Open Access

Identification of key genes associated with the human abdominal aortic aneurysm based on the gene expression profile

  • Authors:
    • Xudong Chen
    • Chengfei Zheng
    • Yunjun He
    • Lu Tian
    • Jianhui Li
    • Donglin Li
    • Wei Jin
    • Ming Li
    • Shusen Zheng
  • View Affiliations

  • Published online on: October 15, 2015     https://doi.org/10.3892/mmr.2015.4448
  • Pages: 7891-7898
  • Copyright: © Chen 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

The present study was aimed at screening the key genes associated with abdominal aortic aneurysm (AAA) in the neck, and to investigate the molecular mechanism underlying the development of AAA. The gene expression profile, GSE47472, including 14 AAA neck samples and eight donor controls, was downloaded from the Gene Expression Omnibus database. The total AAA samples were grouped into two types to avoid bias. Differentially expressed genes (DEGs) were screened in patients with AAA and subsequently compared with donor controls using linear models for microarray data, or the Limma package in R, followed by gene ontology enrichment analysis. Furthermore, a protein‑protein interaction (PPI) network based on the DEGs was constructed to detect highly connected regions using a Cytoscape plugin. In total, 388 DEGs in the AAA samples were identified. These DEGs were predominantly associated with limb development, including embryonic limb development and appendage development. Nuclear receptor co‑repressor 1 (NCOR1), histone 4 (H4), E2F transcription factor 4 (E2F4) and hepatocyte nuclear factor 4α (HNF4A) were the four transcription factors associated with AAA. Furthermore, HNF4A indirectly interacted with the other three transcription factors. Additionally, six clusters were selected from the PPI network. The DEG screening process and the construction of an interaction network enabled an understanding of the mechanism of AAA to be gleaned. HNF4A may exert an important role in AAA development through its interactions with the three other transcription factors (E2F4, NCOR1 and H4), and the mechanism of this coordinated regulation of the transcription factors in AAA may provide a suitable target for the development of therapeutic intervention strategies.

Introduction

Abdominal aortic aneurysm (AAA) is characterized by permanent, localized dilations of the abdominal aorta, which are defined as having diameters 1.5 times greater than normal (or which measure >3 cm) (1). Aortic rupture is the most serious clinical condition resulting from the progression of AAA (2). Almost 80% of patients who experience aortic rupture succumb to mortality (3). Since the majority of aneurysms are usually asymptomatic until rupture occurs, diagnosis is therefore problematic, and no preventative therapies are currently available for patients to effectively limit the progression of AAA (2,4). Therefore, it is important to investigate the mechanisms of AAA initiation and progression in order to assist diagnostic applications and to develop therapeutic options.

AAA is considered to be a particular, localized form of atherothrombosis (5). It shares the usual risk factors with occlusive atherothrombosis, including an increasing age, male gender, smoking, possible genetic susceptibility and low high density lipoprotein-to-cholesterol levels (6). The pathogenesis of AAA is complicated and multifactorial. Unique hemodynamic forces, which particularly impact on the infrarenal area, and variations in the content of elastin and collagen in different parts of the aorta rendered the infrarenal part of the abdominal aorta highly susceptible to AAA development (2). Numerous animal models and clinical studies reported that the initiation of AAA involves an inflammatory response, which is often enforced by the upregulation of adhesion molecules (79). The degradation of the extracellular matrix, which is caused by an increased activity of matrix metalloproteinases (MMPs) and serine proteases, and smooth muscle cell apoptosis are the predominant features associated with the progression of AAA (10,11). Previous-genome wide studies using microarrays have investigated the pathogenesis of AAA (1214). The involvement of the immune system in AAA formation and progression was also reported in previous studies (15,16), however, the molecular mechanisms leading to the development and progression of AAA remain to be fully elucidated.

