1736
UpFOLH1B10.8238534Chr112,163
UpZNF295-AS1   9.3852619Chr211,073
UpAK054990   9.1453539Chr22,070
UpAP001615.9   8.1669081Chr21461
UpGAS5   7.8179616Chr1822
UpLINC00152   7.0158480Chr2455
UpXLOC_003286   6.5502125Chr3409
UpDPY19L2P2   4.4375165Chr73,433
UpAL833634   2.2275523Chr111,885
DownCTD-2536I1.158.1029053Chr15614
DownBC07178946.6526362Chr32,730
DownRP11-548O1.341.2599738Chr3483
DownMEG335.0543457Chr141,351
DownRP11-471J12.130.7697326Chr4892
DownLEMD1-AS124.3438594Chr12,781
DownCLCN620.5708229Chr15,697
DownAL132709.519.7389918Chr14644
DownXLOC_01046317.3764962Chr139,590
DownCACNA1G-AS115.5318244Chr171,450
DownAC079776.212.6763061Chr2400
DownRP11-998D10.210.6026574Chr14548
DownLOC253044   7.5169687Chr151,735
DownPVT1   4.8097586Chr8654
DownAX747026   4.3736710Chr12,133
DownOPA1-AS1   3.4889195Chr3513
DownPTK7   3.1639252Chr64,040
DownRP11-799B12.4   2.5604262Chr18735
DownRPLP0P2   2.4997850Chr11573
DownHOTAIR   2.1863176Chr122,370

[i] lncRNA, long non-coding RNA; Chr, chromosome.

Table II.

Screening of differentially expressed mRNAs (tumor vs. normal).

Table II.

Screening of differentially expressed mRNAs (tumor vs. normal).

RegulationmRNAFold-changeChromosomal localizationRNA length, bp
UpGAL112.8379148Chr11778
UpLAMC2   94.8845376Chr15,623
UpCCNA1   80.0032110Chr131,841
UpMUC1   62.3494142Chr1878
UpWDR69   54.5549954Chr21,669
UpENKUR   47.3980040Chr103,382
UpSTOML3   32.8593469Chr131,936
UpKIAA0101   27.1783242Chr151,345
UpCCNB2   20.5538621Chr151,566
UpSLC1A3   18.5310330Chr53,670
UpSAA2   16.4134886Chr11594
UpFGF18   14.3701010Chr51,999
UpUBE2C   12.9501868Chr20520
UpNAA16   9.5211755Chr131,833
UpKCNIP4   7.5399784Chr42,371
UpSLITRK6   6.5738521Chr134,199
UpCEP44   4.4520673Chr43,290
UpC20orf201   3.2842057Chr20868
UpDHCR7   2.3597491Chr112,665
UpRNLS   2.0603828Chr102,420
DownITM2A110.4209953ChrX1,719
DownZBTB16   82.7721198Chr112,417
DownCPXM1   80.2367909Chr202,409
DownGATA4   69.2038646Chr83,419
DownAPOD   54.0064083Chr31,130
DownDCN   48.0233786Chr121,336
DownGNG11   37.6068614Chr7964
DownDHRS2   32.5328881Chr141,709
DownACADL   28.4221424Chr22,565
DownLCE1C   24.4961395Chr1695
DownMATN2   18.1700061Chr84065
DownLCE2C   16.5275274Chr1614
DownPPP1R14A   10.4409979Chr19782
DownOSR2   8.4374819Chr81,907
DownAKT3   6.6042407Chr17,091
DownIL28RA   5.1905507Chr14,432
DownPIK3IP1   3.6901547Chr222,478
DownSULF1   3.4824364Chr85,716
DownDCAF4L2   2.8399696Chr83,339
DownMARK3   2.6464566Chr143,519

[i] mRNA, messenger RNA; Chr, chromosome.

Validation of de lncRNAs

The results of the microarray analysis were confirmed by RT-qPCR of eight randomly selected lncRNAs. GAPDH was used as a normalization control. Of these randomly selected lncRNAs, four (RP11-1C1.7, XLOC_003286, GAS5 and ZNF295-AS1) were upregulated and the other four (PTK7, MEG3, AC079776.2 and RPLP0P2) were downregulated in epithelial ovarian cancer samples compared with their expression levels in adjacent normal tissues of the same individual. As the results of RT-qPCR and microarray analyses are consistent (Fig. 1), these data can be used with confidence in further research.

