Open Access

Identification of key genes for diabetic kidney disease using biological informatics methods

  • Authors:
    • Fuzhe Ma
    • Tao Sun
    • Meiyan Wu
    • Wanning Wang
    • Zhonggao Xu
  • View Affiliations

  • Published online on: September 29, 2017     https://doi.org/10.3892/mmr.2017.7666
  • Pages: 7931-7938
  • Copyright: © Ma 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

Diabetic kidney disease (DKD) is a common complication of diabetes, which is characterized by albuminuria, impaired glomerular filtration rate or a combination of the two. The aim of the present study was to identify the potential key genes involved in DKD progression and to subsequently investigate the underlying mechanism involved in DKD development. The array data of GSE30528 including 9 DKD and 13 control samples was downloaded from the Gene Expression Omnibus database. The differentially expressed genes (DEGs) in DKD glomerular and tubular kidney biopsy tissues were compared with normal tissues, and were analyzed using the limma package. Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed for DEGs using the GO Function software in Bioconductor. The protein‑protein interaction (PPI) network was then constructed using Cytoscape software. A total of 426 genes (115 up‑ and 311 downregulated) were differentially expressed between the DKD and normal tissue samples. The PPI network was constructed with 184 nodes and 335 edges. Vascular endothelial growth factor A (VEGFA), α‑actinin‑4 (ACTN4), proto‑oncogene, Src family tyrosine kinase (FYN), collagen, type 1, α2 (COL1A2) and insulin‑like growth factor 1 (IGF1) were hub proteins. Major histocompatibility complex, class II, DP α1 (HLA‑DPA1) was the common gene enriched in the rheumatoid arthritis and systemic lupus erythematosus pathways, and the immune response was a GO term enriched in module A. VEGFA, ACTN4, FYN, COL1A2, IGF1 and HLA‑DPA1 may be potential key genes associated with the progression of DKD, and immune mechanisms may serve a part in DKD development.

Introduction

Diabetic kidney disease (DKD) is a common complication of diabetes, which is characterized by albuminuria, an impaired glomerular filtration rate (GFR) or a combination of the two (1,2). DKD accounts for ~50% of all cases of end-stage renal disease (ESRD) in the United States and the US ESRD program is a large medical expense/economic burden and costs a great amount of money to run. However, a number of genes closely associated with DKD development have yet to be identified despite many years of intensive study (3). Therefore, the identification of genes associated with DKD development is urgently required, as well as the subsequent elucidation of its molecular mechanism.

Some advancements have been made in the elucidation of the pathological mechanism involved in the development of DKD. The loss of podocytes is an early feature of DKD (4). The levels of almost all podocyte-specific genes including genes for congenital nephrotic syndrome of the finish type (NPHS1), glomerular podocin (NPHS2), the Wilm's tumor gene (WT1) and vascular endothelial growth factor (VEGF) are all severely reduced in DKD (3). Some other studies have also demonstrated that NPHS1 (5,6), NPHS2 (7), bone morphogenetic protein 7 (8), WT1 (4) and VEGF (9,10) are decreased in DKD. In addition, tubulointerstitial fibrosis is a prominent feature of progressive DKD and is likely to be one of the final common pathways leading to the development of ESRD, with patients subsequently requiring dialysis or transplantation (11,12). A previous study revealed that using angiotensin-converting-enzyme-inhibitors and angiotensin II receptor antagonists in patients with diabetes mellitus can respectively improve mortality and delay the progression of DKD (13). In addition, a human genetic study highlighted that the complement system potentially serves a role in low-grade inflammation and the development of DKD (14). Therefore, the aim of the present study was to identify the key genes associated with the development of DKD and elucidate its underlying mechanism.

In the present study, the microarray data of GSE30528 was downloaded the from Gene Expression Omnibus database (GEO; www.ncbi.nlm.nih.gov/geo/). The gene expression profiles in DKD were analyzed and functional analysis was performed for differentially expressed genes (DEGs) in DKD glomerular and tubular kidney biopsy tissues in comparison with normal tissues. In addition, the protein-protein interaction (PPI) network was also constructed. These results were used to discover the key genes associated with DKD development and to clarify the underlying mechanism.