Using the identical gene expression profile, Biros et al (17) demonstrated that immune pathways are upregulated within the undilated aorta proximal to an AAA. In the present study, the differentially expressed genes (DEGs) featured in the gene expression profile in AAA necks were analyzed. Furthermore, a function and pathway-enrichment analysis was performed on the DEGs, and a protein-protein interaction network (PPI) was constructed to identify those DEGs which have a central role in AAA. The present study also aimed to gain further insights into the molecular mechanisms underlying the development of AAA. Understanding these molecular mechanisms may assist in developing the understanding of the pathogenesis of AAA, and to translate these pathogenic activities into therapeutic applications.

Materials and methods

Microarray data and data pre-processing

The gene expression profile of GSE47472 was downloaded from the Gene Expression Omnibus (GEO) (17) in the National Centre for Biotechnology Information (http://www.ncbi.nlm.nih.gov/geo/) based on the platform of GPL10558 (or the Illumina HumanHT-12 V4.0 expression beadchip). A total of 22 data biopsies were obtained from the AAA neck samples, comprising 14 AAA samples from patients undergoing open AAA repair and eight normal samples from beating heart organ donors following brain mortality. The original data were pre-processed using the beadarray package in R language (version 2.18.0; http://bioconductor.org/packages/release/bioc/html/beadarray.html) (18), and normalized using the quantile method (19). Boxplots of the raw and normalized data were produced.

Screening of DEGs

Multi-dimensional scaling (MDS), which was constructed with the poltMDS (20) function in the linear models for microarray data (Limma) (21) package (version 3.24.15; http://www.bioconductor.org/packages/release/bioc/html/limma.html) was used to investigate the association of the samples as a measure of quality control. From the results of the MDS procedure, the AAA samples were separated into types A and B. The DEGs in the AAA sample types A and B were identified using the Limma package and were compared with the controls. The common DEGs, which featured consistent changes in their expression levels, were selected as the targets for further analysis. The false discovery rate (FDR) was calculated for multiple testing correction using the Benjamini and Hochberg method (22). The threshold for the DEGs was set as the log fold change (FC)>1 and FDR≤0.01. Pearson's correlation coefficient was used to examine the associations between these DEGs (23).

Enrichment analysis of the DEGs

The probe sets, which featured differential expression between the controls and the AAA samples, were annotated to Ensembl gene identifiers (IDs) for ID mapping using the database for annotation, visualization and integrated discovery (DAVID) tool (version 6.7; http://david.abcc.Ncifcrf.gov/ (24,25). Gene ontology (GO; http://www.geneontology.org/) (26) and Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.genome.jp/kegg/pathway.html) analyses were performed on the selected lists of genes. The threshold was set as P≤0.05.

Constructing an interaction network and functional analysis

Following ID mapping, all selected genes were exported into Cytoscape plugin (27) using the BisoGenet module (28) to create network visualizations. The source of the interaction network database was the Biomolecular Interaction Network Database (BIND) (29). Subsequently, a cluster analysis on the resulting network was performed with the Plugin, ClusterONE (http://apps.cytoscape.org/apps/clusterone) (30) program, using a P<0.05 as a cut-off. The significant GO categories of the DEGs in the subnetworks were analyzed using the DAVID tool.

Results

Data pre-processing

The raw data downloaded from the GEO databases were normalized (Fig. 1A). The median values of each sample were almost at the identical level, suggesting that the data were eligible for further analysis. An MDS plot was constructed as a means of visualizing the data. This was performed for all the probes available. The AAA samples were observed to cluster into two groups, types A and B (Fig. 1B).

Screening of the DEGs

Comparing the AAA type A samples with the donor controls yielded the identification of 1,584 DEGs (Fig. 1C). Comparing the AAA type B samples with the donor controls yielded 984 DEGs in total (Fig. 1D). A total of 340 genes were identified as being DEGs common to each group, as revealed by the Venn diagram (Fig. 2A), and therefore, 338 DEGs exhibited a consistent change. An assessment of the correlation of the 338 DEGs revealed a positive pattern of correlation, with a Pearson's correlation coefficient (r) of 0.974 (P<2.2−16). The top three upregulated and downregulated DEGs, with the highest logFC, are listed in Table I. Subsequently, the expression pattern of the DEGs was determined. From the heat map shown in Fig. 2, it was observed that the expression pattern of the DEGs enabled the AAA samples to be distinguished from the donor control samples. Furthermore, the AAA sample types A and B were consistent.

