Open Access

Identification of potential markers for type 2 diabetes mellitus via bioinformatics analysis

  • Authors:
    • Yana Lu
    • Yihang Li
    • Guang Li
    • Haitao Lu
  • View Affiliations

  • Published online on: June 26, 2020     https://doi.org/10.3892/mmr.2020.11281
  • Pages: 1868-1882
  • Copyright: © Lu 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

Type 2 diabetes mellitus (T2DM) is a multifactorial and multigenetic disease, and its pathogenesis is complex and largely unknown. In the present study, microarray data (GSE201966) of β‑cell enriched tissue obtained by laser capture microdissection were downloaded, including 10 control and 10 type 2 diabetic subjects. A comprehensive bioinformatics analysis of microarray data in the context of protein‑protein interaction (PPI) networks was employed, combined with subcellular location information to mine the potential candidate genes for T2DM and provide further insight on the possible mechanisms involved. First, differential analysis screened 108 differentially expressed genes. Then, 83 candidate genes were identified in the layered network in the context of PPI via network analysis, which were either directly or indirectly linked to T2DM. Of those genes obtained through literature retrieval analysis, 27 of 83 were involved with the development of T2DM; however, the rest of the 56 genes need to be verified by experiments. The functional analysis of candidate genes involved in a number of biological activities, demonstrated that 46 upregulated candidate genes were involved in ‘inflammatory response’ and ‘lipid metabolic process’, and 37 downregulated candidate genes were involved in ‘positive regulation of cell death’ and ‘positive regulation of cell proliferation’. These candidate genes were also involved in different signaling pathways associated with ‘PI3K/Akt signaling pathway’, ‘Rap1 signaling pathway’, ‘Ras signaling pathway’ and ‘MAPK signaling pathway’, which are highly associated with the development of T2DM. Furthermore, a microRNA (miR)‑target gene regulatory network and a transcription factor‑target gene regulatory network were constructed based on miRNet and NetworkAnalyst databases, respectively. Notably, hsa‑miR‑192‑5p, hsa‑miR‑124‑5p and hsa‑miR‑335‑5p appeared to be involved in T2DM by potentially regulating the expression of various candidate genes, including procollagen C‑endopeptidase enhancer 2, connective tissue growth factor and family with sequence similarity 105, member A, protein phosphatase 1 regulatory inhibitor subunit 1 A and C‑C motif chemokine receptor 4. Smad5 and Bcl6, as transcription factors, are regulated by ankyrin repeat domain 23 and transmembrane protein 37, respectively, which might also be used in the molecular diagnosis and targeted therapy of T2DM. Taken together, the results of the present study may offer insight for future genomic‑based individualized treatment of T2DM and help determine the underlying molecular mechanisms that lead to T2DM.

Introduction

Type 2 diabetes mellitus (T2DM) has become the third main chronic non-infectious disease following tumors and cardiovascular disease, and threatens human health worldwide (1). In total, ~425 million adults are currently living with diabetes in the world, with the majority of cases being T2DM (2). The International Diabetes Federation reported that in 2045, ~629 million individuals globally will suffer from diabetes, of which ~90% will be T2DM (2). It is well known that insulin resistance and pancreatic β-cell dysfunction are major pathophysiological characteristics of T2DM. Pancreatic β-cells are needed to yield more insulin to meet mounting requirements when insulin resistance occurs (3). Previous studies of pancreatic β-cells provide a basis for improved insight into the pathogenesis and pathophysiology of T2DM, as pancreatic β-cells help in the regulation of the blood glucose level (4,5). T2DM is a complex, polygenic disease that results from the interplay of environmental and genetic factors. Candidate gene association high-throughput methods have been carried out to uncover the genetic aspects of the pathogenesis of T2DM (68).