Materials and methods

Affymetrix microarray data

The array data for GSE30528 was downloaded from the GEO database, which was first recorded by Woroniecka et al (3) and was based on the GPL571 platform (Affymetrix Human Genome U133A 2.0 Array; Affymetrix, Inc.; Thermo Fisher Scientific, Inc., Waltham, MA, USA). A total of 44 samples were used to develop the original array data, and of these 9 DKD [age, 64±13.56 years; 5 females, 4 males; body mass index (BMI), 32.74±7.9 kg/m2] and 13 healthy, disease-free control samples (age, 51.38±12.01 years; 5 females, 8 males; BMI, 29.59±9.08 kg/m2) were selected for analysis in the present study.

Data processing and DEG analysis

The raw expression data was preprocessed using the robust multiarray average algorithm (15) and the Affy package in Bioconductor (bioconductor.org/packages/release/bioc/html/affy.html); the expression levels of the probes were then obtained. If several probes mapped to one gene symbol, then the mean value was set as the final expression value of this gene. The DEGs in DKD glomerular and tubular kidney biopsy tissues where then compared with normal tissues using the limma package (16). |logFC| >1 and P<0.05 were considered as the cutoff criterion.

Gene Ontology (GO) and pathway enrichment analysis

GO is used for the unification of biology, collecting defined, structured and controlled vocabulary for gene annotation, which mainly includes the following 3 categories: Molecular function, biological process and cellular component (17). The Kyoto Encyclopedia of Genes and Genomes (KEGG) is a database for the classification of relevant gene sets into their respective pathways (18).

In order to analyze the DEGs on a functional level, GO annotation and KEGG pathway enrichment analyses were performed for DEGs using GO Function version 1.24.0 (19) software in Bioconductor version 3.5 (www.bioconductor.org/packages/release/bioc/html/GOFunction.html), and gene annotation information was obtained from the org. Hs. eg. db and GO. db package. P<0.05 and gene counts >2 were set as the cut off values.

PPI network analysis

The Search Tool for the Retrieval of Interacting Genes (STRING) database provides the experimental and predicted interaction information of proteins (20). Protein pair interactions in STRING were presented with a combined score. The DEGs were mapped into PPIs and a combined score of >0.7 was identified as the cutoff standard for the important protein pairs. The PPI network was constructed using Cytoscape software version 2.8.2 (www.cytoscape.org/) (21).

Module analysis

ClusterONE version 1.0 (www.paccanarolab.org/cluster-one/) in the Cytoscape software package was used to analyze the PPI network modules with a minimum size of 3 and a minimum density of 0.5. Modules with P<0.01 were set as significant clustering modules.

Results

Data processing and DEGs analysis

As shown in Fig. 1, a total of 426 genes were differentially expressed in DKD samples when compared with normal samples, amongst which 115 genes were upregulated and 311 were downregulated.

GO and pathway enrichment analysis

GO and KEGG pathway analyses were performed for upregulated and downregulated DEGs. The top 5 GO terms are shown in Table I. The overrepresented GO terms of upregulated DEGs were primarily associated with extracellular region, antigen binding, extracellular space, the defense response, the immune response and peptidase regulator activity (Table IA). The downregulated DEGs were mainly involved in cardiovascular system development, circulatory system development, actin cytoskeleton, cell junction, cytoskeletal protein binding and integrin binding (Table IB).

Table I.

Gene Ontology analysis for differentially expressed genes.

Table I.

Gene Ontology analysis for differentially expressed genes.