Pathway analysis

Pathway analysis is a functional method of mapping genes to KEGG pathways (19). Based on the KEGG database (http://www.genome.jp/kegg), KEGG pathway analysis was employed for DE mRNAs. Each P-value denoted the significance of the corresponding pathway, while the EASE Score, Fisher's P-value or hypergeometric P-value denoted the significance of the pathway correlated to the conditions. A low P-value indicated a marked significance of the pathway (P-value cut-off, 0.05). The bar plots in Fig. 2 show the top 10 enrichment scores [-log10 (P-value)] of the significant enrichment pathway. Fig. 2 presents the results of the KEGG pathway analysis for the upregulated and downregulated mRNAs.

GO analysis

The GO project provides a controlled vocabulary to describe gene and gene product attributes in any organism (http://www.geneontology.org). The ontology covers three domains: Biological processes, cellular components and molecular function. Fisher's exact test is used to determine if there are any more overlaps between the DE gene list and the GO annotation list than what is expected by chance. The P-value denotes the significance of enrichment of GO terms in the DE genes. The lower the P-value, the more significant is the GO term (P≤0.05 is recommended) (20). The bar plots in Fig. 3 show the 10 most significant enrichment terms with the most number of DE genes.

Discussion

As increasing research has focused on the function of lncRNAs in epithelial ovarian cancer, an increasing number of lncRNAs have been identified. For example, Gao et al demonstrated that the lncRNA human ovarian cancer-specific transcript 2 promotes tumor cell migration, invasion and proliferation in epithelial ovarian cancer by modulating microRNA let-7b availability (21). lncRNA H19 expression was inhibited by histone H1.3, which contributes to the suppression of epithelial ovarian carcinogenesis (22). However, the genome-wide expression and the biological functional significance of lncRNAs in epithelial ovarian cancer remain unknown.

In the present study, microarray analysis was used to compare lncRNA expression in epithelial ovarian cancer cells and adjacent normal tissues, and 1221 DE lncRNAs (672 upregulated and 549 downregulated) were identified. These results were further confirmed via RT-qPCR for eight randomly selected lncRNAs.

A previous study has reported that Hox transcript antisense intergenic RNA (HOTAIR) is a 2.2-kb lncRNA located at the HOXC locus (23). It has been reported that suppression of HOTAIR expression in highly metastatic epithelial ovarian cancer cell lines significantly reduced cell invasion, and the HOTAIR expression levels were highly positively correlated with the International Federation of Gynecology and Obstetrics stage (24). The MEG3 gene is located in chromosome 14q32 (25), and is expressed in numerous normal tissues, but its expression level has been reported by various previous studies to be either downregulated or absent in a variety of tumor tissues, including ovarian cancer cells and epithelial ovarian cancer tissues (2628). In the present study, HOTAIR was upregulated and MEG3 was downregulated in epithelial ovarian cancer vs. normal tissues. These results confirmed that HOTAIR and MEG3 serve a critical role in the occurrence, development and invasion of epithelial ovarian cancer.

GAS5 is encoded at chromosome 1q25, and was originally isolated from NIH-3T3 cells by subtractive hybridization (29). Several recent studies have shown that GAS5 is an lncRNA that functions as a tumor suppressor. For example, Cao et al noticed that patients with cervical cancer with reduced expression of GAS5 have significantly poorer overall survival than those with higher GAS5 expression (30). Shi et al reported that GAS5 expression was downregulated in non-small cell lung cancer tissues compared with that in noncancerous tissues, and was highly associated with tumor size and tumor-node-metastasis stage (31). However, in the present study, it was observed that the expression of GAS5 was upregulated in epithelial ovarian cancer compared with that in adjacent healthy tissues. The majority of scholars agree that glucocorticoids serve an important role in the regulation of ovarian epithelial function, and they are closely associated with the occurrence and development of ovarian cancer (32,33). In another study, glucocorticoids were demonstrated to significantly inhibit the proliferation of human ovarian cancer cells (34). Therefore, it can be hypothesized that, as a glucocorticoid receptor response element (GRE) analogue, GAS5 may be able to inhibit glucocorticoid production by competing with GRE to associate with the DNA-binding domain of the glucocorticoid receptor (35).

