Open Access

Construction of lncRNA‑miRNA‑mRNA networks reveals functional lncRNAs in abdominal aortic aneurysm

  • Authors:
    • Lu Tian
    • Xiaofeng Hu
    • Yangyan He
    • Ziheng Wu
    • Donglin Li
    • Hongkun Zhang
  • View Affiliations

  • Published online on: September 4, 2018     https://doi.org/10.3892/etm.2018.6690
  • Pages: 3978-3986
  • Copyright: © Tian et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Abdominal aortic aneurysm (AAA) is one of the most significant causes of morbidity and mortality in populations aged >65 years worldwide. However, the underlying mechanisms of AAA based on the competitive endogenous RNA (ceRNA) hypothesis have remained elusive. In the present study, differently expressed long non‑coding RNA (lncRNA)‑microRNA (miRNA)‑mRNA networks in AAA were constructed by analyzing public datasets, including GSE7084, GSE24194 from rats and that of a previous study. A total of 1,219 mRNAs, 2,093 lncRNAs and 57 miRNAs were identified to differently express in AAA. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were performed to explore the potential roles of differently expressed lncRNAs based on their regulating mRNAs. Based on the ceRNA hypothesis, lncRNA‑miRNA‑mRNA networks in AAA were, for the first time, constructed at a system‑wide level. The present study identified 5 upregulated lncRNAs [nuclear paraspeckle assembly transcript 1, cyclin‑dependent kinase inhibitor 2B antisense RNA 1, small Cajal body‑specific RNA 10, AC005224.4 and SUMO1/sentrin/SMT3‑specific peptidase 3‑eukaryotic translation initiation factor 4A1] and the downregulated zinc ribbon domain containing 1 antisense RNA 1 as key lncRNAs in ceRNA networks. To the best of our knowledge, the present study was the first to screen ceRNA networks in AAA. In addition, key lncRNA‑mRNA‑biological processes analysis indicated that these key lncRNAs were involved in regulating signal transduction, protein amino acid phosphorylation, immune response, transcription, development and cell differentiation. The present study provides novel clues to explore the molecular mechanisms of AAA progression in terms of lncRNA implication.

Introduction

Abdominal aortic aneurysm (AAA) is defined as a maximum infrarenal abdominal aortic aneurysm with a diameter of ≥3.0 cm and is characterized by permanent, localized dilations of the abdominal aorta (1,2). AAA is one of the most significant cause of morbidity and mortality in populations aged >65 years worldwide (3,4). Acute rupture is the most dangerous clinical consequence of AAA progression and causes ~80% of associated deaths in the US (5). Pathologic features of AAA include vascular smooth muscle cell apoptosis, infiltration of inflammatory cells, loss of integrity of the arterial wall, increase of oxidative stress and significant matrix degradation (6). However, the precise molecular mechanisms underlying the progression of AAA progression still remain elusive. Therefore, it is important to elucidate the etiological mechanisms of AAA progression to develop novel targets for the diagnosis, treatment, and prognostication of AAA patients.

Long non-coding (lnc)RNAs are a class of non-coding RNAs of >200 nucleotides in length, which have little or no protein-coding function (7). It has been discovered that lncRNAs have important roles in the progression of numerous human diseases, including AAA (8). For instance, Yang et al (9) identified 3,688 differentially expressed lncRNAs between AAA and normal tissues. lncRNAs regulate the protein expression at epigenetic, transcriptional and post-translational levels (1013). One of the most well-known mechanisms of lncRNAs is their action as competing endogenous (ce)RNAs (14). The ceRNA hypothesis, which was proposed by Tay et al (15), holds that pseudogenes, lncRNAs, circular RNAs and mRNAs may impair micro (mi)RNA activity through sequestration, thereby upregulating miRNA target gene expression. Franco-Zorrilla et al (16) reported for the first time that non-coding RNA interferon-β promoter stimulator 1 promoted phosphate metabolism (PHO)2 protein in plants by sequestering miR-399 and preventing it from inhibiting the stability and translation of PHO2 mRNA. Poliseno et al (17) also reported that certain protein-coding genes and their pseudogenes contain the same evolutionarily conserved miRNA binding sites in their 3′-untranslated regions, and that they regulate their respective expression levels by competing for miRNA binding. Emerging studies have indicated that ceRNAs act as important regulators in different types of disease, including cancer, cardiac fibrosis, rheumatoid arthritis and type 2 diabetes mellitus (1820). Thus, constructing ceRNA networks provides a novel perspective to explore the function of yet uncharacterized lncRNAs involved in AAA progression.