A, Upregulated

TermDescriptionCounts (n) P-valuea
GO-BP terms
  GO:0006952Defense response  44<0.0005
  GO:0006955Immune response  41 2.22×10−16
  GO:0002376Immune system process  50 3.00×10−15
  GO:0001775Cell activation  31 1.37×10−14
  GO:0045321Leukocyte activation  26 1.76×10−13
GO-CC terms
  GO:0005576Extracellular region  63 2.06×10−12
  GO:0005615Extracellular space  32 4.37×10−12
  GO:0031982Vesicle  53 9.90×10−11
  GO:0031988Membrane-bounded vesicle  52 1.13×10−10
  GO:0044421Extracellular region part  53 3.50×10−10
GO-MF terms
  GO:0003823Antigen binding     8 2.97×10−07
  GO:0061134Peptidase regulator activity  10 6.67×10−07
  GO:0005539Glycosaminoglycan binding     9 2.84×10−07
  GO:0004866Endopeptidase inhibitor activity     8
8092×10−06
  GO:0061135Endopeptidase regulator activity     8 1.11×10−05

B, Downregulated

TermDescriptionCounts (n) P-valuea

GO-BP terms
  GO:0072358Cardiovascular system development  50 3.77×10−15
  GO:0072359Circulatory system development  50 3.77×10−15
  GO:0009653Anatomical structure morphogenesis  91 4.66×10−15
  GO:0048731System development121 3.40×10−14
  GO:0032502Developmental process147 5.66×10−14
GO-CC terms
  GO:0015629Actin cytoskeleton  31 1.79×10−12
  GO:0030054Cell junction  47 6.04×10−10
  GO:0070161Anchoring junction  28 1.82×10−09
  GO:0005912Adherens junction  27 3.39×10−09
  GO:0044421Extracellular region part  98 3.00×10−08
GO-MF terms
  GO:0008092Cytoskeletal protein binding  40 9.40×10−11
  GO:0005178Integrin binding  12 1.23×10−07
  GO:0032403Protein complex binding  35 4.24×10−07
  GO:0050839Cell adhesion molecule binding  14 8.04×10−07
  GO:0003779Actin binding  21 1.40×10−06

a P<0.00001 vs. normal matched tissues. GO, Gene Ontology; BP, biological process; CC, cellular component; MF, molecular function; Term, the identification number of GO; Description, the name of the GO term; Counts, the number of genes enriched in the GO term.

The upregulated DEGs were mainly enriched in 17 KEGG pathways, including primary immunodeficiency, extracellular matrix-receptor interactions, rheumatoid arthritis and systemic lupus erythematosus (Table IIA). In addition, major histocompatibility complex, class II, DP α1 (HLA-DPA1) was the common gene in the rheumatoid arthritis and systemic lupus erythematosus pathways. The downregulated DEGs were mainly enriched in 16 KEGG pathways, such as tight junction and adherens junction.

Table II.

Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis of the top 10 DEGs.

Table II.

Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis of the top 10 DEGs.

A, Upregulated

TermDescriptionCounts (n)P-value
5150Staphylococcus aureus infection  5 1.89×10−04c
5020Prion diseases  4 3.58×10−04c
5340Primary immunodeficiency  4 3.58×10−04c
4512Extracellular matrix-receptor interaction  5 1.42×10−03b
5323Rheumatoid arthritis  5 1.92×10−03b
5142Chagas disease (American trypanosomiasis)  5 3.45×10−03b
4610Complement and coagulation cascades  4 4.61×10−03b
4974Protein digestion and absorption  4 8.12×10−03b
5322Systemic lupus erythematosus  5 1.06×10−02a
4640Hematopoietic cell lineage  4 1.08×10−02a

B, Downregulated

TermDescriptionCounts (n)P-value

4520Adherens junction  8 3.85×10−05d
4510Focal adhesion13 4.51×10−05d
4810Regulation of actin cytoskeleton13 8.65×10−05d
4530Tight junction  9 5.06×10−04c
5410Hypertrophic cardiomyopathy  7 6.17×10−04c
5414Dilated cardiomyopathy  7 1.00×10−03b
5412Arrhythmogenic right ventricular cardiomyopathy  6 1.87×10−03b
4610Complement and coagulation cascades  5 7.26×10−03b
4360Axon guidance  7 7.65×10−03b
5200Pathways in cancer12 1.21×10−02a

a P<0.05

b P<0.01

c P<0.001

d P<0.0001 vs. normal matched tissues. KEGG, Kyoto Encyclopedia of Genes and Genomes; Term, the identification number of the KEGG pathway; Description, the name of the KEGG pathway; Counts, the number of genes enriched in the KEGG pathway.

