Open Access

Prediction of marker genes associated with hypertension by bioinformatics analyses

  • Authors:
    • Yuan Gao
    • Guo-Xian Qi
    • Zhi-Mei Jia
    • Ying-Xian Sun
  • View Affiliations

  • Published online on: May 25, 2017     https://doi.org/10.3892/ijmm.2017.3000
  • Pages: 137-145
  • Copyright: © Gao 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

This study aimed to explore the underlying marker genes associated with hypertension by bioinformatics analyses. A gene expression profile (GSE54015) was downloaded. The differentially expressed genes (DEGs) between the normotensive female (NF) and hypertensive female (HF), and between the normotensive male (NM) and hypertensive male (HM) groups were analyzed. Gene Ontology (GO) and pathway enrichment analyses were performed, followed by protein-protein interaction (PPI) network construction. The transcription factors (TFs), and the common DEGs between the HF and HM groups were then analyzed. In total, 411 DEGs were identified between the HF and NF groups, and 418 DEGs were identified between the HM and NM groups. The upregulated DEGs in the HF and HM groups were enriched in 9 GO terms, including oxidation reduction, such as cytochrome P450, family 4, subfamily b, polypeptide 1 (Cyp4b1) and cytochrome P450, family 4, subfamily a, polypeptide 31 Cyp4a31). The downregulated DEGs were mainly enriched in GO terms related to hormone metabolic processes. In the PPI network, cytochrome P450, family 2, subfamily e, polypeptide 1 (Cyp2e1) had the highest degree in all 3 analysis methods in the HF group. Additionally, 4 TFs were indentified from the 2 groups of data, including sterol regulatory element binding transcription factor 1 (Srebf1), estrogen receptor 1 (Esr1), retinoid X receptor gamma (Rxrg) and peroxisome proliferator-activated receptor gamma (Pparg). The intersection genes were mainly enriched in GO terms related to the extracellular region. On the whole, our data indicate that the DEGs, Cyp4b1, Cyp4a31 and Loxl2, and the TFs, Esr1, Pparg and Rxrg, are associated with the progression of hypertension, and may thus serve as potential therapeutic targets in this disease.

Introduction

Hypertension, also known as arterial hypertension, is a chronic medical condition in which the blood pressure in the arteries is elevated (1). Hypertension is classified as either primary or secondary hypertension, and primary hypertension accounts for approximately 90–95% of cases (2). In primary hypertension, the cause of the hypertensioin is not clearly defined, whereas in secondary hyptertension, the cause of the high blood pressure is clearly determined. Hypertension can lead to cardiac hypertrophy, aortic dissection and heart failure, due to structural changes in blood vessel walls and in the heart muscle, which has become a major burden to healthcare systems (3). Thus, hypertension is the leading risk factor for cardiovascular disease (4). According to a report of the World Health Organization in 2011, cardiovascular diseases are the leading cause of morbidity and mortality worldwide (5).

As a multifactorial disease, hypertension results from a complex interaction of environmental factors and genetic factors (6). Numerous common genetic variants, as well as some rare genetic variants have been identified to affact blood pressure (7,8). For instance, bone morphogenetic protein receptor type 2, which belongs to the transforming growth factor (TGF)-β superfamily of receptors, has been found to be mutated in the majority of cases of hypertension (9). Interleukin (IL)-17 has also been thought to be involved in hypertension due to its role in increasing the production of some other immune system chemical signals, such as IL-1, IL-6 and tumor necrosis factor (TNF)-α (10). Additionally, Austin et al (11) suggested that mutations in caveolin-1 are associated with pulmonary arterial hypertension. Despite advances in understanding the etiology of hypertension, the genetic basis responsible for this disease remains poorly elucidated.

Presently, there are many species of animals used as models of hypertension. In our study, the microarray dataset, GSE54015, which contained data from Schlager BPH/2J and BPN/3J mouse models was downloaded for analysis. of note, Puig et al (12) also used the same mouse models to identify potential pharmacological targets of hypertension. In this study, we identified the differentially expressed genes (DEGs) between normotensive mice and hypertensive mice of both genders. Additionally, we performed functional enrichment analyses and protein-protein interaction (PPI) networks analysis to study and identify the pathways and DEGs associated with hypertension. Furthermore, transcription factors (TFs) and DEGs in both hypertensive female (HF) and male (HM) groups were analyzed. The results of this study may aid in the diagnosis and treatment of hypertension.

