Open Access

Candidate gene and mechanism investigations in congenital obstructive nephropathy based on bioinformatics analysis

  • Authors:
    • Guangda Xin
    • Rui Chen
    • Xiaofei Zhang
  • View Affiliations

  • Published online on: July 16, 2018     https://doi.org/10.3892/mmr.2018.9284
  • Pages: 2651-2660
  • Copyright: © Xin 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 aim of the present study was to explore the candidate genes, chemicals and mechanisms of congenital obstructive nephropathy (CON). The gene expression profiles of GSE48041, including 24 kidney tissue samples from megabladder (mgb‑/‑) mouse were downloaded from the Gene Expression Omnibus database. Samples were divided into 4 groups: Control, mild, moderate and severe. Differentially expressed genes (DEGs), protein‑protein interaction network, Kyoto Encyclopedia of Genes and Genomes pathways and transcription factor (TF)‑target gene analyses were performed on Set 1 (mild, moderate and severe groups), while Gene Ontology (GO) function enrichment analysis and chemical investigation were performed on Set 2 (severe group). A total of 187 and 139 DEGs were obtained in Set 1 and Set 2, respectively. Chemical carcinogenesis [enriched by genes such as Carbonyl reductase 1 (CBR1)] was one of the most prominent pathways in Set 1. GO analysis for Set 2 revealed that DEGs were mainly assembled in functions such as cellular response to interleukin‑1 and cellular response to tumor necrosis. Furthermore, genes such as Fos Proto‑Oncogene (FOS) were co‑regulated by TFs including RNA polymerase II subunit A (Polr2a) and serum response factor (Srf). Chemical cyclosporine served the most important role in Set 2 by targeting several DEGs in Set 2. DEGs such as CBR1 and FOS, TFs including Polr2a and Srf, and pathways such as chemical carcinogenesis may serve important roles in the process of CON. Interleukin‑1 and tumor necrosis function may be novel targets for CON gene therapy. Furthermore, cyclosporine may be a promising option for future CON therapy.

Introduction

Congenital obstructive nephropathy (CON) is a main cause of kidney insufficiency in child and infant (1). It is associated with various diseases, such as prune belly syndrome (2). As a result, CON generates a huge social burden especially in the morbidity and mortality (2). The different challenges associated with CON in areas like diagnose and therapy highlight the importance of the molecular mechanism of CON (3). Thus, the emerging theories of the biology of CON suggest new targets for therapeutic interventions (4).

Identification of novel biomarkers provides prognostic value for this process remains a major goal in the study of kidney disease (5). In renal and urinary tract, previous study showed that disruption of angiotensin type 2 receptor (AGTR2) induced a huge scale of anomaly (6). Meanwhile, the Monocyte chemotactic protein 1 (MCP-1) as well as the Epidermal growth factor (EGF) seem to be involved in the pathogenesis of tubulointerstitial damage of CON, and their urine excretion may serve as a powerful prognostic marker for this form renal disease (7). Not only gene, but also pathways enriched by differentially expressed genes (DEGs) are closed related with CON. Previous study demonstrated that TGF-β signaling pathway plays an important role in regulating kidney injury process following by obstruction (8). In a rat model, Hermens et al (9) show that there is an age-dependent role of Wnt signaling pathway in the pathophysiology of CON. Despite of revealing the potential genes or pathways, understanding the molecular mechanism of CON also brings a breakthrough for the novel treatment of CON (10). The development of biomarkers contribute to molecular therapies of CON (11). However, the molecular mechanisms underlying these histologic changes are still poorly defined.

To better understand the pathophysiology of CON, Brian Becknell and his colleagues evaluated the global transcription in kidneys with graded hydronephrosis in the megabladder (mgb2/2) mouse (12). Based on normal, mild, moderate, and severe mouse models, they prove that the development of progressive hydronephrosis can result in renal adaptation, including significant changes in the morphology and potential functionality of the renal urothelium. However, the co-regulated gene in all models, transcription factor (TF) associated with DEGs, gene expression in severe disease status, as well as the potential chemicals for CON treatment are still unclear. In the current study, a bioinformatics research was designed based on the gene expression profile provided by Becknell et al (12). Then, investigations of DEGs, function and pathway enrichment, protein-protein interaction network (PPI) analyses, as well as TF-target gene regulatory network were performed in all disease status. Furthermore, the chemical-gene interaction network in severe models were also investigated. We aimed to explore the potential pathoenesis of CON and provided information about novel gene targets, as well as chemicals for CON treatment.