PPI network analysis

Based on the STRING database, a total of 335 protein pairs with a combined score of >0.7 were obtained. As presented in Fig. 2, the PPI network was constructed with 335 edges and 184 nodes. The nodes of VEGFA (degree score, 19), α-actinin-4 (ACTN4; degree score, 17), proto-oncogene, Src family tyrosine kinase (FYN; degree score, 17), collagen, type 1, α2 (COL1A2; degree score, 15) and insulin-like growth factor 1 (IGF1; degree score, 15) were hub proteins in the network.

Modules analysis

Two significant clustering modules were obtained using ClusterONE software (Fig. 3). A total of 8 and 6 nodes were enriched in modules A and B, respectively. As shown in Table III, nodes in module A (density, 1.0; quality, 0.800; P=1.606×10−4) were mainly enriched in GO: G-protein coupled receptor protein signaling pathway, cell surface receptor linked signal transduction, the immune response, the chemokine signaling pathway and the cytokine-cytokine receptor interaction pathway. Nodes in module B (density, 1.0; quality, 0.789; P=0.001) were mainly enriched in GO: regulation of ATPase activity, regulation of system process and regulation of hydrolase activity.

Table III.

Functional enrichment analysis of protein-protein interaction network clustering modules.

Table III.

Functional enrichment analysis of protein-protein interaction network clustering modules.

TermDescriptionCounts (n)P-value
Module A
  GO_BP:0007186G-protein coupled receptor protein signaling pathway8 2.67×10−08a
  GO_BP:0007166Cell surface receptor linked signal transduction8 9.06×10−07a
  GO_BP:0006955Immune response5 2.08×10−04b
  KEGG_ hsa04062Chemokine signaling pathway3 1.25×10−02c
  KEGG_ hsa04060Cytokine-cytokine receptor interaction3 2.38×10−02c
Module B
  GO_BP:0043462Regulation of ATPase activity3 1.31×10−05d
  GO_BP:0044057Regulation of system process3 4.97×10−03e
  GO_BP:0051336Regulation of hydrolase activity3 5.89×10−03f
  KEGG_ hsa04260Cardiac muscle contraction2 2.32×10−04b
  KEGG_ hsa05410Hypertrophic cardiomyopathy3 2.76×10−04b
  KEGG_ hsa05414Dilated cardiomyopathy2 3.24×10−04b

a P<0.00001

b P<0.0005

c P<0.05

d P<0.0001

e P<0.005

f P<0.01vs. normal matched tissues. GO, Gene Ontology; KEGG, KEGG, Kyoto Encyclopedia of Genes and Genomes; Term, the identification number of GO-Biological Process or KEGG pathway; Description, the name of the GO- Biological Process or KEGG pathway; Counts, the number of genes enriched in GO- Biological Process or KEGG pathway.

Discussion

In the present study, using the gene expression patterns downloaded from the GEO database, 426 DEGs in DKD glomerular and tubular kidney biopsy tissues were obtained and compared with matched normal tissues, identifying 115 upregulated genes and 311 downregulated DEGs. The results demonstrated that HLA-DPA1 was the common gene enriched in the rheumatoid arthritis and systemic lupus erythematosus pathways, and the immune response was a significant GO term enriched in module A. In addition, VEGFA, ACTN4, FYN, COL1A2 and IGF1 had higher degrees and were established as hub nodes in the PPI network; they may therefore contribute to the progression of DKD.

A previous study suggested that cells in the immune system may be involved in the progression of DKD (22). Immune cells take part in vascular injury under DKD-associated conditions (23). Other previous studies have also indicated that the immune system is associated with DKD development (2426). Primary immunodeficiency (27), rheumatoid arthritis (28) and systemic lupus erythematosus (29) are associated with the immune system. In the present study, primary immunodeficiency, rheumatoid arthritis and systemic lupus erythematosus were 3 significantly enriched pathways, and the immune response was a GO term enriched in module A. Thus, the results of the present study are in agreement with previous findings, and therefore indicate that immune mechanisms may serve a role in DKD development.