In the present study, differentially expressed RNAs, miRNA and mRNAs in AAA were identified from data provided by the National Center for Biotechnology Information Gene Expression Omnibus (NCBI GEO), and lncRNA-miRNA-mRNA networks were constructed. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were also performed to explore the potential roles of differentially expressed lncRNAs in AAA. The present study aimed to provide useful information to identify novel lncRNAs as biomarkers for AAA.

Materials and methods

Microarray data

In the present study, two public datasets, GSE7084 (21) and that provided by Yang et al (9) were analyzed to identify differentially expressed mRNAs in AAA. The GSE7084 dataset was downloaded from the NCBI GEO database (https://www.ncbi.nlm.nih.gov/geo/). The dataset from Yang et al (9) was downloaded from the supplementary information of their publication. To further identify differentially expressed lncRNAs in AAA, the dataset by Yang et al (9) was analyzed, providing 896 upregulated and 1,197 downregulated lncRNAs. Differently expressed miRNAs were determined from the GSE24194 dataset from rats (22), which was downloaded from the NCBI GEO website. A t-test (23) in the Limma package (24) in R (25) was used to identify differentially expressed genes between normal and AAA samples. The threshold for the differentially expressed genes (DEGs) was set as a corrected P-value of <0.05 and |log2 fold-change (FC)|≥1.

GO and KEGG pathway analysis

To identify functions of DEGs in AAA, GO term enrichment analysis in the categories biological process, cellular component and molecular function was performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID; http://david.ncifcrf.gov/). KEGG pathway enrichment analysis was also performed to identify pathways enriched by DEGs in AAA using DAVID. The P-value was calculated by hypergeometric distribution and a pathway with P<0.05 was considered as significant.

Construction of the lncRNA-miRNA-mRNA network

The StarBase dataset was used to identify potentially dysregulated lncRNA-miRNA pairs. Next, miRcode (26), StarBase (27) and the Targetscan database (http://www.targetscan.org) were used to identify miRNA-mRNA pairs. Finally, lncRNA-miRNA-mRNA networks were constructed. In the present study, only downregulated miRNAs and upregulated mRNAs were integrated into the ceRNA network for upregulated lncRNAs, while only upregulated miRNAs and downregulated mRNAs were integrated into the ceRNA network fir downregulated lncRNAs.

Results

Identification of differentially expressed lncRNAs, mRNAs and miRNAs in AAA

In the present study, two public datasets, GSE7084 (21) and that of Yang et al (9), were analyzed to identify differentially expressed mRNAs (Fig. 1A and B). mRNAs with a FC of ≥2 or ≤0.5 and P<0.05 were selected as differentially expressed mRNAs. A total of 2,292 upregulated and 2,501 downregulated mRNAs were identified from the GSE7084 dataset. In the dataset from Yang et al (9), 1,501 upregulated and 969 downregulated mRNAs were identified. In total, the two datasets had 722 upregulated and 497 downregulated mRNAs in common (Fig. 1C and D). To identify differentially expressed lncRNAs, the dataset from Yang et al (9) was re-analyzed, and 896 upregulated and 1,197 downregulated lncRNAs were obtained (Fig. 1E). Furthermore, miRNAs with a FC of ≥2 or ≤0.5 and P<0.05 were selected as differentially expressed miRNAs. A total of 21 upregulated miRNAs and 36 downregulated miRNAs were selected for further ceRNA network construction by analyzing the GSE24194 dataset obtained from rats (Fig. 1F). Hierarchical clustering provided systematic variations in the expression of mRNAs, miRNAs and lncRNAs in AAA.

Functional prediction of differentially expressed mRNAs in AAA

GO and KEGG pathway analyses were performed to explore the function of the 1,219 differentially expressed mRNAs using DAVID. The results indicated that the upregulated mRNAs were enriched in GO terms including immune response, signal transduction, inflammatory response, chemotaxis, cell adhesion, cell-cell signaling, protein amino acid phosphorylation, proteolysis, cellular defense response, regulation of transcription and apoptosis (Fig. 2A), while the downregulated genes were enriched in GO terms including regulation of transcription, oxidation/reduction, cell adhesion, signal transduction, ion transport, protein amino acid phosphorylation, development, nervous system development, muscle development and cell-matrix adhesion (Fig. 2B).