Materials and methods

Microarray data

Expression data of GSE48041 (12), sequenced on the platform of GPL10787 Agilent-028005 SurePrint G3 Mouse GE 8×60K Microarray, were obtained from the Gene Expression Omnibus (GEO) database (www.ncbi.nlm.nih.gov/geo/). A total of 24 kidney tissue samples from control and megabladder (mgb−/−) mouse aged 23–30 days were included in this dataset, and hydronephrosis was induced by renal ultrasound as previously reported (13). According to the hydronephrosis degrees (www.sfu-urology.org/sfu_hydrone_grading.cfm), animals were divided into 4 groups: Control (n=6), mild (n=7), moderate (n=5) and severe (n=6) according to their disease stages.

Data preprocessing

The preprocessing were performed based on robust multi-array average (RMA) method (14) in Affy package (15) in R (v.3.10.3, bioconductor.org/biocLite.R). Contents of processing included background correct microarray expression intensities, normalize the expression within each array, and expression calculation. The probe ID was convert to the gene symbol based on the chip platform annotation files.

DEGs analysis

Classical Bayes method in Linear Models for Microarray Data (limma) package (16) of R was used to reveal DEGs by comparing expression value among samples from three comparisons: Mild vs. control, moderate vs. control, and severe vs. control. P-value <0.05 and |log2 log fold change (FC)|≥1 were selected as the cut-off criteria for DEG screening.

VennPlot analysis of DEGs

VENNY (v.2.1) (17) is an online tool used for Venn diagram analysis based on gene expression value. The number of up-regulated, down-regulated, and contra-regulated genes can be marked by VennPlex. In the present study, VennPlex software was used to investigate the difference of DEGs among 3 comparative groups. Then, the co-regulated DEGs (Set 1) and severe group DEGs (Set 2) were selected for further investigation.

Enrichment analysis of DEGs

The Database for Annotation, Visualization, and Integrated Discovery (DAVID; v.6.8) (18) provides a synthetic set of functional annotation tools for users for revealing the biological significances among huge scale of genes. Gene Ontology (GO) function enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enrichment analysis were performed using DAVID online tool (19). GO functional categories including molecular function (MF), biological process (BP), and cellular component (CC) (20). In the present study, GO functions assembled by DEGs in severe group, as well as KEGG pathways enriched by the co-regulated DEGs were investigated. P-value <0.05 and enriched gene numbers (count) ≥2 were considered as cut-off values.

PPI network investigation

Search Tool for the Retrieval of Interacting Genes/Proteins (STRING; v.10.0) is an online tool for the predication of PPI based on a large scale of biological database (21). In the present study, STRING was used to predict the interaction relations between DEGs corresponding coding proteins. PPI were revealed based on the STRING database with score (median confidence)=0.4, and nodes represent DEGs in the PPI interaction. The degree was defined as the number of the connections for the target proteins. Cytoscape software (v.3.2.0) (22,23) was used for the visualization of PPI network obtained above. Then, the CytoNCA software (v.2.1.6, apps.cytoscape.org/apps/cytonca) (24) was used for the nodes topological analysis (Parameter: Without weight). Furthermore, the sub-networks with score >4.5 were identified by MOCDE (v.1.4.2; apps.cytoscape.org/apps/MCODE) (23) plugin in Cytoscape software.

TF-target gene regulatory network construction

TF is an important class of participators in the regulation of gene expression. Analysis of TF binding sites is of great significance for the study of gene regulation system. In the current research, the TFs-co-regulated DEGs network was constructed with iRegulon (25) (v.1.3, apps.cytoscape.org/apps/iRegulon). The Parameters of this analysis were set as: Minimum identity between orthologous genes=0.05, and maximum false discovery rate (FDR) on motif similarity=0.001. TF-target gene relations with normalized enrichment score (NES) >4 were considered as the meaningful results.