To understand the function of the targets of DE lncRNAs, GO terms and KEGG pathway annotation were applied in the present study to the target gene pool. The GO analysis revealed that the DE genes were associated with mitogen-activated protein kinase phosphatase activity, major histocompatibility complex class II receptor activity and DNA metabolic processes, which is consistent with previous research (3638). Previous studies have demonstrated that signaling pathways, including the Ras, p53 and transforming growth factor-β signaling pathways, serve a critical role in the regulation of pathophysiological processes in ovarian cancer (3941). In addition to these signaling pathways, the present study also demonstrated that focal adhesion, extracellular matrix-receptor interaction, cell adhesion molecules, cell cycle, transcriptional misregulation in cancer and other signaling pathways were involved in the pathogenesis of epithelial ovarian cancer.

In summary, the present study identified lncRNAs that were aberrantly expressed in epithelial ovarian cancer compared with their expression in matched normal tissue. Further studies are required to reveal the possible biological functions and mechanism of these lncRNAs.

Acknowledgements

The present study was supported by the Science and Technology Development Foundation of Nanjing Medical University (grant no. 2014NJMUZD050). The authors thank Kangchen Biotech Co., Ltd. (Shanghai, China) for their technical assistance.

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Lowe KA, Chia VM, Taylor A, O'Malley C, Kelsh M, Mohamed M, Mowat FS and Goff B: An international assessment of ovarian cancer incidence and mortality. Gynecol Oncol. 130:107–114. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Chiang YC, Chen CA, Chiang CJ, Hsu TH, Lin MC, You SL, Cheng WF and Lai MS: Trends in incidence and survival outcome of epithelial ovarian cancer: 30-year national population-based registry in Taiwan. J Gynecol Oncol. 24:342–351. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Wang B, Liu SZ, Zheng RS, Zhang F, Chen WQ and Sun XB: Time Trends of Ovarian Cancer Incidence in China. Asian Pac J Cancer Prev. 15:191–193. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Lawrie TA, Bryant A, Cameron A, Gray E and Morrison J: Pegylated liposomal doxorubicin for relapsed epithelial ovarian cancer. Cochrane Database Syst Rev: CD006910. 2013. View Article : Google Scholar

6 

Gadducci A, Cosio S, Zola P, Landoni F, Maggino T and Sartori E: Surveillance procedures for patients treated for epithelial ovarian cancer: A review of the literature. Int J Gynecol Cancer. 17:21–31. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Adams SV, Quraishi SM, Shafer MM, Passarelli MN, Freney EP, Chlebowski RT, Luo J, Meliker JR, Mu L, Neuhouser ML and Newcomb PA: Dietary cadmium exposure and risk of breast, endometrial and ovarian cancer in the Women's Health Initiative. Environ Health Perspect. 122:594–600. 2014.PubMed/NCBI

8 

Schock H, Surcel HM, Zeleniuch-Jacquotte A, Grankvist K, Lakso HÅ, Fortner RT, Kaaks R, Pukkala E, Lehtinen M, Toniolo P and Lundin E: Early pregnancy sex steroids and maternal risk of epithelial ovarian cancer. Endocr Relat Cancer. 21:831–844. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Haruta S, Furukawa N, Yoshizawa Y, Tsunemi T, Nagai A, Kawaguchi R, Tanase Y, Yoshida S and Kobayashi H: Molecular genetics and epidemiology of epithelial ovarian cancer. Oncol Rep. 26:1347–1356. 2011.PubMed/NCBI