KEGG pathway analysis revealed that the upregulated mRNAs were mainly involved in cytokine-cytokine receptor interaction, hematopoietic cell lineages, the B-cell receptor signaling pathway, cell adhesion molecules, leukocyte transendothelial migration, the T-cell receptor signaling pathway, graft-versus-host disease, Toll-like receptor signaling pathway and the mitogen-activated protein kinase signaling pathway (Fig. 2C), while the downregulated genes were involved in regulation of the actin cytoskeleton, calcium signaling pathway, focal adhesion, neuroactive ligand-receptor interaction, tight junctions, tryptophan metabolism, and valine, leucine and isoleucine degradation (Fig. 2D).

Construction of lncRNA-miRNA-mRNA networks in AAA

To explore the functions of lncRNAs acting as miRNA sponges in AAA, two lncRNA-mediated ceRNA networks were first constructed by using bioinformatics analysis. First, the StarBase dataset was used to identify potentially dysregulated lncRNA-miRNA pairs. Next, miRcode (26), StarBase (27) and the Targetscan database were used to identify miRNA-mRNA pairs. Finally, lncRNA-miRNA-mRNA networks were constructed. In the present study, only downregulated miRNAs and upregulated mRNAs were integrated into the ceRNA network mediated by upregulated lncRNAs, while, only upregulated miRNAs and downregulated mRNAs were integrated into the ceRNA network mediated by downregulated lncRNAs.

As presented in Fig. 3, the upregulated lncRNA-miRNA-mRNA network contained 38 lncRNA nodes, 374 mRNA nodes, 27 miRNA nodes and 2,021 edges. In addition, the downregulated lncRNA-miRNA-mRNA network comprised 28 lncRNA nodes, 238 mRNA nodes, 12 miRNA nodes and 729 edges.

Identification of key lncRNA-mRNA-biological processes sub-network

According to the ceRNA network analysis, all node degrees were calculated to identify hub lncRNAs, which have critical roles in biological networks. Five upregulated lncRNAs [nuclear paraspeckle assembly transcript 1 (NEAT1), cyclin-dependent kinase inhibitor 2B (CDKN2B)-antisense RNA 1 (AS1), small Cajal body-specific RNA 10 (SCARNA10), AC005224.4 and SUMO1/sentrin/SMT3-specific peptidase 3 (SENP3)-eukaryotic translation initiation factor 4A1 (EIF4A1)] and the downregulated zinc ribbon domain containing 1 (ZNRD1-AS1) were identified as key lncRNAs in the ceRNA networks and interacted with >5 different miRNAs (Fig. 4).

Furthermore, key lncRNA-mRNA-biological processes analysis indicated that NEAT1, CDKN2B-AS1, SCARNA10, AC005224.4 and SENP3-EIF4A1 were involved in regulating signal transduction, protein amino acid phosphorylation and immune response (Fig. 5A). ZNRD1-AS1 was associated with the regulation of transcription, development, and cell differentiation (Fig. 5B).

Discussion

AAA is one of the most significant causes of morbidity and mortality in populations aged >65 years worldwide (3,4). Previous studies reported that various proteins, including mitochondrial uncoupling protein-2, c-Jun N-terminal kinase and matrix metalloproteinase-9 and miRNAs, including miRNA-103a and miRNA-516a-5p, were associated with AAA progression (28). Of note, the underlying mechanisms of AAA have remained to be fully elucidated. lncRNAs are a class of non-coding RNAs with no protein-coding function (7). Several studies have focused on exploring the roles of lncRNAs in aortic aneurysm disease. Hypoxia-inducible factor 1α-AS1 was the first lncRNA reported to be involved in the pathogenesis of thoracic aortic aneurysm (8). Yang et al (9) identified 3,688 differentially expressed lncRNAs between AAA and normal tissues. However, the molecular functions and detailed mechanisms of lncRNAs in AAA remain largely elusive. In the present study, DEGs in AAA were explored by analyzing a series of public datasets, including GSE7084, the dataset from Yang et al (9) and GSE24194 obtained from rats. A total of 1,219 mRNAs, 2,093 lncRNAs and 57 miRNAs were identified to differently express in AAA.