In recent years, single gene research and genome-wide association studies have determined genetic susceptibility genes for the increased risk of T2DM (911). Previous studies of gene expression in T2DM demonstrated that decreased expression of insulin (12,13), and a reduced expression of syntaxin 1A and transcription factor 7 like 2 contributed to impaired insulin secretion (12,14,15). The downregulation of FXYD domain containing ion transport regulator 2-stimulated β-cell proliferation (13) and the upregulation of genes δ like non-canonical Notch ligand 1, diacylglycerol kinase β and zinc finger MIZ-type containing 1 were implicated in T2DM (11,16). Previous genetic studies identified several dozen genes leading to monogenic diabetes due to impaired insulin secretion (17,18). These genes play a key role in pancreatic β-cell lineage, phenotype and function. How genetic and epigenetic factors are involved in β-cell development, proliferation, differentiation and function requires further investigation. Understanding of the underlying mechanisms is vital to the development of new therapeutic methods to prevent β-cell dysfunction and failure in the development of T2DM. The identification of T2DM candidate genes has been challenging in biomedical research and the majority of the genes have yet to be discovered. The aim of the present study was to contribute to research efforts to identify the biological markers and signaling pathways associated with T2DM. These molecular mechanisms may provide insight for aspects of T2DM pathogenesis or pathophysiology.

High-throughput sequencing is becoming an important tool, extensively applied in life sciences, including in cancer detection (1921) and for identifying global gene expression changes in T2DM (22). Knowledge of the subcellular localization of proteins provides new insight into protein function and the complex pathways that modulate biological processes on a sub-cellular level, contributing to the current understanding of the proteins that interact with each other and with other molecules in the cellular environment (23). Accordingly, subcellular proteomics, as an important step to functional proteomics, has been the focus of the prediction of subcellular protein location, which is associated with molecular cell biology, proteomics, systems biology and drug discovery (2426); it is used to better understand complex diseases (27), such as breast cancer (28), ovarian carcinoma (29), ischemic dilated cardiomyopathy (30), esophageal squamous cell carcinoma (31) and asthma (32). It was previously demonstrated that an integrative analysis of gene expression and a protein-protein interaction (PPI) network could offer insight of the molecular mechanisms of a variety of diseases (3335). Consequently, the present study proposed a comprehensive bioinformatics analysis of gene expression data combining protein subcellular localization information and the construction of a layered PPI network (as opposed to a traditional PPI network) to identify candidate genes. Functional enrichment analyses were performed for candidate genes. A microRNA (miRNA/miR)-target gene regulatory network and transcription factor (TF)-target gene regulatory network were also constructed to identify miRNAs and TFs, which could be involved in T2DM development. The findings of the present study may help in the discovery of potentially novel predictive and prognostic markers for T2DM, and provide insight into the underlying molecular mechanisms of T2DM.

Materials and methods

Data acquisition, preprocessing and differentially expressed genes (DEGs) analysis

GSE20966 (36), the gene chip datasets of β-cells acquired from cadaver pancreases of non-diabetic subjects (control group, n=10) and T2D subjects (T2D group, n=10), was assessed and obtained from the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo/). The annotation information of GeneChip was acquired from the GPL1352 Affymetrix Human X3P Array platform (Affymetrix; Thermo Fisher Scientific, Inc.). The probes were mapped to gene names based on the GPL1352 platform and the average expression value for the probes was calculated when there was more than one gene corresponding to the same probe. In the original gene expression profiles, after normalization, MATLAB 2018a (https://www.ilovematlab.cn/forum.php?mod=home) was used to identify the DEGs by value of a fold change >1.5 and a false discovery rate_Benjamini & Hochberg (fdr_BH) <0.1. The differences in gene expression between the control and T2DM subjects were assessed using hierarchical clustering and principal component analysis (PCA).

PPI network, layering and network analysis

The PPI data were retrieved from the Human Protein Reference Database v9.0 (HPRD) (37), BioGRID v3.5 (38), IntACT v4.2 (39) and STRING v10.5 (40) databases. First, single nodes, self-loops and duplicates were removed from the PPI data. Second, the total DEGs were mapped to the PPI data. To improve the reliability, only the direct interaction proteins of these DEGs were matched. Third, the integrated PPI network was visualized and analyzed using Cytoscape v3.6.1 (41). Then, the subcellular localization information for each protein in the integrated PPI network obtained from the HPRD, the UniProt database (http://www.uniprot.org/help/uniprotkb) and the Human Protein Atlas database (http://www.proteinatlas.org/) (42) was input as a node attribute. The Cerebral plug-in in Cytoscape was applied to redistribute nodes on the basis of subcellular localization without changing their interactions (43). The layered PPI network was split into five layers: Extracellular, plasma membrane, cytoplasm, nucleus and mitochondria. Hub protein nodes that encoded DEGs in the layered network with a connectivity degree >8 were screened as candidate genes.