10 

Costa FF: Non-coding RNAs, epigenetics and complexity. Gene. 410:9–17. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Taft RJ, Pang KC, Mercer TR, Dinger M and Mattick JS: Non-coding RNAs: Regulators of disease. J Pathol. 220:126–139. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Hung T and Chang HY: Long noncoding RNA in genome regulation: Prospects and mechanisms. RNA Biol. 7:582–585. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Bonasio R and Shiekhattar R: Regulation of transcription by long noncoding RNAs. Annu Rev Genet. 48:433–455. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Haemmerle M and Gutschner T: Long non-coding RNAs in cancer and development: Where do we go from here? Int J Mol Sci. 16:1395–1405. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Kunej T, Obsteter J, Pogacar Z, Horvat S and Calin GA: The decalog of long non-coding RNA involvement in cancer diagnosis and monitoring. Crit Rev Clin Lab Sci. 51:344–357. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Zuo C, Wang Z, Lu H, Dai Z, Liu X and Cui L: Expression profiling of lncRNAs in C3H10T1/2 mesenchymal stem cells undergoing early osteoblast differentiation. Mol Med Rep. 8:463–467. 2013.PubMed/NCBI

17 

Li J, Long W, Li Q, Zhou Q, Wang Y, Wang H, Zhou B and Li J: Distinct expression profiles of lncRNAs between regressive and mature scars. Cell Physiol Biochem. 35:663–675. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

19 

Kanehisa M: Molecular network analysis of diseases and drugs in KEGG. Methods Mol Biol. 939:263–275. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Gu S, Li G, Zhang X, Yan J, Gao J, An X, Liu Y and Su P: Aberrant expression of long noncoding RNAs in chronic thromboembolic pulmonary hypertension. Mol Med Rep. 11:2631–2643. 2015.PubMed/NCBI

21 

Gao Y, Meng H, Liu S, Hu J, Zhang Y, Jiao T, Liu Y, Ou J, Wang D, Yao L, et al: LncRNA-HOST2 regulates cell biological behaviors in epithelial ovarian cancer through a mechanism involving microRNA let-7b. Hum Mol Genet. 24:841–852. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Medrzycki M, Zhang Y, Zhang W, Cao K, Pan C, Lailler N, McDonald JF, Bouhassira EE and Fan Y: Histone h1.3 suppresses h19 noncoding RNA expression and cell growth of ovarian cancer cells. Cancer Res. 74:6463–6473. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, Goodnough LH, Helms JA, Farnham PJ, Segal E and Chang HY: Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 129:1311–1323. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Qiu JJ, Lin YY, Ye LC, Ding JX, Feng WW, Jin HY, Zhang Y, Li Q and Hua KQ: Overexpression of long non-coding RNA HOTAIR predicts poor patient prognosis and promotes tumor metastasis in epithelial ovarian cancer. Gynecol Oncol. 134:121–128. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Miyoshi N, Wagatsuma H, Wakana S, Shiroishi T, Nomura M, Aisaka K, Kohda T, Surani MA, Kaneko-Ishino T and Ishino F: Identification of an imprinted gene, Meg3/Gtl2 and its human homologue MEG3, first mapped on mouse distal chromosome 12 and human chromosome 14q. Genes Cells. 5:211–220. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Zhang X, Zhou Y, Mehta KR, Danila DC, Scolavino S, Johnson SR and Klibanski A: A pituitary-derived MEG3 isoform functions as a growth suppressor in tumor cells. J Clin Endocrinol Metab. 88:5119–5126. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Huarte M, Guttman M, Feldser D, Garber M, Koziol MJ, Kenzelmann-Broz D, Khalil AM, Zuk O, Amit I, Rabani M, et al: A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell. 142:409–419. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Sheng X and Li J, Yang L, Chen Z, Zhao Q, Tan L, Zhou Y and Li J: Promoter hypermethylation influences the suppressive role of maternally expressed 3, a long non-coding RNA, in the development of epithelial ovarian cancer. Oncol Rep. 32:277–285. 2014.PubMed/NCBI

29 

Schneider C, King RM and Philipson L: Genes specifically expressed at growth arrest of mammalian cells. Cell. 54:787–793. 1988. View Article : Google Scholar : PubMed/NCBI

30 

Cao S, Liu W, Li F, Zhao W and Qin C: Decreased expression of lncRNA GAS5 predicts a poor prognosis in cervical cancer. Int J Clin Exp Pathol. 7:6776–6783. 2014.PubMed/NCBI