Data collection methods

Affymetrix microarray data

The microarray data of GSE54015 were downloaded, which was deposited in the National Center of Biotechnology Information (NCBI) Gene Expression Omnibus (GEO) (http://www.ncbi.nlm.nih.gov/geo/) database by Chiu et al (2). The platform is [MoGene-1_1-st] Affymetrix Mouse Gene 1.1 ST array [transcript (gene) version]. In the study by Chiu et al (2), 12- to 13-week-old Schlager hypertensive male and hypertensive female BPH/2J, and normotensive male and normotensive female BPN/3J mice (n=6/group) were used, and their kidney tissues were collected for the microarrays. In the present study, 4 types of mouse samples were used for analysis: normotensive female (NF) BPN/3J, hypertensive female (HF) BPH/2J, normotensive male (NM) BPN/3J and hypertensive male (HM) BPH/2J mice.

Data pre-processing and analysis of DEGs

The CEL raw data were downloaded and preprocessed using oligo (13) in R of Bioconductor (http://www.bioconductor.org/packages/release/bioc/html/) with the method of robust multiarray average (RMA) (14). The preprocessing processes included background correction, quartile data normalization and probe summarization.

The paired t-test based on the limma package (15) (no. 1406, Ritchie, 2015) in Bioconductor was used to identify the DEGs between the HF and NF groups, and between the HM and NM groups. The false discovery rate (FDR) was calculated to adjust the P-value with the Benjamini-Hochberg method (16). Only genes with |log2FC| ≥0.58 and an adjusted P-value <0.05 were regarded as DEGs.

Gene ontology (GO) and pathway enrichment analyses

The GO (http://www.geneontology.org) project is used to provide consistent descriptors for gene products in different databases, and to standardize classifications for sequences (17). Kyoto Encyclopedia of Genes and Genomes (KEGG) (http://www.genome.ad.jp/kegg/) is a knowledge base used to analyze gene functions, and to link genomic information with higher order functional information (18). In this study, the obtained DEGs were performed GO and KEGG pathway enrichment analyses to obtain the biological process (BP), molecular function (MF), cellular component (CC) terms and pathways. The analysis tool was the Database for Annotation, Visualization and Integrated Discovery (DAVID) (http://david.niaid.nih.gov) (19), which is an integrated biological knowledgebase used for the systematic and integrative analysis of large gene protein lists. The P-value <0.05 and enrichment count ≥2 were considered as thresholds.

PPI network construction

The Search Tool for the Retrieval of Interacting Genes (STRING) (http://string-db.org/) database (20) is a precomputed global resource used for evaluating PPI information. In the present study, the STRING online tool was applied to predict the PPI pairs of DEGs with a combined score of >0.4. The PPI network was constructed using cytoscape (21). Additionally, 3 methods including degree centrality, betweenness centrality and subgraph centrality were used to analyze the hub gene in the PPI network using the CytoNCA cytoscape plugin (http://apps.cytoscape.org/apps/cytonca) (22).

TF analysis

In order to predict the role of TFs in hypertension, we selected the TFs from the DEG corresponding proteins, and predicted the target associations between the TFs and the DEGs based on the TRANSFAC database in the iRegulon cytoscape plugin (http://iregulon.aertslab.org) (23), as previously described. FDR on motif similarity <0.05 was regarded as the threshold.

Analysis of DEGs in the HF and HM groups

In the present study, we screened the common DEGs between the HF and HM groups in order to analyze the similarity of hypertension in females and males. Subsequently, these selected DEGs were subjected to GO functional enrichment analysis using DAVID. Furthermore, the enriched GO terms were subjected to correlation analysis using the enrichmentmap cytoscape plugin (http://baderlab.org/Software/EnrichmentMap/) (24) (P<0.05).

Results

Identification of DEGs

In total, 201 upregulated and 210 downregulated DEGs were identified between the HF and NF groups. Moreover, 204 upregulated and 214 downregulated DEGs were identified between the HM and NM groups. The results are presented in the heatmaps in Fig. 1A and B.