The work of Woroniecka et al (3) suggested that HLA-DPA1 was a differentially expressed transcript in the tubulointerstitium of patients with DKD when compared with normal samples. Previous studies revealed that HLA-DPA1, which is the closest centromeric gene expressed to HLA-DOα, may contribute to the differences in the associated risks of diabetes (30,31), including DKD, which is a complication of diabetes. In the present study, HLA-DPA1 was the common gene enriched in the rheumatoid arthritis and systemic lupus erythematosus pathways. Therefore, these results are in line with previous findings and suggest that HLA-DPA1 may contribute to DKD development.

In addition, VEGFA, ACTN4, FYN, COL1A2 and IGF1 were identified as hub proteins in the PPI network. VEGFA is an important angiogenic growth factor that regulates endothelial cells' permeability and vasculogenesis (32). It is also important for the differentiation, proliferation, survival and migration of endothelial cells within the glomerulus (33). Previous studies have suggested that VEGFA may serve a significant role in retaining glomerular endothelial cell function as a reduction in VEGFA levels induced abnormal remodeling of glomerular capillaries (34,35). VEGF may also serve a role in the pathogenesis of DKD (36) and the dysregulation of VEGFA may serve a pathogenic role in inducing glomerular injury (37). In DKD, VEGFA has reduced mRNA expression and may be a potential factor that can lead to the development of DKD by inducing microvascular rarefaction and tubular atrophy (9). In addition, neoangiogenesis, which is caused by overexpression of VEGFA, may stimulate the development of DKD and therefore blocking VEGFA or its signaling may ameliorate DKD (38). These findings indicate that VEGFA may serve a role in DKD progression.

ACTNs are actin-binding proteins that are critical in cell adhesion and in the organization of the cytoskeleton (39). Increasing evidence has revealed that in diabetes, there are cytoskeletal changes in podocytes. For instance, advanced glycosylation end products and high glucose can decrease the expression of ACTN4 (40), and a reduced expression of ACTN4 may lead to proteinuria (a symptom of DKD) (41). In addition, FYN is a tyrosine-specific phospho-transferase that belongs to the Src family of tyrosine protein kinases (42). FYN phosphorylation is transiently stimulated by high glucose levels (43). Src/FYN kinase inhibitors disrupt signaling molecules in the VEGF signal transduction pathway (44), and as mentioned above, VEGF may be associated with DKD; thus, FYN may in turn be involved in DKD. In addition, the accumulation of extracellular matrix proteins such as COL1A2 is a key feature of DKD (45). A previous report demonstrated that some key microRNAs (miR) act as effectors of transforming growth factor (TGF)-β and the actions of high glucose in DKD (46). In mesangial cells and the kidney, experimental diabetes was associated with the increased expression of COL1A2, and miR-192 was increased by TGF-β treatment (45). IGF1 as a growth factor receptor has been associated with type 1 DKD (47). In addition, IGF-1 has the capacity to mediate the histological changes characteristic of DKD (48). These previous studies all indicate that these proteins are associated with DKD. Therefore, the results of the present study are in agreement with these findings and provide further evidence that VEGFA, ACTN4, FYN, COL1A2 and IGF1 may serve important roles in DKD development directly or indirectly.

In conclusion, the results of the present study indicated that in addition to VEGFA, ACTN4, FYN, COL1A2, IGF1 and HLA-DPA1, immune mechanisms may also serve an important role in the development of DKD. These genes may serve as target genes for the treatment of DKD in future clinical practice. However, this conclusion has no experimental verification; therefore, further evaluation of the potential applications in clinical practice is required.

Acknowledgements

The present study was supported by the National Natural Science Foundation of China (grant nos. 81070578 and 81270809).

References

1 

Association AD: Standards of medical care in diabetes-2010. Diabetes care. 33 Suppl:S11–S61. 2010. View Article : Google Scholar :

2 

Levin A and Rocco M: KDOQI clinical practice guidelines and clinical practice recommendations for diabetes and chronic kidney disease. Am J Kidney Dis. 49(2 Suppl 2): S12–S154. 2007.