The ceRNA hypothesis was first proposed by Salmena et al (18) in 2011. This hypothesis holds that lncRNAs act as miRNA ‘sponges’ to promote the expression of target genes of miRNAs, which provided a novel basis for exploring the molecular functions of lncRNAs. Emerging studies have reported that ceRNAs act as important regulators in different types of disease, including cancer, cardiac fibrosis and rheumatoid arthritis (20). For instance, Karreth (29) reported that the B-Raf pseudogene functions as a competitive endogenous RNA and induces lymphoma in vivo. In the present study, deregulated lncRNA-miRNA-mRNA networks in AAA were constructed. The results indicated that several key lncRNAs served important roles in regulating AAA progression by regulating a series of miRNAs and mRNAs. To the best of our knowledge, the present study was the first to screen ceRNA networks in AAA.

To date, only few studies have focused on exploring the roles of lncRNAs in AAA. In the present study, hub lncRNAs with critical roles in biological networks were identified. The upregulated lncRNAs NEAT1, CDKN2B-AS1, SCARNA10, AC005224.4 and SENP3-EIF4A1, and the downregulated ZNRD1-AS1 were identified as key lncRNAs in the ceRNA networks and interacted with >5 different miRNAs. Furthermore, key lncRNA-mRNA-biological processes analysis revealed that NEAT1, CDKN2B-AS1, SCARNA10, AC005224.4 and SENP3-EIF4A1 were involved in regulating signal transduction, protein amino acid phosphorylation and immune response, while ZNRD1-AS1 was associated with regulation of transcription, development and cell differentiation. Of these key lncRNAs, NEAT1 was reported to be overexpressed in different types of solid tumor, including lung cancer, oesophageal cancer, colorectal cancer and hepatocellular carcinoma (3033). CDKN2B-AS1 was reported to interact with protein regulator of cytokinesis 1 and −2, leading to epigenetic silencing of CDKN2B (34). CDKN2B-AS1 has key roles in the progression of several cancer types (3537), intracranial aneurysm (38), type-2 diabetes (39,40), periodontitis (41,42), Alzheimer's disease (43,44), endometriosis (45), frailty in the elderly (46) and glaucoma (36,47).

Several limitations of the present study should be noted. First, further validation of key lncRNA expression levels in AAA samples is required. Although the datasets used in the present study have been reported and validated, further study to confirm the differences in lncRNA expression between AAA and normal samples is required. Another limitation is that the public datasets for analyzing AAA-associated lncRNAs were limited. An urgent requirement remains to identify differently expressed lncRNAs in AAA by using high-throughput methods, including lncRNA expression array and RNA sequencing. Finally, additional functional investigations of these lncRNAs on AAA progression are still required.

Based on the ceRNA hypothesis, the present study was the first to constructed lncRNA-miRNA-mRNA networks in AAA, to the best of our knowledge. In the present study, a total of 1,219 mRNAs, 2,093 lncRNAs and 57 miRNAs were identified to be differently expressed in AAA based on analyzing public datasets including GSE7084, the dataset by Yang et al (9) and GSE24194. The present study further identified 5 upregulated lncRNAs (NEAT1, CDKN2B-AS1, SCARNA10, AC005224.4 and SENP3-EIF4A1) and the downregulated ZNRD1-AS1 as key lncRNAs in the ceRNA networks. Key lncRNA-mRNA-biological processes analysis indicated that these key lncRNAs are involved in regulating signal transduction, protein amino acid phosphorylation, immune response, transcription, development and cell differentiation. The present study provides novel clues for exploring the mechanistic involvement of lncRNA in AAA progression.

Acknowledgements

Not applicable.

Funding

No funding received.

Availability of data and materials

High-resolution versions of Figs. 3 and 4 are available on request. The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request

Authors' contributions

HZ, LT and DL contributed to the conception and design of the study. LT and XH developed the methodology. YH collected the samples. ZW analyzed and interpreted the data. LT, DL and HZ wrote, reviewed and revised the manuscript. The final version of the manuscript has been read and approved by all authors.