GO and pathway enrichment analyses of DEGs

The upregulated DEGs in the HF group were mainly enriched in GO terms related to ion transport and oxidation reduction, and in the pathway of histidine metabolism. The downregulated DEGs were mainly enriched in the GO terms related to steroid metabolic process and microsome, and in the pathway of arachidonic acid metabolism (Table I).

Table I

Functional enrichment analysis for the DEGs of the HF group.

Table I

Functional enrichment analysis for the DEGs of the HF group.

TermCountP-value
UpBPGO:0006811-ion transport195.68E-05
GO:0019751-polyol metabolic process46.56E-03
GO:0055114-oxidation reduction131.66E-02
CC GO:0005576-extracellular region324.98E-04
GO:0042383-sarcolemma41.17E-02
GO:0005886-plasma membrane411.75E-02
MFGO:0022836-gated channel activity101.00E-03
GO:0005216-ion channel activity104.35E-03
GO:0022838-substrate specific channel activity105.32E-03
KEGGmmu00340:Histidine metabolism31.96E-02
mmu03320:PPAR signaling pathway43.10E-02
DownBPGO:0008202-steroid metabolic process97.92E-05
GO:0055114-oxidation reduction172.25E-04
GO:0010565-regulation of cellular ketone metabolic process49.96E-04
CC GO:0005792-microsome92.16E-04
GO:0042598-vesicular fraction92.71E-04
GO:0005783-endoplasmic reticulum196.14E-04
MF GO:0030414-peptidase inhibitor activity112.89E-06
GO:0004866-endopeptidase inhibitor activity101.06E-05
GO:0004857-enzyme inhibitor activity114.83E-05
KEGG mmu00590:Arachidonic acid metabolism53.56E-03
mmu00511:Other glycan degradation36.44E-03
mmu00830:Retinol metabolism41.49E-02
mmu04610:Complement and coagulation cascades41.94E-02

[i] DEGs, differentially expressed genes; HF, hypertensive female; up, upregulated DGEs; down, downregulated DEGs; BP, biological process; CC, cellular component; MF, molecular function; KEGG, Kyoto Encyclopedia of Genes and Genomes.

In the HM group, the upregulated DEGs were mainly enriched in GO terms related to co-factor metabolic process and oxidation reduction, and in the pathway of PPAR signaling. The downregulated DEGs were mainly enriched in GO terms related to hormone metabolic process and endoplasmic reticulum (Table II).

Table II

Functional enrichment analysis for the DEGs of the HM group.

Table II

Functional enrichment analysis for the DEGs of the HM group.

TermCountP-value
UpBPGO:0051186-cofactor metabolic process81.04E-03
GO:0055114-oxidation reduction152.04E-03
GO:0019748-secondary metabolic process53.02E-03
CC GO:0005576-extracellular region374.68E-06
GO:0005777-peroxisome52.03E-02
GO:0042579-microbody52.03E-02
MFGO:0019842-vitamin binding101.64E-06
GO:0016290-palmitoyl-CoA hydrolase activity38.12E-03
GO:0030170-pyridoxal phosphate binding41.33E-02
KEGGmmu03320:PPAR signaling pathway56.25E-03
mmu00590:Arachidonic acid metabolism57.43E-03
DownBPGO:0042445-hormone metabolic process71.63E-04
GO:0010817-regulation of hormone levels71.07E-03
GO:0042398-cellular amino acid derivative biosynthetic process49.31E-03
CC GO:0005783-endoplasmic reticulum151.29E-02
MF GO:0004866-endopeptidase inhibitor activity73.52E-03
GO:0030414-peptidase inhibitor activity75.43E-03
GO:0004867-serine-type endopeptidase inhibitor activity52.09E-02

[i] DEGs, differentially expressed genes; HM, hypertensive male; up, upregulated DGEs; down, downregulated DEGs; BP, biological process; CC, cellular component; MF, molecular function; KEGG, Kyoto Encyclopedia of Genes and Genomes.

PPI network construction

The constructed PPI network based on the DEGs between the HF and NF groups is shown in Fig. 2A. The PPI network consisted of 193 nodes and 301 interaction pairs. The top 10 hub genes were selected based on the analysis of degree centrality, betweenness centrality and subgraph centrality. Among the 10 genes, cytochrome P450, family 2, subfamily E, polypeptide 1 (Cyp2e1) had the highest degree in all the 3 analysis methods (Table III).