Functional interpretations for the candidate genes

To investigate the functions of the candidate genes, functional enrichment analysis was performed using the ClueGO and the CluePedia plug-ins (44) for Cytoscape v3.6.1 software (45). ClueGO was used to decipher functionally grouped Gene Ontology (GO) (46) and pathway annotation networks to understand their implication in three different classifications; biological process (BP), molecular function (MF) and cell component (CC), in addition to the Kyoto Encyclopedia of Genes and Genomes (KEGG) (47) signaling pathway. The relationship between the terms was calculated using κ statistics and the ClueGO network was built based on the similarity of their related genes. The CluePedia plug-in is a search tool for new markers potentially associated to pathways, and can provide a broad viewpoint of a pathway using integrated experimental and in silico data. In the present study, the enrichment analysis of gene-BP and gene-pathway was statistically validated using the Cytoscape plug-ins ClueGO and CluePedia. BPs/signaling pathways were functionally split into several groups with κ score ≥0.4 and a network was constructed, where the node represents a BP/pathway and the edge between two nodes indicates that the two BPs/pathways share common genes.

Prediction of target miRNAs and TFs for the candidate genes

Genes need to interact to react to the environment of an organism, as they cannot alone to regulate the organism. Gene expression is modulated by TFs and miRNAs at the transcriptional and post-transcriptional levels. Information on TFs, miRNAs and their corresponding target genes could provide insight into the processes of T2DM. miRNet v2.0 (http://www.mirnet.ca/) (48) was used to predict the miRNAs associated with candidate genes noted in miRTarBase v7.6 (49) and miRecords (50). The 8 most captivating groups (top 15) and a minimum of two genes for each of the groups were picked as the threshold. Then, the TFs encoded by candidate genes were used for prediction coupled with human TF information (NetworkAnalyst v3.0; http://www.networkanalyst.ca) (51) noted in Binding and Expression Target Analysis v1.0.7 (BETA) (http://cistrome.org/BETA/) (52). The miRNA-target gene regulatory network and TF-target gene regulatory network were visualized using Cytoscape.

Results

Screening for DEGs

Following data preprocessing, 108 DEGs were identified to be differentially expressed in 10 control subjects and 10 T2DM subjects, with 66 upregulated and 42 downregulated genes, as presented in the heat map of the cluster analysis of DEGs, according to the cut-off criteria of fold change >1.5 and fdr_BH <0.1 (Fig. 1A; Table SI). The PCA plot demonstrated that the DEGs could roughly divide the majority of T2DM subjects from the non-diabetic controls (Fig. 1B).

PPI network, layering network construction and network analysis

The identification of proteins that interact directly with proteins encoded by the 108 DEGs could help understand the molecular mechanism underlying T2DM pathophysiology. In the present study, a PPI network was built from the 108 DEGs with Cytoscape and was composed of 1,546 nodes and 1,842 edges, including 52 proteins that were encoded by upregulated genes, 36 that were encoded by downregulated genes and 1,458 nodes marking proteins that were not encoded by DEGs (Fig. 2; Table I).

Table I.

Distribution of nodes.

Table I.

Distribution of nodes.