Table I

Differentially expressed genes in Type A and Type B.

Table I

Differentially expressed genes in Type A and Type B.

Probe IDExpression changeGene symbolType A
Type B
logFCadj.P.VallogFCadj.P.Val
ILMN_1899549UpregulatedNA3.3697510.0089943.1120590.001856
ILMN_1717168UpregulatedPCDHGA42.753187 1.33e−102.225494 5.06e−11
ILMN_2177965UpregulatedRPS19BP12.678406 4.24e−82.365836 6.39e−9
ILMN_2231051DownregulatedTCP11L2−3.164650 6.34e−8−1.0032800.001986
ILMN_1787591Downregulatedxpa−3.391970 8.9e−8−1.1629700.001270
ILMN_1717733DownregulatedNA−4.015280 3.99e−11−2.217670 5.8e−9

[i] Positive values for logFC represent upregulated and negative values represent downregulated genes, ID, identifier; FC, fold change, adj.P.Val, adjusted P-value.

GO and KEGG enrichment analyses

The results of the GO and KEGG enrichment analyses of the common DEGs using the DAVID tool are presented in Fig. 3. A total of 18 significantly enriched categories were identified, which comprised 15 biological processes, two molecular functions and one KEGG pathway, grouped and annotated manually. The DEGs were predominantly associated with limb development, including embryonic limb morphogenesis and appendage development. The tyrosine kinase signaling pathway was also significantly enriched.

Interaction network and functional analysis

The 338 probe sets, which were retained, were mapped, in total, to 297 gene symbols. Interactions between the genes were searched for in BIND and the results are presented in Fig. 4. The transcriptional factors nuclear receptor corepressor 1 (NCOR1), histone 4 (H4), E2F transcription factor 4 (E2F4) and hepatocyte nuclear factor 4α (HNF4A) were identified in the network to regulate other DEGs. Six noteworthy clusters emerged from the clustering analysis (Table II) and the subnetwork diagram is presented in Fig. 5.

Table II

Noteworthy clusters in the network.

Table II

Noteworthy clusters in the network.

ClusterGO IDNodesDensityQualityP-valueDescriptionGenes in cluster
1004356560.7330.9170.003Sequence-specific DNA bindingBACH2, JUN, MAKF, MAFG, ATF7, MAFB
2004566540.66710.011Negative regulation of neuron differentiationPBX1, PAX6, IPO13, HXB
3003114340.66710.011PseudopodiumACTN2, MYOZ2, MYOZ1, LDB3
4000724230.66710.026Intracellular signaling cascadeNDKB, ITGB1BP1, KRIT1
5003011730.66710.026Membrane coatAP3D1, AP3S2, AP3S1
6000175930.66710.026Induction of an organFGR1, FGF1, FGF2

[i] GO, gene ontology; ID, identifier.

Discussion

AAA is a complex disease, of which the pathobiology remains to be fully elucidated (2). Previous studies have revealed that the disease has a marked genetic component (12,14,31). Assessing gene expression profiling in cases of disease may reveal the underlying changes in gene activity, which contribute to the disease and enable targets for therapeutic intervention to be identified. In the present study, the gene expression profile downloaded from the GEO database was used to examine the mechanism of AAA development. A total of 340 genes were identified to be commonly DEGs in AAA samples of type A compared with donor controls and AAA samples of type B, among which 388 genes exhibited consistent changes. The analysis of the DEGs indicated that they were predominantly involved in limb development. The PPI network analysis revealed that four transcription factors regulated these DEGs in the network, and that NCOR1, H4 and E2F4 were clustered across the network in association with HNF4A. Furthermore, c-jun proto-oncogene (JUN) and its downstream components, which are regulated upstream in the pathway by HNF4A, were enriched in cluster 1.