Table III

The top 10 network nodes in the HF group based on the analysis of degree centrality, betweenness centrality and subgraph centrality.

Table III

The top 10 network nodes in the HF group based on the analysis of degree centrality, betweenness centrality and subgraph centrality.

GeneDegreeBetweennessSubgragh
Cyp2e118Cyp2e17915.4395Cyp2e1295.83835
Cyp4a12a16Aldh1a14667.3003Cyp4a12a279.15607
Cyp2b1011Pla2g54572.402Cyp2b10179.15228
Pla2g59Srebf14370.598Slc22a30100.235176
Srebf19Dkk14108.5645Pla2g599.77931
Serpina1d9Cyp4a12a3427.9216Slco1a199.73588
Cyp7b19Pld13171.1284Cyp4f1881.0327
Slc22a309Serpina1d3132.0713Cyp4f1481.03269
Anxa18Prok13062.5642Aldh1a173.60265
Gm2668Plcl22823.1924Aldh1a769.66662

[i] HF, hypertensive female.

The PPI network constructed by the DEGs between HM and NM included 182 nodes and 248 PPI interaction pairs (Fig. 2B). The top 10 hub genes respectively analyzed by 3 methods are shown in Table IV, namle UDP glucuronosyltransferase 2 family, polypeptide B37 (Ugt2b37), formimidoyltransferase cyclodeaminase (Ftcd) and cytochrome P450, family 4, subfamily a, polypeptide 14 (Cyp4a14).

Table IV

The top 10 network nodes in the HM group based on the analysis of degree centrality, betweenness centrality and subgraph centrality.

Table IV

The top 10 network nodes in the HM group based on the analysis of degree centrality, betweenness centrality and subgraph centrality.

GeneDegreeBetweennessSubgragh
Ugt2b3711Ftcd5603.764Cyp4a1457.048008
Cyp4a1010Lep4363.4756Cyp4a1055.98465
Cyp4a1410Ugt2b374147.9277Ppbp52.882313
Lep10Serpina1d4005.5542Ugt2b3750.838398
Ppbp9Cyp4a143500.8042Aldh1a734.876118
Tgfb28Fcgr2b2844.4468Aldh1a134.272568
Ftcd8Pld12784.8557Aplnr28.300505
Aldh1a77Plcl22771.22Grm728.300499
Aldh1a17Il10rb2728.78Adra2b28.300493
A2m7Synj22569.02Oxgr128.300491

[i] HM, hypertensive male.

TF analysis

Based on the TRANSFAC database in iRegulon, the TFs in the HF and HM samples were predicted and the transcriptional regulatory networks were constructed (Fig. 3A and B). In the HF group, the TFs were sterol regulatory element binding transcription factor 1 (Srebf1) and estrogen receptor 1 (Esr1). In the HM group, the TFs were retinoid X receptor gamma (Rxrg) and peroxisome proliferator-activated receptor gamma (Pparg).

Analysis of DEGs in the HF and HM groups

There were 118 intersection genes among the upregulated DEGs in the HF and HM samples; they were mainly distributed in chromosomes 4 and 7. Among the downregulated DEGs, 112 were intersection genes and they were mainly distributed in chromosomes 11 and 14. Moreover, these intersection genes were mainly enriched in BP terms related to catecholamine biosynthetic process, CC terms related to extracellular region and MF terms related to endopeptidase inhibitor activity (Table V). For instance, lysyl oxidase-like 2 (Loxl2) was enriched in the CC term related to extracellular region. The interaction network for these terms are shown in Fig. 4.

Table V

The significant functions enriched by the intersection genes.

Table V

The significant functions enriched by the intersection genes.

TermCountP-value
BP GO:0042423-catecholamine biosynthetic process30.004975549
GO:0006638-neutral lipid metabolic process40.007615151
GO:0042445-hormone metabolic process50.008732351
CC GO:0005576-extracellular region314.56E-04
GO:0000267-cell fraction120.02798221
GO:0005626-insoluble fraction110.030455631
MF GO:0004866-endopeptidase inhibitor activity91.17E-04
GO:0030414-peptidase inhibitor activity92.16E-04
GO:0004867-serine-type endopeptidase inhibitor activity76.59E-04

[i] BP, biological process; CC, cellular component; MF, molecular function.