LocalizationUpregulatedDownregulatedUnchangedTotal
Extracellular135   132   150
Plasma membrane79   244   260
Cytoplasm218   427   456
Nucleus814   516   538
Mitochondrion30   139   142
Total52361,4581,546

Subcellular protein localization is a crucial process in numerous cells. Following synthesis, proteins are transported to distinct compartments depending on their molecular function within the cell. Certain proteins are even transported to distant sites. Protein localization data can contribute to the elucidation of protein functions. The subcellular localization information for each protein in the integrated PPI network obtained from the HPRD, the UniProt database (http://www.uniprot.org/help/uniprotkb) and the Human Protein Atlas database (http://www.proteinatlas.org/) (42), was input as a node attribute. Then, the layered network was created from the PPI network using the Cerebral plug-in (43) of Cytoscape, according to the subcellular localization information of 1,546 proteins, which was split into five layers as follows: Extracellular, plasma membrane, cytoplasm, nucleus and mitochondrion (Fig. 3).

The degree distribution of a network is a standard feature of scale-free networks. The degree distributions of the layered network closely followed the power law distribution, with an R2=0.865. This suggested that the integrated PPI network is a true cellular complex biological network and scale-free. The other topological parameters of the network are presented in Table II. The results also suggested that a small number of nodes are hubs with a number of links to nodes. A total of 83 DEGs were identified as hub genes with an interaction degree ≥8 and were selected as candidate genes (Table SII). The top 20 candidate genes are presented in Table III. Of these candidate genes, ISG15 ubiquitin like modifier (ISG15) had the highest degree (185), followed by phosphoenolpyruvate carboxykinase 1 (PCK1) (85) and neural precursor cell expressed, developmentally downregulated 9 (NEDD9) (50). The present study identified 83 candidate genes for T2DM (Table IV). The identified candidate genes may serve as biological markers for future T2DM treatment research.

Table II.

Topological parameters of network.

Table II.

Topological parameters of network.

ParametersValue
y=βxα y=73.313×−1.347
R20.865
Correlation0.946
Clustering coefficient0.2
Network centralization0.118
Network density0.002

Table III.

Top 20 hub genes.

Table III.

Top 20 hub genes.

No.Gene nameDegreeGene expression
1ISG15185Upregulated
2PCK185Upregulated
3NEDD950Downregulated
4TMSB4X44Upregulated
5SYT144Upregulated
6IGFBP341Downregulated
7NEFL41Upregulated
8TENC137Upregulated
9TIMM4436Upregulated
10HMMR35Upregulated
11ASB934Downregulated
12TRAF3IP234Downregulated
13KLF532Upregulated
14SERPINA331Upregulated
15NEFM30Upregulated
16PHC129Downregulated
17RASGRF129Downregulated
18SERPING128Upregulated
19MYCN27Downregulated
20SERPINA526Upregulated

Table IV.

Candidate gene identification for type 2 diabetes mellitus.

Table IV.

Candidate gene identification for type 2 diabetes mellitus.

A, Upregulated genes, n=46

Gene namePIMID
CCR4PMID: 17244154, PMID: 12464673
CELPMID: 19760265
CKS2
CPA2
CTRB1
DKK3
DPYSL3
EFHD2
EFNA3
FHIT
FXYD3PMID: 25058609
GAD1
GIPC2
HMMR
ISG15PMID: 25031023
KANK4
KLF5
MDFIC
NCF4
NEFL
NEFM
NEU3PMID: 17292733
NHLH2
PAIP2B
PCK1PMID: 24089092, PMID: 25997216
PLA2G1BPMID: 16567514
PLXNA1
PRIM2
PRSS2
REG1A
REG1B
RENBP
RPS19BP1
SERPINA3PMID: 28150914
SERPINA5
SFTPD
SIX6PMID: 23478426
SLC22A3
SMOC1PMID: 28163738
SPINK1
SYT1
TCTEX1D1PMID: 15144884
TENC1
TIMM44PMID: 25749183
TMPRSS2PMID: 25749183, PMID: 9419343
TMSB4X

B, Downregulated, n=37

Gene namesPIMID

SERPING1PMID: 23277452
ANKRD23PMID: 26398569
ASB9
CDK2AP2
CHD5
CHL1PMID: 22768844
CTGFPMID: 22045431
EDN3
ESM1PMID: 27756187
FAM105APMID: 20644627
GALNT14
GLIS3PMID: 23737756
IGFBP3PMID: 22554827, PMID: 26880678
KCNG3
LPAR3
MBNL3
MCOLN3
MYCN
NEDD9
NXPH1
ODC1
PCOLCE2
PHC1
PIGA
PLA1A
PPM1EPMID: 20801214
PPP1R1APMID: 25489054
RASGRF1
RCAN2
SLC2A2PMID: 28052964
TMEM27PMID: 24905913
TMEM37PMID: 29185012
TRAF3IP2PMID: 23085260
UBL3
UNC13B
ZNF610
ZNF697

[i] PIMID, PubMed number.