Ethical approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Alcorn HG, Wolfson SK Jr, Sutton-Tyrrell K, Kuller LH and O'Leary D: Risk factors for abdominal aortic aneurysms in older adults enrolled in the cardiovascular health study. Arterioscler Thromb Vasc Biol. 16:963–970. 1996. View Article : Google Scholar : PubMed/NCBI

2 

Humphrey JD and Taylor CA: Intracranial and abdominal aortic aneurysms: Similarities, differences, and need for a new class of computational models. Annu Rev Biomed Eng. 10:221–246. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Hallett JW Jr, Marshall DM, Petterson TM, Gray DT, Bower TC, Cherry KJ Jr, Gloviczki P and Pairolero PC: Graft-related complications after abdominal aortic aneurysm repair: Reassurance from a 36-year population-based experience. J Vasc Surg. 25:277–284; discussion 285–276. 1997. View Article : Google Scholar : PubMed/NCBI

4 

Johansson G and Swedenborg J: Ruptured abdominal aortic aneurysms: A study of incidence and mortality. Br J Surg. 73:101–103. 1986. View Article : Google Scholar : PubMed/NCBI

5 

Maegdefessel L, Azuma J, Toh R, Deng A, Merk DR, Raiesdana A, Leeper NJ, Raaz U, Schoelmerich AM, McConnell MV, et al: MicroRNA-21 blocks abdominal aortic aneurysm development and nicotine-augmented expansion. Sci Transl Med. 4:122ra1222012. View Article : Google Scholar

6 

Miyake T and Morishita R: Pharmacological treatment of abdominal aortic aneurysm. Cardiovasc Res. 83:436–443. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Huarte M: The emerging role of lncRNAs in cancer. Nat Med. 21:1253–1261. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Duggirala A, Delogu F, Angelini TG, Smith T, Caputo M, Rajakaruna C and Emanueli C: Non coding RNAs in aortic aneurysmal disease. Front Genet. 6:1252015. View Article : Google Scholar : PubMed/NCBI

9 

Yang YG, Li MX, Kou L, Zhou Y, Qin YW, Liu XJ and Chen Z: Long noncoding RNA expression signatures of abdominal aortic aneurysm revealed by microarray. Biomed Environ Sci. 29:713–723. 2016.PubMed/NCBI

10 

Geisler S and Coller J: RNA in unexpected places: Long non-coding RNA functions in diverse cellular contexts. Nat Rev Mol Cell Biol. 14:699–712. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Gibney ER and Nolan CM: Epigenetics and gene expression. Heredity (Edinb). 105:4–13. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Wilusz JE, Sunwoo H and Spector DL: Long noncoding RNAs: Functional surprises from the RNA world. Genes Dev. 23:1494–1504. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Wan X, Huang W, Yang S, Zhang Y, Pu H, Fu F, Huang Y, Wu H, Li T and Li Y: Identifcation of androgen-responsive lncRNAs as diagnostic and prognostic markers for prostate cancer. Oncotarget. 7:60503–60518. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Guo LL, Song CH, Wang P, Dai LP, Zhang JY and Wang KJ: Competing endogenous RNA networks and gastric cancer. World J Gastroenterol. 21:11680–11687. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Tay Y, Rinn J and Pandolfi PP: The multilayered complexity of ceRNA crosstalk and competition. Nature. 505:344–352. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Franco-Zorrilla JM, Valli A, Todesco M, Mateos I, Puga MI, Rubio-Somoza I, Leyva A, Weigel D, García JA and Paz-Ares J: Target mimicry provides a new mechanism for regulation of microRNA activity. Nat Genet. 39:1033–1037. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ and Pandolfi PP: A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature. 465:1033–1038. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Salmena L, Poliseno L, Tay Y, Kats L and Pandolfi PP: A ceRNA hypothesis: The rosetta stone of a hidden RNA language? Cell. 146:353–358. 2011. View Article : Google Scholar : PubMed/NCBI

19 

van Rooij E and Olson EN: MicroRNA therapeutics for cardiovascular disease: opportunities and obstacles. Nat Rev Drug Discov. 11:860–872. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Chen Y, Li C, Tan C and Liu X: Circular RNAs: A new frontier in the study of human diseases. J Med Genet. 53:359–365. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Lenk GM, Tromp G, Weinsheimer S, Gatalica Z, Berguer R and Kuivaniemi H: Whole genome expression profiling reveals a significant role for immune function in human abdominal aortic aneurysms. BMC Genomics. 8:2372007. View Article : Google Scholar : PubMed/NCBI