Chemical-DEGs interaction network construction

Comparative Toxicogenomics Database (CTD) provides relationships, such as chemical-gene/protein interactions, chemical-disease as well as gene-disease (26). Relationship between disease (Nephrosis, congenital) associated chemical-gene or interactions was revealed by CDT. Then Chemical-DEGs interaction network was constructed by Cytoscape software.

Results

DEGs investigation in mild, moderate and severe groups

Considering a huge amount of calculation in gene expression profile, the original data was analyzed and filtered. The results revealed that totally 220 up-regulated DEGs and 225 down-regulated DEGs were obtained in the mild group. In the moderate group, there were 375 up-regulated DEGs and 142 down-regulated DEGs screened out. Furthermore, a total of 484 up-regulated DEGs and 280 down-regulated DEGs uncovered in the severe group.

VennPlot analysis

VennPlot for DEGs in the mild, moderate, and severe groups were investigated in the present study (Fig. 1). The results revealed 187 co-regulated DEGs in Set 1, including 87 up-regulated DEGs and 100 down-regulated DEGs. In Set 1, Serpina6, Hpd, and Nr4a1 were the 3 most outstanding up-regulated genes, while Proz, 4122401K19Rik, and Nudt19 were the 3 most outstanding down-regulated genes. Meanwhile, there were 139 DEGs in Set 2, including 121 up-regulated DEGs and 18 down-regulated DEGs. In Set 2, Sprr2f, Lcn2 and Saa2 were the 3 most outstanding up-regulated gene, while Symbol, Vps8 and Ang2 were the 3 most down-regulated gene.

KEGG and GO enrichment analysis

The KEGG analysis of DEGs in Set 1 showed that a total of 16 pathways were significantly enriched (Table I). The up-regulated DEGs were mostly enriched in TNF signaling pathway (mmu04668, P=3.59×10−4), while the down-regulated DEGs were mainly assembled in Retinol metabolism (mmu00830, P=1.36×10−7). Among these pathways, Chemical carcinogenesis (mmu05204, Gene: CBR1, UGT2B38, GSTA2, UGT2B37 and UGT2B36) was the common pathway in all 3 group of Set 1. The detail pathway map of Chemical carcinogenesis was showed in Fig. 2.

Table I.

Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis for common DEGs in the mild, moderate and severe groups.

Table I.

Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis for common DEGs in the mild, moderate and severe groups.

RegulationPathway IDPathway nameCountP-value
ALLmmu00980Metabolism of xenobiotics by cytochrome P45092.84×10-7
mmu00830Retinol metabolism103.16×10-7
mmu05204Chemical carcinogenesis104.21×10-7
mmu00982Drug metabolism-cytochrome P45085.09×10-6
mmu00053Ascorbate and aldarate metabolism51.64×10-4
mmu01100Metabolic pathways274.04×10-4
mmu00040Pentose and glucuronate interconversions55.13×10-4
mmu00860Porphyrin and chlorophyll metabolism58.48×10-4
mmu00983Drug metabolism-other enzymes51.93×10-3
mmu00140Steroid hormone biosynthesis62.12×10-3
mmu00590Arachidonic acid metabolism62.34×10-3
mmu04668TNF signaling pathway65.60×10-3
mmu04976Bile secretion56.42×10-3
mmu00071Fatty acid degradation41.45×10-2
mmu05140Leishmaniasis42.93×10-2
mmu04610Complement and coagulation cascades44.53×10-2
UPmmu04668TNF signaling pathway63.59×10-4
mmu05140Leishmaniasis45.65×10-3
mmu04010MAPK signaling pathway61.39×10-2
mmu05204Chemical carcinogenesis41.52×10-2
mmu05152Tuberculosis51.74×10-2
mmu05166HTLV–I infection62.01×10-2
mmu05142Chagas disease (American trypanosomiasis)42.06×10-2
mmu04978Mineral absorption32.26×10-2
DOWNmmu00830Retinol metabolism81.36×10-7
mmu00053Ascorbate and aldarate metabolism56.48×10-6
mmu00980Metabolism of xenobiotics by cytochrome P45061.03×10-5
mmu00982Drug metabolism-cytochrome P45061.20×10-5
mmu01100Metabolic pathways181.73×10-5
mmu00040Pentose and glucuronate interconversions52.11×10-5
mmu00860Porphyrin and chlorophyll metabolism53.57×10-5
mmu00140Steroid hormone biosynthesis64.64×10-5
mmu05204Chemical carcinogenesis66.07×10-5
mmu00983Drug metabolism-other enzymes58.52×10-5
mmu04146Peroxisome46.63×10-3
mmu00071Fatty acid degradation32.17×10-2

[i] Count values indicate the number of genes enriched in a pathway. P<0.05 and the gene number (count) ≥2 were considered the threshold values for significant differences. DEGs, differentially expressed genes; UP, upregulated; DOWN, downregulated; ALL, up- and downregulated.