Discussion

In the present study, 411 DEGs were identified between the HF and NF samples, and 418 DEGs were identified between the HM and NM samples. The upregulated DEGs, Cyp4b1 and Cyp4a31, were mainly enriched in BP terms related to oxidation reduction in both the HF and HM samples. In addition, Esr1, Pparg and Rxrg were the key TFs involved in the transcriptional regulatory networks. Following the analysis of DEGs in the HF and HM samples, many intersection DEGs were identified between the 2 groups, such as Loxl2. These genes and TFs may play important roles in the progression of hypertension.

Previous studies have indicated that reactive oxygen species play an important pathophysiological role in the development of cardiovascular diseases, including hypertension (25,26). In human hypertension, biomarkers of systemic oxidative stress have been found to be elevated (27). In the present study, BP terms related to oxidation reduction were significant terms enriched by the DEGs in both the HF and HM samples. For instance, several cytochrome P450 (CYP)-associated genes, including Cyp4b1 and Cyp4a31 were enriched in this BP term. CYPs are important enzymes involved in the oxidative and reductive metabolism of numerous endogenous compounds in mammals (28). In the study by Chiu et al (2), Cyp4b1 was validated to be differentially expressed between hypertensive and normotensive mice by qPCR. CYP4A proteins have been found in vascular tissues (29) and have been suggested to be linked to hypertension in numerous experimental models (30). Sacerdoti et al (31) demonstrated that the inhibition or depletion of CYP4A activity can lower blood pressure in the spontaneously hypertensive rat. Of note, Cyp4a31 was found enriched in the PPAR signaling pathway in HF and HM samples. The PPARs are members of the nuclear receptor family of ligand-activated TFs, which play an important role in the regulation of cardiovascular circadian rhythms (32). Therefore, the upregulation of Cyp4b1 and Cyp4a31 in our study was in accordance with previous research. The 2 DEGs may be critical biomarkers associated with human hypertension.

In addition, Esr1 was a key TF in the transcriptional regulatory networks of the HF sample group. Esr1 is a member of the family of ESRs, belonging to superfamily of nuclear receptor TFs. This receptor is expressed in a wide range of tissues and cells, including vascular smooth muscle and vascular endothelial cells (33). Estrogen can increase vasodilatation and inhibit the development of atherosclerosis (33). Specifically, the incidence of cardiovascular disease, including hypertension differs between males and females (34). Accumulating evidence suggests that estrogen deficiency plays a major role in the pathogenesis of cardiovascular diseases in post-menopausal women (35). In our study, the TF, Esr1, was also identified in female hypertensive mice, which indicated that Esr1 may play an important role in the progression of hypertension in females.

Furthermore, the TFs, Pparg and Rxrg, were involved in the transcriptional regulatory networks in the HM samples. Pparg is a member of PPARs which, as mentioned above, play an important role in the regulation of cardiovascular circadian rhythms. Pparg has been implicated in atherosclerosis (36), coronary heart disease (37) and hypertension (38). RXRs are members of the NR2B family of nuclear receptors and are major heterodimerization partners of PPARs (39). RXRs are involved in mediating the anti-proliferative effects of retinoic acid, which can comprise a collection of molecules with retinoids to maintain homeostasis in adults (40). A growing number of studies have reported the role of RXRs and PPARs in controlling cellular metabolism in tissues, including the myocardium (40,41). Nohara et al (39) suggested that a variant of Rxrg contributes to human genetic dyslipidemia. Taken together, the TFs, Pparg and Rxrg, may have utility in the diagnose and treatment of hypertension.