Functional enrichment analysis

To clarify possible biological functions of candidate genes, and examine the relationship between the functional groups and their underlying annotations in the networks, BP enrichment analyses were performed for the 46 upregulated and 37 downregulated candidate genes using ClueGO and CluePedia. A κ score >0.4 was set as the criterion. The results are presented in Fig. 4. Specifically, for the upregulated groups, the results yielded BPs related to the activation of ‘cellular response to cytokine stimulus’, ‘chemotaxis’, ‘inflammatory response’, ‘lipid metabolic process’, ‘macromolecule catabolic process’, ‘positive regulation of biosynthetic process’, ‘neurogenesis’, ‘regulation of cell differentiation’, ‘regulation of peptidase activity’, ‘regulation of transport’, ‘response to external biotic stimulus’ and ‘response to wounding’ (Fig. 4A). For the downregulated groups, the results yielded BPs related to the activation of ‘ion transport’, ‘neuron differentiation’, ‘positive regulation of cell death’, ‘positive regulation of cell proliferation’, ‘positive regulation of cellular component biogenesis’, ‘positive regulation of signaling’, ‘regulation of cell cycle process’ and ‘regulation of ion transport’ (Fig. 4B).

To obtain an improved understanding of the functional involvement of these candidate genes, pathway-based functional enrichment analyses was performed using ClueGO and CluePedia. A κ score >0.4 was set as the criterion. These genes were involved in pathways associated with ‘amyotrophic lateral sclerosis (ALS)’ [neurofilament light (NEFL) neurofilament medium], ‘axon guidance’ [ephrin A3 (EFNA3) and plexin A1 (PLXNA1)], ‘cellular senescence’ [insulin like growth factor binding protein 3 (IGFBP3) and TRAF3 interacting protein 2 (TRAF3IP2)], ‘complement and coagulation cascades’ [serpin family A member 5 (SERPINA5) and serpin family G member 1 (SERPING1)], ‘fat digestion and absorption’ [carboxyl ester lipase (CEL) and phospholipase A2 group IB (PLA2G1B)], ‘glucagon signaling pathway’ (PCK1 and solute carrier family 2 member 2), ‘influenza A’ [serine protease 2 (PRSS2) and transmembrane serine protease 2 (TMPRSS2)], ‘MAPK signaling pathway’ [EFNA3 and Ras protein specific guanine nucleotide releasing factor 1 (RASGRF1)], ‘neuroactive ligand-receptor interaction’ [lysophosphatidic acid receptor 3 (LPAR3) and PRSS2], ‘PI3K-Akt signaling pathway’ (EFNA3, LPAR3 and PCK1), ‘pancreatic secretion’ [CEL, carboxypeptidase A2 (CPA2), chymotrypsinogen B1 (CTRB1), PLA2G1B and PRSS2], ‘pathways in cancer’ [CDC28 protein kinase regulatory subunit 2 (CKS2) and LPAR3), ‘phagosome’ [neutrophil cytosolic factor 4 (NCF4) and surfactant protein D (SFTPD)], ‘protein digestion and absorption’ (CPA2, CTRB1 and PRSS2), ‘purine metabolism’ [fragile histidine triad diadenosine triphosphatase (FHIT) and DNA primase subunit 2), ‘Rap1 signaling pathway’ (EFNA3 and LPAR3), ‘Ras signaling pathway’ (EFNA3, phospholipase A1 member A, PLA2G1B and RASGRF1), ‘small cell lung cancer’ (CKS2 and FHIT), ‘synaptic vesicle cycle’ [synaptotagmin (SYT1) and unc-13 homolog B (UNC13B)] and ‘transcriptional misregulation in cancer’ (IGFBP3, MYCN proto-oncogene, bHLH transcription factor and TMPRSS2) (Fig. 5).