GO functional analysis was performed in the severe group DEGs. With count number >10, the top 10 of BP, CC, and MF functions were revealed, respectively. The DEGs in the severe group was mainly assembled in functions like inflammatory response (BP, GO: 0006954, P=1.43×10−3), cellular response to interleukin-1 (BP, GO: 0071347, P=8.87×10−3), cellular response to tumor necrosis (BP, GO: 0071356, P=6.34×10−4), extracellular region (CC, GO: 0005576, P=3.53×10−7), and cytokine activity (MF, GO: 0005125, P=7.63×10−5). The detail information was showed in Fig. 3.

PPI network and modules analysis

To dig out the potential interactions of the DEGs, PPI network and related modules were constructed based on the protein interactions of DEGs. A total of 102 nodes (Jun, Fos, Ptgs2, Ugt2b37, Junb, Atf3, Cyp4a12a, Adh1, Ugt2b5, Egr1, etc.) and 320 interactions were identified in the PPI network of Set 1. With score >4.5, 2 modules were further obtained from the Set 1 PPI network. There were 13 nodes (Jun, Fos, Ptgs2, Junb, Atf3, Egr1, Dusp1, Nr4a1, Zfp36, Btg2, etc.) and 60 interactions in module a (score=10), while 8 nodes (Ugt2b5, Cyp2j13, Acsm3, Cyp4a12b, Cyp4a14, Cyp4b1 Slc22a30 and Slc7a13) and 20 interactions contained in module b (score=5.714) (Fig. 4). Furthermore, there were a total of 73 nodes (Ccl2, Timp1, Acta2, Spp1, Actg2, Thbs1, Edn1, Aldh1a1, Mmp3, Aldh1a7, etc.) and 143 interactions included in the PPI network of Set 2. With score >4.5, one module was further obtained from the Set 2 PPI network. A total of 5 nodes (Aldh1a1, Aldh1a7, Aifm3, Hbb-b1 and Acad9) and 9 interactions were revealed in the module of Set 2 (Fig. 5).

TF-target gene regulatory network analysis

By using the iRegulon software, 2 TFs (Polr2a and Srf) were obtained from the Set 1 PPI network. To further investigate the relationship between TFs and their target genes, the TF-target gene network was visualized by cytoscape software. With NES >4, the results showed that there were totally 52 regulatory relations contained in this network. Some genes like FOS were co-regulated by both Polr2a and Srf. The detail information was showed in Fig. 6.

Chemical-gene interaction network analysis

The present study revealed totally 12012 chemical-gene interactions related to Nephrosis or congenital. Among these interactions, totally 60 interactions were matched with the 139 DEGs in Set 2. The results showed that Cyclosporine was the most important chemical that target with either the most number of up-regulated or down-regulated DEGs in Set 2. Detailed information was listed in Table II.

Table II.

Chemical-target gene interaction network.

Table II.

Chemical-target gene interaction network.