Following the analysis of DEGs in the HF and HM samples, we found that there were many intersection DEGs between the 2 groups, such as Loxl2. This DEG was found enriched in the BP terms related to oxidation reduction and CC terms related to extracellular region. Loxl2 encodes a member of the lysyl oxidase gene family, which are extracellular copper-dependent amine oxidases initiating the formation of crosslinks in collagen and elastin (42). The collagen and elastin are important components of the extracellular matrix (43). It has been found that the activity of lysyl oxidase is essential to maintain the features of connective tissues, including cardiovascular systems (44). The deregulation of lysyl oxidase is associated with aneurysms, aortic rupture and different stages of the atherosclerotic process (45,46). Presently, there are few studies on the association between lysyl oxidase and hypertension. Thus, we speculated that Loxl2 may be a potential biomarker for the progression of hypertension.

In conclusion, our study provides a comprehensive bioinformatics analysis of biomarkers in hypertension, which may provide new insight into the mechanisms responsible for this disease. The DEGs, Cyp4b1, Cyp4a31 and Loxl2, and the TFs, Esr1, Pparg and Rxrg, may be associated with the progression of hypertension, and may serve as potential therapeutic targets in this disease.

However, there was a limitation to this study. The present study was implemented by means of bioinformatics; therefore, the screened genes and TFs have not been validated by actual experiments. As a result, further experimental studies with a larger sample size are warranted to confirm the results of our study.

References

1 

Lupton SJ, Chiu CL and Lind JM: A hypertension gene: are we there yet? Twin Res Hum Genet. 14:295–304. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Chiu CL, Jackson KL, Hearn L, Steiner N, Head GA and Lind JM: Identification of genes with altered expression in male and female Schlager hypertensive mice. BMC Med Genet. 15:1012014. View Article : Google Scholar : PubMed/NCBI

3 

Williams SS: Advances in genetic hypertension. Curr Opin Pediatr. 19:192–198. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Chobanian AV, Bakris GL, Black HR, Cushman WC, Green LA, Izzo JL Jr, Jones DW, Materson BJ, Oparil S, Wright JT Jr, et al Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure. National Heart, Lung, and Blood Institute; National High Blood Pressure Education Program Coordinating Committee: Seventh report of the joint national committee on prevention, detection, evaluation, and treatment of high blood pressure. Hypertension. 42:1206–1252. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Mendis S, Puska P and Norrving B: Global atlas on cardiovascular disease prevention and control. World Health Organization; Geneva: 2011

6 

Northcott CA, Glenn JP, Shade RE, Kammerer CM, Hinojosa-Laborde C, Fink GD, Haywood JR and Cox LA: A custom rat and baboon hypertension gene array to compare experimental models. Exp Biol Med (Maywood). 237:99–110. 2012. View Article : Google Scholar

7 

Ehret GB, Munroe PB, Rice KM, Bochud M, Johnson AD, Chasman DI, Smith AV, Tobin MD, Verwoert GC, Hwang SJ, et al International Consortium for Blood Pressure Genome-Wide Association Studies; CARDIoGRAM consortium; CKDGen Consortium; KidneyGen Consortium; EchoGen consortium: CHARGE-HF consortium: Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature. 478:103–109. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Lifton RP, Gharavi AG and Geller DS: Molecular mechanisms of human hypertension. Cell. 104:545–556. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Machado RD, Eickelberg O, Elliott CG, Geraci MW, Hanaoka M, Loyd JE, Newman JH, Phillips JA III, Soubrier F, Trembath RC, et al: Genetics and genomics of pulmonary arterial hypertension. J Am Coll Cardiol. 54(Suppl 1): S32–S42. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Gooch JL and Sharma AC: Targeting the immune system to treat hypertension: where are we? Curr Opin Nephrol Hypertens. 23:473–479. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Austin ED, Ma L, LeDuc C, Berman Rosenzweig E, Borczuk A, Phillips JA III, Palomero T, Sumazin P, Kim HR, Talati MH, et al: Whole exome sequencing to identify a novel gene (caveolin-1) associated with human pulmonary arterial hypertension. Circ Cardiovasc Genet. 5:336–343. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Puig O, Wang I-M, Cheng P, Zhou P, Roy S, Cully D, Peters M, Benita Y, Thompson J and Cai TQ: Transcriptome profiling and network analysis of genetically hypertensive mice identifies potential pharmacological targets of hypertension. Physiol Genomics. 42A:24–32. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Carvalho BS and Irizarry RA: A framework for oligonucleotide microarray preprocessing. Bioinformatics. 26:2363–2367. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U and Speed TP: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Smyth GK: Limma: linear models for microarray data. Bioinformatics and Computational Biology Solutions using R and Bioconductor. Gentleman R, Carey V, Huber W, Irizarry R and Dudoit S: Springer; New York: pp. 397–420. 2005, View Article : Google Scholar