3 

Woroniecka KI, Park AS, Mohtat D, Thomas DB, Pullman JM and Susztak K: Transcriptome analysis of human diabetic kidney disease. Diabetes. 60:2354–2369. 2011. View Article : Google Scholar :

4 

Susztak K, Raff AC, Schiffer M and Böttinger EP: Glucose-induced reactive oxygen species cause apoptosis of podocytes and podocyte depletion at the onset of diabetic nephropathy. Diabetes. 55:225–233. 2006. View Article : Google Scholar

5 

Langham RG, Kelly DJ, Cox AJ, Thomson NM, Holthöfer H, Zaoui P, Pinel N, Cordonnier DJ and Gilbert RE: Proteinuria and the expression of the podocyte slit diaphragm protein, nephrin, in diabetic nephropathy: Effects of angiotensin converting enzyme inhibition. Diabetologia. 45:1572–1576. 2002. View Article : Google Scholar

6 

Doublier S, Salvidio G, Lupia E, Ruotsalainen V, Verzola D, Deferrari G and Camussi G: Nephrin expression is reduced in human diabetic nephropathy: Evidence for a distinct role for glycated albumin and angiotensin II. Diabetes. 52:1023–1030. 2003. View Article : Google Scholar

7 

Koop K, Eikmans M, Baelde HJ, Kawachi H, de Heer E, Paul LC and Bruijn JA: Expression of podocyte-associated molecules in acquired human kidney diseases. J Am Soc Nephrol. 14:2063–2071. 2003. View Article : Google Scholar

8 

Turk T, Leeuwis JW, Gray J, Torti SV, Lyons KM, Nguyen TQ and Goldschmeding R: BMP signaling and podocyte markers are decreased in human diabetic nephropathy in association with CTGF overexpression. J Histochem Cytochem. 57:623–631. 2009. View Article : Google Scholar :

9 

Lindenmeyer MT, Kretzler M, Boucherot A, Berra S, Yasuda Y, Henger A, Eichinger F, Gaiser S, Schmid H, Rastaldi MP, et al: Interstitial vascular rarefaction and reduced VEGF-A expression in human diabetic nephropathy. J Am Soc Nephrol. 18:1765–1776. 2007. View Article : Google Scholar

10 

Hohenstein B, Hausknecht B, Boehmer K, Riess R, Brekken RA and Hugo CP: Local VEGF activity but not VEGF expression is tightly regulated during diabetic nephropathy in man. Kidney Int. 69:1654–1661. 2006. View Article : Google Scholar

11 

Nath KA: The tubulointerstitium in progressive renal disease. Kidney Int. 54:992–994. 1998. View Article : Google Scholar

12 

Gilbert RE and Cooper ME: The tubulointerstitium in progressive diabetic kidney disease: More than an aftermath of glomerular injury? Kidney Int. 56:1627–1637. 1999. View Article : Google Scholar

13 

Strippoli GF, Craig M, Deeks JJ, Schena FP and Craig JC: Effects of angiotensin converting enzyme inhibitors and angiotensin II receptor antagonists on mortality and renal outcomes in diabetic nephropathy: Systematic review. BMJ. 329:8282004. View Article : Google Scholar :

14 

Hansen TK, Forsblom C, Saraheimo M, Thorn L, Wadén J, Høyem P, Østergaard J, Flyvbjerg A and Groop PH; FinnDiane Study Group, : Association between mannose-binding lectin, high-sensitivity C-reactive protein and the progression of diabetic nephropathy in type 1 diabetes. Diabetologia. 53:1517–1524. 2010. 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 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Altermann E and Klaenhammer TR: PathwayVoyager: Pathway mapping using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. BMC Genomics. 6:602005. View Article : Google Scholar : PubMed/NCBI