GeneChemical nameDescriptionGeneChemical nameDescriptionGeneChemical nameDescription
ACTA2CyclosporineUPEREGCyclosporineUPRGS16CyclosporineUP
ACTG2CyclosporineUPFGBCyclosporineUPRHOUCyclosporineUP
AIFM3CyclosporineDOWNFOXA1CyclosporineUPS100A6CyclosporineUP
ALDH1A1CyclosporineUPFOXQ1CyclosporineUPSAA2CyclosporineUP
APOC2CyclosporineUPGDF15CyclosporineUPSAMD5CyclosporineUP
BCAS1CyclosporineUPGPR87CyclosporineUPSDC1CyclosporineUP
BCL2L14CyclosporineUPIGFBP1CyclosporineUPSERPINA3NCyclosporineUP
CAPGCyclosporineUPIGFBP6CyclosporineUPSLC16A3CyclosporineUP
CCL2CyclosporineUPITGA11CyclosporineUPSLC17A3CyclosporineDOWN
CCL6CyclosporineUPLCN2CyclosporineUPSLC39A4CyclosporineUP
CD14CyclosporineUPLOXCyclosporineUPSPP1CyclosporineUP
CES1ECyclosporineDOWNLTBP2CyclosporineUPTACSTD2CyclosporineUP
CLDN4CyclosporineUPMDKCyclosporineDOWNTAGLNCyclosporineUP
CNN1CyclosporineUPMFAP5CyclosporineUPTGFB3CyclosporineUP
COL27A1CyclosporineDOWNMGST2CyclosporineUPTHBS1CyclosporineUP
CPCyclosporineUPMMP3CyclosporineUPTIMP1CyclosporineUP
CPECyclosporineUPMMP7CyclosporineUPTRPV6CyclosporineUP
CTSECyclosporineUPOSMRCyclosporineUPTUBB2BCyclosporineUP
EDN1CyclosporineUPPAK3CyclosporineUPUCHL1CyclosporineUP
ENTPD3AzathioprineUPPLAURCyclosporineUPUPK3ACyclosporineUP

[i] UP, upregulated; DOWN, downregulated.

Discussion

CON is the leading cause of chronic kidney disease in child. However, the molecular mechanisms underlying disease progression are still poorly defined. To reveal the potential genes or pathways associated with CON, the present bioinformatics study was performed on mild, moderate, and severe CONs. The results showed that totally 187 and 139 co-regulated DEGs were obtained in Set 1 and Set 2, respectively. KEGG analysis for Set 1 showed that DEGs were mainly enriched in pathways like chemical carcinogenesis. Meanwhile, the GO analysis for Set 2 showed that DEGs were mainly assembled in functions like cellular response to interleukin-1 and cellular response to tumor necrosis. Two modules in Set 1 and one module in Set 2 were revealed based on PPI network analysis. Furthermore, the genes like FOS were co-regulated by TFs like Polr2a and Srf. Cyclosporine was the most important chemical that target with DEGs in Set 2.

Based on the Set 1 (including all disease status like mild, moderate and severe), KEGG pathway analysis, PPI network analysis, and TF-target investigation were carried out in this study to provide a potential mechanism understanding for the process of CON. Chemical carcinogenesis is a process that the cells need genetic and epigenetic variations based on elements including oncogenes and tumor suppressors (27,28). Carbonyl reductase 1 (CBR1) metabolized by the body many toxic environmental quinones and pharmacological relevant substrates, such as the anticancer doxorubicin (27). Previous study has shown that CBR1 is vital for the cell protection in functions like oxidative stress and apoptosis (29). Pathway analysis showed that the chemical carcinogenesis was the common pathway enriched by both up- and down-regulated DEGs (especially CBR1 in Set 1). Thus, we speculated that DEGs like CBR1 in chemical carcinogenesis pathway might be the potential pathogenesis of CON. However, the related study associated with chemical carcinogenesis pathway about CBR1 and CON is very rare. Furthermore, c-Fos (FOS) gene has been considered as the regulators of cell proliferation, differentiation, transformation, and apoptotic cell death (30,31). Otsuka et al (32) indicates that the expression of FOS is potentially an early indicator of late radiation damage to the kidney. In rats model, previous study indicated that the renoprotective effects of drugs partly through inhibiting the upregulation of FOS mRNA and protein in renal cortices (33). In the present study, the PPI network analysis showed that FOS was one of the most outstanding DEGs in Set 1. Interestingly, the TF-target gene network in this study showed that FOS was the co-target gene of Polr2a and Srf. Thus, we speculated that DEGs like FOS might play an important role in the process of CON via TFs like Polr2a and Srf. However, the related study associated with FOS and CON is very rare. Thus, an additional research with a wide scale of gene expression analysis is necessary to confirm this speculation.