16 

Ferreira JA: The Benjamini-Hochberg method in the case of discrete test statistics. Int J Biostat. 3:112007. View Article : Google Scholar

17 

Harris MA, Clark J, Ireland A, Lomax J, Ashburner M, Foulger R, Eilbeck K, Lewis S, Marshall B, Mungall C, et al: Gene Ontology Consortium: The Gene Ontology (GO) database and informatics resource. Nucleic Acids Res. 32:D258–D261. 2004. View Article : Google Scholar

18 

Kanehisa M and Goto S: KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar

19 

Huang 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

20 

Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, et al: STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 43:D447–D452. 2015. View Article : Google Scholar

21 

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

22 

Tang Y, Li M, Wang J, Pan Y and Wu FX: CytoNCA: a cytoscape plugin for centrality analysis and evaluation of protein interaction networks. Biosystems. 127:67–72. 2015. View Article : Google Scholar

23 

Janky R, Verfaillie A, Imrichová H, Van de Sande B, Standaert L, Christiaens V, Hulselmans G, Herten K, Naval Sanchez M, Potier D, et al: iRegulon: from a gene list to a gene regulatory network using large motif and track collections. PLoS Comput Biol. 10:e10037312014. View Article : Google Scholar : PubMed/NCBI

24 

Merico D, Isserlin R, Stueker O, Emili A and Bader GD: Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS One. 5:e139842010. View Article : Google Scholar : PubMed/NCBI

25 

Touyz RM: Reactive oxygen species in vascular biology: role in arterial hypertension. Expert Rev Cardiovasc Ther. 1:91–106. 2003. View Article : Google Scholar

26 

Chabrashvili T, Tojo A, Onozato ML, Kitiyakara C, Quinn MT, Fujita T, Welch WJ and Wilcox CS: Expression and cellular localization of classic NADPH oxidase subunits in the spontaneously hypertensive rat kidney. Hypertension. 39:269–274. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Redón J, Oliva MR, Tormos C, Giner V, Chaves J, Iradi A and Sáez GT: Antioxidant activities and oxidative stress byproducts in human hypertension. Hypertension. 41:1096–1101. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Nelson DR and Nebert DW: Cytochrome P450 (CYP) gene superfamily. eLS. 2011. View Article : Google Scholar

29 

Marji JS, Wang M-H and Laniado-Schwartzman M: Cytochrome P-450 4A isoform expression and 20-HETE synthesis in renal preglomerular arteries. Am J Physiol Renal Physiol. 283:F60–F67. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Singh H, Cheng J, Deng H, Kemp R, Ishizuka T, Nasjletti A and Schwartzman ML: Vascular cytochrome P450 4A expression and 20-hydroxyeicosatetraenoic acid synthesis contribute to endothelial dysfunction in androgen-induced hypertension. Hypertension. 50:123–129. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Sacerdoti D, Escalante B, Abraham NG, Mcgiff JC, Levere RD and Schwartzman ML: Treatment with tin prevents the development of hypertension in spontaneously hypertensive rats. Science. 243:388–390. 1989. View Article : Google Scholar : PubMed/NCBI

32 

Kobayashi N, Ohno T, Yoshida K, Fukushima H, Mamada Y, Nomura M, Hirata H, Machida Y, Shinoda M, Suzuki N, et al: Cardioprotective mechanism of telmisartan via PPAR-γ-eNOS pathway in dahl salt-sensitive hypertensive rats. Am J Hypertens. 21:576–581. 2008. View Article : Google Scholar : PubMed/NCBI

33 

Mendelsohn ME and Karas RH: The protective effects of estrogen on the cardiovascular system. N Engl J Med. 340:1801–1811. 1999. View Article : Google Scholar : PubMed/NCBI

34 

Barrett-Connor E: Sex differences in coronary heart disease. Why are women so superior? The 1995 Ancel Keys Lecture. Circulation. 95:252–264. 1997. View Article : Google Scholar : PubMed/NCBI