NCOR1, which mediates transcriptional repression by certain nuclear receptors, is a part of a complex, which promotes histone deacetylation and the formation of repressive chromatin structures, and which may impede the access of basal transcription factors (32). MMPs exert a significant role in the degradation of the extracellular matrix of the vessel wall, which gradually leads to the formation of AAA (33). Mannello et al (34) demonstrated that MMP-3 may degrade NCOR1 to prevent transcriptional repression of the connective tissue growth factor promoter. In line with previous studies, NCOR1 may exert a critical role in the development of AAA, based on the PPI network analysis.

H4 is a core histone of the nucleosome, which functions in nucleosomal wrapping and occupies a central role in transcriptional regulation, DNA repair, DNA replication and chromosomal stability (35). Santos-Rosa et al (36) reported that the histone code set up by post-translational histone modifications also respond to DNA damage, which is frequently observed in cancer cells. HNF4A is a transcriptionally controlled transcription factor, and it may be essential for the development of the kidney, liver and intestine (37,38). A previous study revealed that phospholipase A2 g10-deficent mice were protected from angiotensin-II-induced aortic aneurysms (39). Guan et al (40) demonstrated that the transcription levels of Pla2g12b were regulated by the transcription factor HNF4A and its co-activator. Furthermore, Soutoglou et al (41) reported that HNF4A may interact with histone acetyltransferases, a process which is dependent on the acetylation status of the histones. In this context, and based on the results of the present study, it was postulated that there may be an interaction between the expression of H4 and HNF4A in the development of AAA.

E2F4 is a transcriptional activator, which binds DNA co-operatively with differentially regulated transcription factor proteins, whose products are involved in DNA replication and in cell cycle regulation (42). The transcription factor DRTF1/E2F complex functions in the control of cell cycle progression from G1 to S phase (43). Tung et al (44) identified genes, which are involved in cell cycle regulation in AAA, using a membrane-based complementary DNA expression array. Talianidis et al (45) demonstrated that complex interactions between HNF4 bound to the proximal promoter and SP1 bound to multiple distal regulatory sites may lead to transcriptional activation of liver-specific human apolipoprotein CIII gene. SP1 is a cellular transcription factor involved in a wide variety of processes, and it has been determined to bind to different promoters to regulate transcription (46). In addition, Price et al (47) determined that differences in allelic expression of the MMP-2 gene, which exerts a major role in AAA formation, were attributed to the elimination of SP1 binding. Furthermore, the expression levels of HNF4A as a hepatocyte marker for hepatic differentiation were increased in the early G1 phase in human embryonic stem cells during their cell cycle (48). Therefore, an important interaction may occur between HNF4A and E2F4 in the pathogenesis of AAA.

A previous study revealed that HNF4A may be a key upstream mediator of JUN, whereas JUN was intimately associated with BTB and CNC homology 1, basic leucine zipper transcription factor 2 (BACH2), which is involved in the lymphocyte signaling pathway in cluster 1 (49). Henderson et al (50) demonstrated the death of smooth muscle cells and the expression of mediators of apoptosis by T lymphocytes in human AAA. A previous study revealed that the inhibition of c-Jun N-terminal kinase caused regression of AAA. In this context, it is surmised that JUN may interact with BACH2 in the progression of AAA.

In conclusion, genes which are critical in the development of AAA have been assessed based on the microarray data. HNF4A exerts an important role in the development of AAA through the interactions made with three other transcription factors (E2F4, NCOR1 and H4), and the coordinated regulation of these transcription factors may represent potential novel targets for the mechanism underlying the development of AAA. The present study offers novel insights into the pathobiology of AAA. Further studies are required to confirm these intriguing results in terms of the possible associations with the transcriptional factors.