Based on Set 2 (only severe group), GO function analysis and chemical-gene interaction network investigation in this study were performed and intended to provide a potential therapy strategy for CON. Interleukin-1 family plays a vital role in the immune and inflammatory responses regulation (34). Previous study showed that the protective effect of interleukin-1 in renal injury might be induced by reducing the induction of the nuclear factors (35). Actually, Pindjakova et al (36) indicate that the production of interleukin-1 based on the resident dendritic cells enhances the situation of acute kidney injury. Moreover, tumor necrosis factor (TNF) is a superfamily of cytokines that can lead to cell death, and it is a potent pyrogen that can be caused by stimulation of interleukin-1 secretion (37). Meldrum et al (38) prove that TNF-α mediates renal tubular cell apoptosis in the process of obstructive uropathy. Recent study indicates that the production of TNF-α by macrophages take part in the progression of renal injury (39). Actually, interleukin-1 and TNF-α gene polymorphism are associated with the susceptibility, disease activity and long-term outcome in human nephropathy (40). In the current research, the results of GO analysis showed that cellular response to interleukin-1 and cellular response to tumor necrosis is two of most outstanding function assembled by DEGs. Thus, we speculated that these two functions might be vital for the process of severe CON. The deteriorated or even canceration might take place in severe CON, which needed an early therapy intervention. Furthermore, cyclosporine, also spelled ciclosporin or cyclosporin, is an immunosuppressant medication and natural product (41). As an immunosuppressant with therapeutic indications in various immunological diseases, the use of cyclosporine is reported to be associated with chronic nephropathy (42). In the present study, cyclosporine was the most outstanding chemical associated with DEGs identified in severe CON. Thus, cyclosporine might still be the preferences for the therapy of CON. However, due to the negative effects of cyclosporine in CON clinical treatment, a more effective drug with less adverse reaction is needed.

However, there were still some limitations in this study. First, findings in the present study were all analyzed by bioinformatics methods, and experimental validations were deficient. Second, parameters used in in silico analyses were set manually, and some results might be missed or overemphasized. Therefore, further experimental validations in both in vivo and in vitro were required in the future.

In conclusion, the DEGs like CBR1 and FOS, TFs like Polr2a and Srf, as well as pathways like chemical carcinogenesis might play important roles in the process of CON. Interleukin-1 and tumor necrosis function were associated with the deteriorate of CON, and might be novel targets for CON treatment. Furthermore, a more effective drug with less adverse reaction for severe CON is needed.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

GX contributed to the conceptualization of the research. GX and RC acquired the data and performed the analysis. RC and XZ interpreted the data. GX wrote the manuscript, and XZ revised the manuscript for important intellectual content.

Ethics 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 

Piepsz A, Ham H and Josephson S: RE: Biomarkers of congenital obstructive nephropathy: Past, present and future. J Urol. 173:2207–2208. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Ingraham SE and Mchugh KM: Current perspectives on congenital obstructive nephropathy. Pediatr Nephrol. 26:1453–1461. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Chevalier RL, Thornhill BA, Forbes MS and Kiley SC: Mechanisms of renal injury and progression of renal disease in congenital obstructive nephropathy. Pediatr Nephrol. 25:687–697. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Liapis H: Biology of congenital obstructive nephropathy. Nephron Exp Nephrol. 93:e87–e91. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Chevalier RL: Prognostic factors and biomarkers of congenital obstructive nephropathy. Pediatr Nephrol. 31:1411–1420. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Hahn H, Ku SE, Kim KS, Park YS, Yoon CH and Cheong HI: Implication of genetic variations in congenital obstructive nephropathy. Pediatr Nephrol. 20:1541–1544. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Grandaliano G, Gesualdo L, Bartoli F, Ranieri E, Monno R, Leggio A, Paradies G, Caldarulo E, Infante B and Schena FP: MCP-1 and EGF renal expression and urine excretion in human congenital obstructive nephropathy. Kidney Int. 58:182–192. 2000. View Article : Google Scholar : PubMed/NCBI