35 

Mendelsohn ME: Protective effects of estrogen on the cardiovascular system. Am J Cardiol. 89:12E–17E. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Kersten S, Desvergne B and Wahli W: Roles of PPARs in health and disease. Nature. 405:421–424. 2000. View Article : Google Scholar : PubMed/NCBI

37 

Yilmaz-Aydogan H, Kurnaz O, Kurt O, Akadam-Teker B, Kucukhuseyin O, Tekeli A and Isbir T: Effects of the PPARG P12A and C161T gene variants on serum lipids in coronary heart disease patients with and without type 2 diabetes. Mol Cell Biochem. 358:355–363. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Matsuda Y, Hoshikawa Y, Ameshima S, Suzuki S, Okada Y, Tabata T, Sugawara T, Matsumura Y and Kondo T: Effects of peroxisome proliferator-activated receptor gamma ligands on monocrotaline-induced pulmonary hypertension in rats. Nihon Kokyuki Gakkai Zasshi. 43:283–288. 2005.In Japanese. PubMed/NCBI

39 

Nohara A, Kawashiri M-A, Claudel T, Mizuno M, Tsuchida M, Takata M, Katsuda S, Miwa K, Inazu A, Kuipers F, et al: High frequency of a retinoid X receptor γ gene variant in familial combined hyperlipidemia that associates with atherogenic dyslipidemia. Arterioscler Thromb Vasc Biol. 27:923–928. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Sucov HM, Dyson E, Gumeringer CL, Price J, Chien KR and Evans RM: RXR alpha mutant mice establish a genetic basis for vitamin A signaling in heart morphogenesis. Genes Dev. 8:1007–1018. 1994. View Article : Google Scholar : PubMed/NCBI

41 

Osorio JC, Stanley WC, Linke A, Castellari M, Diep QN, Panchal AR, Hintze TH, Lopaschuk GD and Recchia FA: Impaired myocardial fatty acid oxidation and reduced protein expression of retinoid X receptor-α in pacing-induced heart failure. Circulation. 106:606–612. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Rodríguez C, Martínez-González J, Raposo B, Alcudia JF, Guadall A and Badimon L: Regulation of lysyl oxidase in vascular cells: lysyl oxidase as a new player in cardiovascular diseases. Cardiovasc Res. 79:7–13. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Mäki JM, Räsänen J, Tikkanen H, Sormunen R, Mäkikallio K, Kivirikko KI and Soininen R: Inactivation of the lysyl oxidase gene Lox leads to aortic aneurysms, cardiovascular dysfunction, and perinatal death in mice. Circulation. 106:2503–2509. 2002. View Article : Google Scholar : PubMed/NCBI

44 

Lucero HA and Kagan HM: Lysyl oxidase: an oxidative enzyme and effector of cell function. Cell Mol Life Sci. 63:2304–2316. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Sibon I, Sommer P, Lamaziere JM and Bonnet J: Lysyl oxidase deficiency: a new cause of human arterial dissection. Heart. 91:e33. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Nakashima Y and Sueishi K: Alteration of elastic architecture in the lathyritic rat aorta implies the pathogenesis of aortic dissecting aneurysm. Am J Pathol. 140:959–969. 1992.PubMed/NCBI

Related Articles

Journal Cover

July-2017
Volume 40 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gao Y, Qi G, Jia Z and Sun Y: Prediction of marker genes associated with hypertension by bioinformatics analyses. Int J Mol Med 40: 137-145, 2017
APA
Gao, Y., Qi, G., Jia, Z., & Sun, Y. (2017). Prediction of marker genes associated with hypertension by bioinformatics analyses. International Journal of Molecular Medicine, 40, 137-145. https://doi.org/10.3892/ijmm.2017.3000
MLA
Gao, Y., Qi, G., Jia, Z., Sun, Y."Prediction of marker genes associated with hypertension by bioinformatics analyses". International Journal of Molecular Medicine 40.1 (2017): 137-145.
Chicago
Gao, Y., Qi, G., Jia, Z., Sun, Y."Prediction of marker genes associated with hypertension by bioinformatics analyses". International Journal of Molecular Medicine 40, no. 1 (2017): 137-145. https://doi.org/10.3892/ijmm.2017.3000