22 

Liu G, Huang Y, Lu X, Lu M, Huang X, Li W and Jiang M: Identification and characteristics of microRNAs with altered expression patterns in a rat model of abdominal aortic aneurysms. Tohoku J Exp Med. 222:187–193. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Cui X and Churchill GA: Statistical tests for differential expression in cDNA microarray experiments. Genome Biol. 4:2102003. View Article : Google Scholar : PubMed/NCBI

24 

Diboun I, Wernisch L, Orengo CA and Koltzenburg M: Microarray analysis after RNA amplification can detect pronounced differences in gene expression using limma. BMC Genomics. 7:2522006. View Article : Google Scholar : PubMed/NCBI

25 

Gondro C, Porto-Neto LR and Lee SH: R for genome-wide association studies. Methods Mol Biol. 1019:1–17. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Jeggari A, Marks DS and Larsson E: miRcode: A map of putative microRNA target sites in the long non-coding transcriptome. Bioinformatics. 28:2062–2063. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Li JH, Liu S, Zhou H, Qu LH and Yang JH: starBase v2.0: Decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 42:D92–D97. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Moran CS, McCann M, Karan M, Norman P, Ketheesan N and Golledge J: Association of osteoprotegerin with human abdominal aortic aneurysm progression. Circulation. 111:3119–3125. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Karreth FA, Reschke M, Ruocco A, Ng C, Chapuy B, Léopold V, Sjoberg M, Keane TM, Verma A, Ala U, et al: The BRAF pseudogene functions as a competitive endogenous RNA and induces lymphoma in vivo. Cell. 161:319–332. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Guo S, Chen W, Luo Y, Ren F, Zhong T, Rong M, Dang Y, Feng Z and Chen G: Clinical implication of long non-coding RNA NEAT1 expression in hepatocellular carcinoma patients. Int J Clin Exp Pathol. 8:5395–5402. 2015.PubMed/NCBI

31 

Clemson CM, Hutchinson JN, Sara SA, Ensminger AW, Fox AH, Chess A and Lawrence JB: An architectural role for a nuclear noncoding RNA: NEAT1 RNA is essential for the structure of paraspeckles. Mol Cell. 33:717–726. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Sun C, Li S, Zhang F, Xi Y, Wang L, Wang L and Li D: Long non-coding RNA NEAT1 promotes non-small cell lung cancer progression through regulation of miR-377-3p-E2F3 pathway. Oncotarget. 7:51784–51814. 2016.PubMed/NCBI

33 

Chen X, Kong J, Ma Z, Gao S and Feng X: Up regulation of the long non-coding RNA NEAT1 promotes esophageal squamous cell carcinoma cell progression and correlates with poor prognosis. Am J Cancer Res. 5:2808–2815. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Kotake Y, Nakagawa T, Kitagawa K, Suzuki S, Liu N, Kitagawa M and Xiong Y: Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene. 30:1956–1962. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Yang XR, Liang XY, Pfeiffer RM, Wheeler W, Maeder D, Burdette L, Yeager M, Chanock S, Tucker MA and Goldstein AM: Associations of 9p21 variants with cutaneous malignant melanoma, nevi, and pigmentation phenotypes in melanoma-prone families with and without CDKN2A mutations. Fam Cancer. 9:625–633. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Falchi M, Bataille V, Hayward NK, Duffy DL, Bishop JA, Pastinen T, Cervino A, Zhao ZZ, Deloukas P, Soranzo N, et al: Genome-wide association study identifies variants at 9p21 and 22q13 associated with development of cutaneous nevi. Nat Genet. 41:915–919. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Sherborne AL, Hosking FJ, Prasad RB, Kumar R, Koehler R, Vijayakrishnan J, Papaemmanuil E, Bartram CR, Stanulla M, Schrappe M, et al: Variation in CDKN2A at 9p21.3 influences childhood acute lymphoblastic leukemia risk. Nat Genet. 42:492–494. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Helgadottir A, Thorleifsson G, Magnusson KP, Grétarsdottir S, Steinthorsdottir V, Manolescu A, Jones GT, Rinkel GJ, Blankensteijn JD, Ronkainen A, et al: The same sequence variant on 9p21 associates with myocardial infarction, abdominal aortic aneurysm and intracranial aneurysm. Nat Genet. 40:217–224. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Broadbent HM, Peden JF, Lorkowski S, Goel A, Ongen H, Green F, Clarke R, Collins R, Franzosi MG, Tognoni G, et al: Susceptibility to coronary artery disease and diabetes is encoded by distinct, tightly linked SNPs in the ANRIL locus on chromosome 9p. Hum Mol Genet. 17:806–814. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Cugino D, Gianfagna F, Santimone I, de Gaetano G, Donati MB, Iacoviello L and Di Castelnuovo A: Type 2 diabetes and polymorphisms on chromosome 9p21: A meta-analysis. Nutr Metab Cardiovasc Dis. 22:619–625. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Schaefer AS, Richter GM, Groessner-Schreiber B, Noack B, Nothnagel M, El Mokhtari NE, Loos BG, Jepsen S and Schreiber S: Identification of a shared genetic susceptibility locus for coronary heart disease and periodontitis. Plos Genet. 5:e10003782009. View Article : Google Scholar : PubMed/NCBI