8 

Inazaki K, Kanamaru Y, Kojima Y, Sueyoshi N, Okumura K, Kaneko K, Yamashiro Y, Ogawa H and Nakao A: Smad3 deficiency attenuates renal fibrosis, inflammation, and apoptosis after unilateral ureteral obstruction. Kidney Int. 66:597–604. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Hermens JS, Thelen P, Ringert RH and Seseke F: Alterations of selected genes of the Wnt signal chain in rat kidneys with spontaneous congenital obstructive uropathy. J Pediatr Urol. 3:86–95. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Chevalier RL: Promise for gene therapy in obstructive nephropathy. Kidney Int. 66:1709–1710. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Chevalier RL: Obstructive nephropathy: Towards biomarker discovery and gene therapy. Nat Clin Pract Nephrol. 2:157–168. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Becknell B, Carpenter AR, Allen JL, Wilhide ME, Ingraham SE, Hains DS and McHugh KM: Molecular basis of renal adaptation in a murine model of congenital obstructive nephropathy. PLoS One. 8:e727622013. View Article : Google Scholar : PubMed/NCBI

13 

Carpenter AR, Becknell B, Ingraham SE and McHugh KM: Ultrasound imaging of the murine kidney. Methods Mol Biol. 886:403–410. 2012. 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 

Gautier L, Cope L, Bolstad BM and Irizarry RA: affy-analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 20:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Smyth GK: Limma: Linear models for microarray data. In: Bioinformatics and Computational Biology Solutions Using R and BioconductorStatistics for Biology and Health. Gentleman R, Carey VJ, Huber W, Irizarry RA and Dudoit S: Springer; New York, NY: pp. 397–420. 2005, View Article : Google Scholar

17 

Oliveros JC: Venny. An interactive tool for comparing lists with Venn Diagrams. BioinfoGP of CNB-CSIC. http://bioinfogp.cnnb.csic.es/tools/venny/index.htJuly 1–2017

18 

da 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 : PubMed/NCBI

19 

Dennis G Jr, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC and Lempicki RA: DAVID: Database for annotation, visualization, and integrated discovery. Genome Biol. 4:P32003. View Article : Google Scholar : PubMed/NCBI

20 

Gene Ontology Consortium: The gene ontology in 2010: Extensions and refinements. Nucleic Acids Res. 38:(Database Issue). D331–D335. 2010. View Article : Google Scholar : PubMed/NCBI

21 

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:(Database Issue). D447–D452. 2015. View Article : Google Scholar : PubMed/NCBI

22 

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

23 

Bandettini WP, Kellman P, Mancini C, Booker OJ, Vasu S, Leung SW, Wilson JR, Shanbhag SM, Chen MY and Arai AE: MultiContrast Delayed Enhancement (MCODE) improves detection of subendocardial myocardial infarction by late gadolinium enhancement cardiovascular magnetic resonance: A clinical validation study. J Cardiovasc Magn Reson. 14:832012. View Article : Google Scholar : PubMed/NCBI

24 

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 : PubMed/NCBI

25 

Janky R, Verfaillie A, Imrichová H, Van de Sande B, Standaert L, Christiaens V, Hulselmans G, Herten K, Sanchez Naval 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

26 

Davis AP, Grondin CJ, Lennon-Hopkins K, Saraceni-Richards C, Sciaky D, King BL, Wiegers TC and Mattingly CJ: The comparative toxicogenomics database's 10th year anniversary: Update 2015. Nucleic Acids Res. 43:(Database Issue). D914–D920. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Arlt VM, Zuo J, Trenz K, Roufosse CA, Lord GM, Nortier JL, Schmeiser HH, Hollstein M and Phillips DH: Gene expression changes induced by the human carcinogen aristolochic acid I in renal and hepatic tissue of mice. Int J Cancer. 128:21–32. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Jin K, Su KK, Li T, Zhu XQ, Wang Q, Ge RS, Pan ZF, Wu BW, Ge LJ, Zhang YH, et al: Hepatic premalignant alterations triggered by human nephrotoxin aristolochic acid I in canines. Cancer Prev Res (Phila). 9:324–334. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Ismail E, Al-Mulla F, Tsuchida S, Suto K, Motley P, Harrison PR and Birnie GD: Carbonyl reductase: A novel metastasis-modulating function. Cancer Res. 60:1173–1176. 2000.PubMed/NCBI

30 

Gao S, Tang K, Zhang J, Yang Z, Cui H, Li P, Tang H and Zhou M: Department of Orthopedics, Orthopedic Center of Chinese PLA, Southwest Hospital, Third Military Medical University; Department of Neurobiology, Third Military Medical University: Effect of cyclic stretch on expression of c-fos gene in rat Achilles-derived tendon stem cells. Chin J Rep Reconstr Surg. 2017.