31 

Shi X, Sun M, Liu H, Yao Y, Kong R, Chen F and Song Y: A critical role for the long non-coding RNA GAS5 in proliferation and apoptosis in non-small-cell lung cancer. Mol Carcinog. 54:E1–E12. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Melhem A, Yamada SD, Fleming GF, Delgado B, Brickley DR, Wu W, Kocherginsky M and Conzen SD: Administration of glucocorticoids to ovarian cancer patients is associated with expression of the anti-apoptotic genes SGK1 and MKP1/DUSP1 in ovarian tissues. Clin Cancer Res. 15:3196–3204. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Dickinson RE, Fegan KS, Ren X, Hillier SG and Duncan WC: Glucocorticoid regulation of SLIT/ROBO tumour suppressor genes in the ovarian surface epithelium and ovarian cancer cells. PLoS One. 6:e277922011. View Article : Google Scholar : PubMed/NCBI

34 

Xu MJ, Fang GE, Liu YJ and Song LN: Effects of glucocorticoid on proliferation, differentiation and glucocorticoid receptor expression in human ovarian carcinoma cell line 3AO. Acta Pharmacol Sin. 23:819–823. 2002.PubMed/NCBI

35 

Kino T, Hurt DE, Ichijo T, Nader N and Chrousos GP: Noncoding RNA gas5 is a growth arrest- and starvation-associated repressor of the glucocorticoid receptor. Sci Signal. 3:ra82010. View Article : Google Scholar : PubMed/NCBI

36 

Beauchamp MC, Yasmeen A, Knafo A and Gotlieb WH: Targeting insulin and insulin-like growth factor pathways in epithelial ovarian cancer. J Oncol. 2010:2570582010. View Article : Google Scholar : PubMed/NCBI

37 

Dadmarz RD, Ordoubadi A, Mixon A, Thompson CO, Barracchini KC, Hijazi YM, Steller MA, Rosenberg SA and Schwartzentruber DJ: Tumor-infiltrating lymphocytes from human ovarian cancer patients recognize autologous tumor in an MHC class II-restricted fashion. Cancer J Sci Am. 2:263–272. 1996.PubMed/NCBI

38 

Beesley J, Jordan SJ, Spurdle AB, Song H, Ramus SJ, Kjaer SK, Hogdall E, DiCioccio RA, McGuire V, Whittemore AS, et al: Association between single-nucleotide polymorphisms in hormone metabolism and DNA repair genes and epithelial ovarian cancer: results from two Australian studies and an additional validation set. Cancer Epidemiol Biomarkers Prev. 16:2557–2565. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Mei FC, Young TW, Liu J and Cheng X: RAS-mediated epigenetic inactivation of OPCML in oncogenic transformation of human ovarian surface epithelial cells. FASEB J. 20:497–499. 2006.PubMed/NCBI

40 

Li J, Zhang Y, Gao Y, Cui Y, Liu H, Li M and Tian Y: Downregulation of HNF1 homeobox B is associated with drug resistance in ovarian cancer. Oncol Rep. 32:979–988. 2014.PubMed/NCBI

41 

Ji M, Shi H, Xie Y, Zhao Z, Li S, Chang C, Cheng X and Li Y: Ubiquitin specific protease 22 promotes cell proliferation and tumor growth of epithelial ovarian cancer through synergy with transforming growth factor β1. Oncol Rep. 33:133–140. 2015.PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 14 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ding Y, Yang DZ, Zhai YN, Xue K, Xu F, Gu XY and Wang SM: Microarray expression profiling of long non‑coding RNAs in epithelial ovarian cancer. Oncol Lett 14: 2523-2530, 2017
APA
Ding, Y., Yang, D., Zhai, Y., Xue, K., Xu, F., Gu, X., & Wang, S. (2017). Microarray expression profiling of long non‑coding RNAs in epithelial ovarian cancer. Oncology Letters, 14, 2523-2530. https://doi.org/10.3892/ol.2017.6448
MLA
Ding, Y., Yang, D., Zhai, Y., Xue, K., Xu, F., Gu, X., Wang, S."Microarray expression profiling of long non‑coding RNAs in epithelial ovarian cancer". Oncology Letters 14.2 (2017): 2523-2530.
Chicago
Ding, Y., Yang, D., Zhai, Y., Xue, K., Xu, F., Gu, X., Wang, S."Microarray expression profiling of long non‑coding RNAs in epithelial ovarian cancer". Oncology Letters 14, no. 2 (2017): 2523-2530. https://doi.org/10.3892/ol.2017.6448