Abbreviations:

AAA

abdominal aortic aneurysm

MMP

matrix metalloproteinase

DEG

differentially expressed gene

GEO

gene expression omnibus

MDS

multi-dimensional scaling

FDR

false discovery rate

FC

fold change

References

1 

Golledge J, Muller J, Daugherty A and Norman P: Abdominal aortic aneurysm: Pathogenesis and implications for management. Arterioscler Thromb Vasc Biol. 26:2605–2613. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Maegdefessel L, Dalman RL and Tsao PS: Pathogenesis of abdominal aortic aneurysms: MicroRNAs, proteases, genetic associations. Annu Rev Med. 65:49–62. 2014. View Article : Google Scholar

3 

Verhoeven EL, Kapma MR, Groen H, Tielliu IF, Zeebregts CJ, Bekkema F and van den Dungen JJ: Mortality of ruptured abdominal aortic aneurysm treated with open or endovascular repair. J Vasc Surg. 48:1396–1400. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Golledge J and Norman PE: Current status of medical management for abdominal aortic aneurysm. Atherosclerosis. 217:57–63. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Sakalihasan N, Limet R and Defawe O: Abdominal aortic aneurysm. The Lancet. 365:1577–1589. 2005. View Article : Google Scholar

6 

Michel JB, Martin-Ventura JL, Egido J, Sakalihasan N, Treska V, Lindholt J, Allaire E, Thorsteinsdottir U, Cockerill G and Swedenborg J; FAD EU Consortium: Novel aspects of the pathogenesis of aneurysms of the abdominal aorta in humans. Cardiovasc Res. 90:18–27. 2011. View Article : Google Scholar :

7 

Thomas M, Gavrila D, McCormick ML, Miller FJ Jr, Daugherty A, Cassis LA, Dellsperger KC and Weintraub NL: Deletion of p47phox attenuates angiotensin II-induced abdominal aortic aneurysm formation in apolipoprotein E-deficient mice. Circulation. 114:404–413. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Sharma AK, Lu G, Jester A, Johnston WF, Zhao Y, Hajzus VA, Saadatzadeh MR, Su G, Bhamidipati CM, Mehta GS, et al: Experimental abdominal aortic aneurysm formation is mediated by IL-17 and attenuated by mesenchymal stem cell treatment. Circulation. 126(Suppl 1): S38–S45. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Harrison SC, Smith AJ, Jones GT, Swerdlow DI, Rampuri R, Bown MJ; Aneurysm Consortium; Folkersen L, Baas AF, de Borst GJ, et al: Interleukin-6 receptor pathways in abdominal aortic aneurysm. Eur Heart J. 34:3707–3716. 2013. View Article : Google Scholar :

10 

Kunieda T, Minamino T, Nishi J, Tateno K, Oyama T, Katsuno T, Miyauchi H, Orimo M, Okada S, Takamura M, et al: Angiotensin II induces premature senescence of vascular smooth muscle cells and accelerates the development of atherosclerosis via a p-21 dependent pathway. Circulation. 114:953–960. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Manning MW, Cassis LA and Daugherty A: Differential effects of doxycycline, a broad-spectrum matrix metalloproteinase inhibitor, on angiotensin II-induced atherosclerosis and abdominal aortic aneurysms. Arterioscler Thromb Vasc Biol. 23:483–488. 2003. View Article : Google Scholar

12 

Hinterseher I, Tromp G and Kuivaniemi H: Genes and abdominal aortic aneurysm. Ann Vasc Surg. 25:388–412. 2011. View Article : Google Scholar :

13 

Hinterseher I, Erdman R, Elmore JR, Stahl E, Pahl MC, Derr K, Golden A, Lillvis JH, Cindric MC, Jackson K, et al: Novel pathways in the pathobiology of human abdominal aortic aneurysms. Pathobiology. 80:1–10. 2013. View Article : Google Scholar :