19 

Wang J, Zhou X, Zhu J, Gu Y, Zhao W, Zou J and Guo Z: GO-function: Deriving biologically relevant functions from statistically significant functions. Brief Bioinform. 13:216–227. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Szklarczyk D, Franceschini A, Kuhn M, Simonovic M, Roth A, Minguez P, Doerks T, Stark M, Muller J, Bork P, et al: The STRING database in 2011: Functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res. 39:D561–D568. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Kohl M, Wiese S and Warscheid B: Cytoscape: Software for visualization and analysis of biological networks. Methods Mol Biol. 696:291–303. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Ichinose K, Kawasaki E and Eguchi K: Recent advancement of understanding pathogenesis of type 1 diabetes and potential relevance to diabetic nephropathy. Am J Nephrol. 27:554–564. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Galkina E and Ley K: Leukocyte recruitment and vascular injury in diabetic nephropathy. J Am Soc Nephrol. 17:368–377. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Chow F, Ozols E, Nikolic-Paterson DJ, Atkins RC and Tesch GH: Macrophages in mouse type 2 diabetic nephropathy: Correlation with diabetic state and progressive renal injury. Kidney Int. 65:116–128. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Tuttle KR: Linking metabolism and immunology: Diabetic nephropathy is an inflammatory disease. J Am Soc Nephrol. 16:1537–1538. 2005. View Article : Google Scholar : PubMed/NCBI

26 

Mora C and Navarro JF: Inflammation and diabetic nephropathy. Curr Diab Rep. 6:463–468. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Johnston SL, Virgo PF and Unsworth DJ: Type 1 diabetes mellitus masking primary antibody deficiency. J Clin Pathol. 53:236–237. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Prevoo ML, Van't Hof MA, Kuper HH, Van Leeuwen MA, Van de Putte LB and Van Riel PL: Modified disease activity scores that include twenty-eight-joint counts, development and validation in a prospective longitudinal study of patients with rheumatoid arthritis. Arthritis Rheum. 38:44–48. 1995. View Article : Google Scholar : PubMed/NCBI

29 

Feng PH: Systemic lupus erythematosus: the face of Asia. Ann N Y Acad Sci. 1108:114–120. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Varney MD, Valdes AM, Carlson JA, Noble JA, Tait BD, Bonella P, Lavant E, Fear AL, Louey A, Moonsamy P, et al: HLA DPA1, DPB1 alleles and haplotypes contribute to the risk associated with type 1 diabetes: Analysis of the type 1 diabetes genetics consortium families. Diabetes. 59:2055–2062. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Santin I, Castellanos-Rubio A, Aransay AM, Gutierrez G, Gaztambide S, Rica I, Vicario JL, Noble JA, Castaño L and Bilbao JR: Exploring the diabetogenicity of the HLA-B18-DR3 CEH: Independent association with T1D genetic risk close to HLA-DOA. Genes Immun. 10:596–600. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Dvorak HF, Brown LF, Detmar M and Dvorak AM: Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis. Am J Pathol. 146:1029–1039. 1995.PubMed/NCBI

33 

Schrijvers BF, Flyvbjerg A and De Vriese AS: The role of vascular endothelial growth factor (VEGF) in renal pathophysiology. Kidney Int. 65:2003–2017. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Baelde HJ, Eikmans M, Doran PP, Lappin DW, de Heer E and Bruijn JA: Gene expression profiling in glomeruli from human kidneys with diabetic nephropathy. Am J Kidney Dis. 43:636–650. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Bortoloso E, Del Prete D, Vestra M Dalla, Gambaro G, Saller A, Antonucci F, Baggio B, Anglani F and Fioretto P: Quantitave and qualitative changes in vascular endothelial growth factor gene expression in glomeruli of patients with type 2 diabetes. Eur J Endocrinol. 150:799–807. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Cha DR, Kim NH, Yoon JW, Jo SK, Cho WY, Kim HK and Won NH: Role of vascular endothelial growth factor in diabetic nephropathy. Kidney Int Suppl. 77:S104–S112. 2000. View Article : Google Scholar : PubMed/NCBI