31 

Ge R, Wang Z, Zeng Q, Xu X and Olumi AF: F-box protein 10, an NF-κB-dependent anti-apoptotic protein, regulates TRAIL-induced apoptosis through modulating c-Fos/c-FLIP pathway. Cell Death Differ. 18:1184–1195. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Otsuka M, Hatakenaka M, Ishigami K and Masuda K: Expression of the c-myc and c-fos genes as a potential indicator of late radiation damage to the kidney. Int J Radiat Oncol Biol Phys. 49:169–173. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Zhou F, Zhang L, Su Y, Zhang J and An Z: Inhibitory effect of artemisinin on the upregulation of c-fos and c-jun gene expression in kidney tissue of diabetic rats. Modern Journal of Integrated Traditional Chinese & Western Medicine. 23:2294–2295. 2014.

34 

Yazdi AS and Ghoreschi K: The interleukin-1 family. Adv Exp Med Biol. 941:21–29. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Rusai K, Huang H, Sayed N, Strobl M, Roos M, Schmaderer C, Heemann U and Lutz J: Administration of interleukin-1 receptor antagonist ameliorates renal ischemia-reperfusion injury. Transpl Int. 21:572–580. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Pindjakova J, Hanley SA, Duffy MM, Sutton CE, Weidhofer GA, Miller MN, Nath KA, Mills KH, Ceredig R and Griffin MD: Interleukin-1 accounts for intrarenal Th17 cell activation during ureteral obstruction. Kidney Int. 81:379–390. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Wajant H, Pfizenmaier K and Scheurich P: Tumor necrosis factor signaling. Cell Death Differ. 10:45–65. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Meldrum K, Misseri R, Rink R and Meldrum D: Tumor necrosis factor alpha (TNF) mediates renal tubular cell apoptosis during obstructive uropathy. J Am Coll Surg. 197:S912003.

39 

Awad AS, You H, Gao T, Cooper TK, Nedospasov SA, Vacher J, Wilkinson PF, Farrell FX and Reeves Brian W: Macrophage-derived tumor necrosis factor-α mediates diabetic renal injury. Kidney Int. 88:722–733. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Shu KH, Lee SH, Cheng CH, Wu MJ and Lian JD: Impact of interleukin-1 receptor antagonist and tumor necrosis factor-alpha gene polymorphism on IgA nephropathy. Kidney Int. 58:783–789. 2000. View Article : Google Scholar : PubMed/NCBI

41 

World Health Organization, . Stuart MC, Kouimtzi M and Hill S: WHO model formulary 2008. World Health Organization; Geneva: 2009

42 

Sattarinezhad E, Panjehshahin MR, Torabinezhad S, Kamali-Sarvestani E, Farjadian S, Pirsalami F and Moezi L: Protective effect of edaravone against cyclosporine-induced chronic nephropathy through antioxidant and nitric oxide modulating pathways in rats. Iran J Med Sci. 42:170–178. 2017.PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 18 Issue 3

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
Xin G, Chen R and Zhang X: Candidate gene and mechanism investigations in congenital obstructive nephropathy based on bioinformatics analysis. Mol Med Rep 18: 2651-2660, 2018
APA
Xin, G., Chen, R., & Zhang, X. (2018). Candidate gene and mechanism investigations in congenital obstructive nephropathy based on bioinformatics analysis. Molecular Medicine Reports, 18, 2651-2660. https://doi.org/10.3892/mmr.2018.9284
MLA
Xin, G., Chen, R., Zhang, X."Candidate gene and mechanism investigations in congenital obstructive nephropathy based on bioinformatics analysis". Molecular Medicine Reports 18.3 (2018): 2651-2660.
Chicago
Xin, G., Chen, R., Zhang, X."Candidate gene and mechanism investigations in congenital obstructive nephropathy based on bioinformatics analysis". Molecular Medicine Reports 18, no. 3 (2018): 2651-2660. https://doi.org/10.3892/mmr.2018.9284