14 

Golledge J and Kuivaniemi H: Genetics of abdominal aortic aneurysm. Curr Opin Cardiol. 28:290–296. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Duftner C, Seiler R, Dejaco C, Fraedrich G and Schirmer M: Increasing evidence for immune-mediated processes and new therapeutic approaches in abdominal aortic aneurysms-a review. Ann N Y Acad Sci. 1085:331–338. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Kuivaniemi H, Platsoucas CD and Tilson MD III: Aortic aneurysms: An immune disease with a strong genetic component. Circulation. 117:242–252. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Biros E, Moran CS, Rush CM, Gäbel G, Schreurs C, Lindeman JH, Walker PJ, Nataatmadja M, West M, Holdt LM, et al: Differential gene expression in the proximal neck of human abdominal aortic aneurysm. Atherosclerosis. 233:211–218. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Dunning MJ, Smith ML, Ritchie ME and Tavaré S: Beadarray: R classes and methods for Illumina bead-based data. Bioinformatics. 23:2183–2184. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Bolstad BM: Probe level quantile normalization of high density oligonucleotide array data. Unpublished manuscript. 2001

20 

Choi J: Guide: A desktop application for analysing gene expression data. BMC Genomics. 14:6882013. View Article : Google Scholar : PubMed/NCBI

21 

Smyth GK: Limma: Linear models for microarray data. Bioinformatics and computational biology solutions using R and Bioconductor. Springer; New York: pp. 397–420. 2005, View Article : Google Scholar

22 

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.

23 

Makova KD and Li WH: Divergence in the spatial pattern of gene expression between human duplicate genes. Genome Res. 7:1638–1645. 2003. View Article : Google Scholar

24 

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

25 

Huang da W, Sherman BT and Lempicki RA: Bioinformatics enrichment tools: Paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37:1–13. 2009. View Article : Google Scholar :

26 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology. The gene ontology consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Martin A, Ochagavia ME, Rabasa LC, Miranda J, Fernandez-de-Cossio J and Bringas R: BisoGenet: A new tool for gene network building, visualization and analysis. BMC Bioinformatics. 11:912010. View Article : Google Scholar : PubMed/NCBI

29 

Bader GD, Betel D and Hogue CW: BIND: The biomolecular interaction network database. Nucleic Acids Res. 31:248–250. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Nepusz T, Yu H and Paccanaro A: Detecting overlapping protein complexes in protein protein interaction networks. Nat Methods. 9:471–472. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Estrelinha M, Hinterseher I and Kuivaniemi H: Gene expression studies in human abdominal aortic aneurysm. Rev Vasc Med. 2:77–82. 2014. View Article : Google Scholar

32 

Yoon HG, Chan DW, Reynolds AB, Qin J and Wong J: N-CoR mediates DNA methylation-dependent repression through a methyl CpG binding protein Kaiso. Mol Cell. 12:723–734. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Saratzis A, Abbas AA, Kiskinis D, Melas N, Saratzis N and Kitas GD: Abdominal aortic aneurysm: A review of the genetic basis. Angiology. 62:18–32. 2011. View Article : Google Scholar

34 

Mannello F and Medda V: Nuclear localization of matrix metalloproteinases. Prog Histochem Cytochem. 47:27–58. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Turner BM, Birley AJ and Lavender J: Histone H4 isoforms acetylated at specific lysine residues define individual chromosomes and chromatin domains in Drosophila polytene nuclei. Cell. 69:375–384. 1992. View Article : Google Scholar : PubMed/NCBI

36 

Santos-Rosa H and Caldas C: Chromatin modifier enzymes, the histone code and cancer. Eur J Cancer. 41:2381–2402. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Kanazawa T, Konno A, Hashimoto Y and Kon Y: Hepatocyte nuclear factor 4 alpha is associated with survival of the condensed mesenchyme in the developing mouse kidney. Dev Dyn. 239:1145–1154. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Sandovici I, Smith NH, Nitert MD, Ackers Johnson M, Uribe-Lewis S, Ito Y, Jones RH, Marquez VE, Cairns W, Tadayyon M, et al: Maternal diet and aging alter the epigenetic control of a promoter-enhancer interaction at the Hnf4a gene in rat pancreatic islets. Proc Natl Acad Sci USA. 108:5449–5454. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Zack M, Boyanovsky BB, Shridas P, Bailey W, Forrest K, Howatt DA, Gelb MH, de Beer FC, Daugherty A and Webb NR: Group X secretory phospholipase A(2) augments angiotensin II induced inflammatory responses and abdominal aortic aneurysm formation in apoE-deficient mice. Atherosclerosis. 214:58–64. 2011. View Article : Google Scholar