42 

Ernst FD, Uhr K, Teumer A, Fanghänel J, Schulz S, Noack B, Gonzales J, Reichert S, Eickholz P, Holtfreter B, et al: Replication of the association of chromosomal region 9p21.3 with generalized aggressive periodontitis (gAgP) using an independent case-control cohort. BMC Med Genet. 11:1192010. View Article : Google Scholar : PubMed/NCBI

43 

Yu JT, Yu Y, Zhang W, Wu ZC, Li Y, Zhang N and Tan L: Single nucleotide polymorphism rs1333049 on chromosome 9p21.3 is associated with alzheimer's disease in han chinese. Clin Chim Acta. 411:1204–1207. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Emanuele E, Lista S, Ghidoni R, Binetti G, Cereda C, Benussi L, Maletta R, Bruni AC and Politi P: Chromosome 9p21.3 genotype is associated with vascular dementia and Alzheimer's disease. Neurobiol Aging. 32:1231–1235. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Uno S, Zembutsu H, Hirasawa A, Takahashi A, Kubo M, Akahane T, Aoki D, Kamatani N, Hirata K and Nakamura Y: A genome-wide association study identifies genetic variants in the CDKN2BAS locus associated with endometriosis in Japanese. Nat Genet. 42:707–710. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Melzer D, Frayling TM, Murray A, Hurst AJ, Harries LW, Song H, Khaw K, Luben R, Surtees PG, Bandinelli SS, et al: A common variant of the p16 INK4a genetic region is associated with physical function in older people. Mech Ageing Dev. 128:370–377. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Ramdas WD, van Koolwijk LME, Lemij HG, Pasutto F, Cree AJ, Thorleifsson G, Janssen SF, Jacoline TB, Amin N, Rivadeneira F, et al: Common genetic variants associated with open-angle glaucoma. Hum Mol Genet. 20:2464–2471. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 16 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tian L, Hu X, He Y, Wu Z, Li D and Zhang H: Construction of lncRNA‑miRNA‑mRNA networks reveals functional lncRNAs in abdominal aortic aneurysm. Exp Ther Med 16: 3978-3986, 2018
APA
Tian, L., Hu, X., He, Y., Wu, Z., Li, D., & Zhang, H. (2018). Construction of lncRNA‑miRNA‑mRNA networks reveals functional lncRNAs in abdominal aortic aneurysm. Experimental and Therapeutic Medicine, 16, 3978-3986. https://doi.org/10.3892/etm.2018.6690
MLA
Tian, L., Hu, X., He, Y., Wu, Z., Li, D., Zhang, H."Construction of lncRNA‑miRNA‑mRNA networks reveals functional lncRNAs in abdominal aortic aneurysm". Experimental and Therapeutic Medicine 16.5 (2018): 3978-3986.
Chicago
Tian, L., Hu, X., He, Y., Wu, Z., Li, D., Zhang, H."Construction of lncRNA‑miRNA‑mRNA networks reveals functional lncRNAs in abdominal aortic aneurysm". Experimental and Therapeutic Medicine 16, no. 5 (2018): 3978-3986. https://doi.org/10.3892/etm.2018.6690