37 

Eremina V, Sood M, Haigh J, Nagy A, Lajoie G, Ferrara N, Gerber HP, Kikkawa Y, Miner JH and Quaggin SE: Glomerular-specific alterations of VEGF-A expression lead to distinct congenital and acquired renal diseases. J Clin Invest. 111:707–716. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Caldwell RB, Bartoli M, Behzadian MA, El-Remessy AE, Al-Shabrawey M, Platt DH and Caldwell RW: Vascular endothelial growth factor and diabetic retinopathy: Pathophysiological mechanisms and treatment perspectives. Diabetes Metab Res Rev. 19:442–455. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Nikolopoulos SN, Spengler BA, Kisselbach K, Evans AE, Biedler JL and Ross RA: The human non-muscle alpha-actinin protein encoded by the ACTN4 gene suppresses tumorigenicity of human neuroblastoma cells. Oncogene. 19:380–386. 2000. View Article : Google Scholar : PubMed/NCBI

40 

HA TS: High glucose and advanced glycosylated end-products affect the expression of alpha-actinin-4 in glomerular epithelial cells. Nephrology (Carlton). 11:435–441. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Jefferson JA, Shankland SJ and Pichler RH: Proteinuria in diabetic kidney disease: A mechanistic viewpoint. Kidney Int. 74:22–36. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Resh MD: Fyn, a Src family tyrosine kinase. Int J Biochem Cell Biol. 30:1159–1162. 1998. View Article : Google Scholar : PubMed/NCBI

43 

Taniguchi K, Xia L, Goldberg HJ, Lee KW, Shah A, Stavar L, Masson EA, Momen A, Shikatani EA, John R, et al: Inhibition of Src kinase blocks high glucose-induced EGFR transactivation and collagen synthesis in mesangial cells and prevents diabetic nephropathy in Mice. Diabetes. 62:3874–3886. 2013. View Article : Google Scholar : PubMed/NCBI

44 

van Nieuw Amerongen GP and van Hinsbergh VW: Targets for pharmacological intervention of endothelial hyperpermeability and barrier function. Vascular pharmacol. 39:257–272. 2002. View Article : Google Scholar

45 

Kato M, Zhang J, Wang M, Lanting L, Yuan H, Rossi JJ and Natarajan R: MicroRNA-192 in diabetic kidney glomeruli and its function in TGF-beta-induced collagen expression via inhibition of E-box repressors. Proc Natl Acad Sci USA. 104:pp. 3432–3437. 2007; View Article : Google Scholar : PubMed/NCBI

46 

Kato M, Arce L and Natarajan R: MicroRNAs and their role in progressive kidney diseases. Clin J Am Soc Nephrol. 4:1255–1266. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Ewens KG, George RA, Sharma K, Ziyadeh FN and Spielman RS: Assessment of 115 candidate genes for diabetic nephropathy by transmission/disequilibrium test. Diabetes. 54:3305–3318. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Schreiber B, Hughes M and Groggel G: Insulin-like growth factor-1 stimulates production of mesangial cell matrix components. Clin Nephrol. 43:368–374. 1995.PubMed/NCBI

Related Articles

Journal Cover

December-2017
Volume 16 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
Ma F, Sun T, Wu M, Wang W and Xu Z: Identification of key genes for diabetic kidney disease using biological informatics methods. Mol Med Rep 16: 7931-7938, 2017
APA
Ma, F., Sun, T., Wu, M., Wang, W., & Xu, Z. (2017). Identification of key genes for diabetic kidney disease using biological informatics methods. Molecular Medicine Reports, 16, 7931-7938. https://doi.org/10.3892/mmr.2017.7666
MLA
Ma, F., Sun, T., Wu, M., Wang, W., Xu, Z."Identification of key genes for diabetic kidney disease using biological informatics methods". Molecular Medicine Reports 16.6 (2017): 7931-7938.
Chicago
Ma, F., Sun, T., Wu, M., Wang, W., Xu, Z."Identification of key genes for diabetic kidney disease using biological informatics methods". Molecular Medicine Reports 16, no. 6 (2017): 7931-7938. https://doi.org/10.3892/mmr.2017.7666