40 

Guan M, Qu L, Tan W, Chen L and Wong CW: Hepatocyte nuclear factor-4 alpha regulates liver triglyceride metabolism in part through secreted phospholipase A2 GXIIB. Hepatology. 53:458–466. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Soutoglou E, Katrakili N and Talianidis I: Acetylation regulates transcription factor activity at multiple levels. Mol Cell. 5:745–751. 2000. View Article : Google Scholar : PubMed/NCBI

42 

Beijersbergen RL, Kerkhoven RM, Zhu L, Carlée L, Voorhoeve PM and Bernards R: E2F-4, a new member of the E2F gene family, has oncogenic activity and associates with p107 in vivo. Genes Dev. 8:2680–2690. 1994. View Article : Google Scholar : PubMed/NCBI

43 

Zheng N, Fraenkel E, Pabo CO and Pavletich NP: Structural basis of DNA recognition by the heterodimeric cell cycle transcription factor E2F-DP. Genes Dev. 13:666–674. 1999. View Article : Google Scholar : PubMed/NCBI

44 

Tung WS, Lee JK and Thompson RW: Simultaneous analysis of 1176 gene products in normal human aorta and abdominal aortic aneurysms using a membrane-based complementary DNA expression array. J Vasc Surg. 34:143–150. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Talianidis I, Tambakaki A, Toursounova J and Zannis VI: Complex interactions between SP1 bound to multiple distal regulatory sites and HNF-4 bound to the proximal promoter lead to transcriptional activation of liver-specific human APOCIII gene. Biochemistry. 34:10298–10309. 1995. View Article : Google Scholar : PubMed/NCBI

46 

Cook T, Gebelein B and Urrutia R: Sp1 and its likes: Biochemical and functional predictions for a growing family of zinc finger transcription factors. Ann N Y Acad Sci. 880:94–102. 1999. View Article : Google Scholar : PubMed/NCBI

47 

Price SJ, Greaves DR and Watkins H: Identification of novel, functional genetic variants in the human matrix metalloproteinase-2 gene: Role of Sp1 in allele-specific transcriptional regulation. J Biol Chem. 276:7549–7558. 2001. View Article : Google Scholar

48 

Pauklin S and Vallier L: The cell cycle state of stem cells determines cell fate propensity. Cell. 155:135–147. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Cooper MD and Alder MN: The evolution of adaptive immune systems. Cell. 124:815–822. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Henderson EL, Geng YJ, Sukhova GK, Whittemore AD, Knox J and Libby P: Death of smooth muscle cells and expression of mediators of apoptosis by T lymphocytes in human abdominal aortic aneurysms. Circulation. 99:96–104. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2015
Volume 12 Issue 6

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
Chen X, Zheng C, He Y, Tian L, Li J, Li D, Jin W, Li M and Zheng S: Identification of key genes associated with the human abdominal aortic aneurysm based on the gene expression profile. Mol Med Rep 12: 7891-7898, 2015
APA
Chen, X., Zheng, C., He, Y., Tian, L., Li, J., Li, D. ... Zheng, S. (2015). Identification of key genes associated with the human abdominal aortic aneurysm based on the gene expression profile. Molecular Medicine Reports, 12, 7891-7898. https://doi.org/10.3892/mmr.2015.4448
MLA
Chen, X., Zheng, C., He, Y., Tian, L., Li, J., Li, D., Jin, W., Li, M., Zheng, S."Identification of key genes associated with the human abdominal aortic aneurysm based on the gene expression profile". Molecular Medicine Reports 12.6 (2015): 7891-7898.
Chicago
Chen, X., Zheng, C., He, Y., Tian, L., Li, J., Li, D., Jin, W., Li, M., Zheng, S."Identification of key genes associated with the human abdominal aortic aneurysm based on the gene expression profile". Molecular Medicine Reports 12, no. 6 (2015): 7891-7898. https://doi.org/10.3892/mmr.2015.4448