miRNA-target gene regulatory network

The miRNAs for DEGs were predicted using the two microRNA prediction tools through miRNet. The miRNA-gene regulatory network was built, which included 22 upregulated target genes, 19 downregulated target genes and 12 miRNAs (Fig. 6). A total of 12 miRNAs were selected, including hsa-mir-335-5p (degree=15), hsa-mir-8485 (degree=4), hsa-mir-1277-5p (degree=5), hsa-mir-190a-3p (degree=5), hsa-mir-5011-5p (degree=5), hsa-mir-124-3p (degree=6), hsa-mir-7106-5p (degree=5), hsa-let-7a-5p (degree=5), hsa-mir-192-5p (degree=5), hsa-mir-26b-5p (degree=6), hsa-let-7b-5p (degree=5) and hsa-mir-98-5p (degree=5).

TF-target gene regulatory network

In order to understand the topology and dynamics of the transcriptional regulatory network, TFs with a P<0.05 in BETA with its target genes via network analysis were built into a TF-target gene regulatory network using Cytoscape. The network consisted of 127 edges and 66 nodes (Fig. 7). Based on the degree, the top 8 TFs were selected to be enhancers of SUZ12 polycomb repressive complex 2 subunit (SUZ12; degree=10), enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2; degree=15), BCL6 transcription repressor (BCL6; degree=9), zinc finger protein 580 (ZNF580; degree=10), Kruppel like factor 9 (KLF9; degree=8), MYC associated zinc finger protein (MAZ; degree=15), activating transcription factor 1 (ATF1; degree=12), structure specific recognition protein 1 (SSRP1; degree=10), WRN helicase interacting protein 1 (WRNIP1; degree=10), chromodomain helicase DNA binding protein 1 (CHD1; degree=10) and SMAD5 (degree=11).

Discussion

T2DM is a multifactorial and multigenetic disease, and its pathogenesis is complex and largely unknown. A PPI network and a layered network for DEGs were constructed and it was observed that the majority of the proteins were localized in the cytoplasm, followed by the nucleus. The modules were mined from the PPI network and ISG15, PCK1, NEDD9, thymosin β 4 X-linked (TMSB4X), SYT1, IGFBP3, NEFL, tensin 2, translocase of inner mitochondrial membrane 44 (TIMM44), hyaluronan mediated motility receptor (HMMR), ankyrin repeat and SOCS box containing 9 and TRAF3IP2 were screened as the candidate genes with the highest degree of connectivity.

ISG15 has an anti-apoptotic capability on MIN6 cells (53). Upregulated ISG15 could be a potential therapeutic approach for type 1 diabetes (T1D) in pancreatic β-cells (53). PCK1 has been a candidate gene for T2DM susceptibility (54). SYT1 is a Ca2+ sensor that plays a central role in insulin release, which is a characteristic deterioration in the early stages of T2DM (55,57). Higher levels of IGFBP3 might raise the risk of T2DM (57,58). The TIMM44 gene could be a new target for T2DM therapy (59). The procession of vascular diseases can be delayed by targeting TRAF3IP2 during diabetes and atherosclerosis, as TRAF3IP2 reconciles high glucose-induced NF-κB and AP-1-dependent inflammatory signaling and endothelial dysfunction (60). TRAF3IP2 may play a role in the pathogenesis of T1D (61). Notably, to the best of the authors' knowledge, ribosomal protein S19 binding protein 1 (RPS19BP1) and SFTPD have not been previously reported to be dysregulated in T2DM. RPS19BP1 is a direct regulator of NAD-dependent deacetylase sirtuin-1 (SIRT1), which is a promising molecular target for the treatment of obesity. RPS19BP1 can serve as a prognostic indicator via the direct regulation of SIRT1 in obese patients with T2DM (6264). SFTPD is an element of lung innate immunity that strengthens pathogen clearance and regulates inflammatory responses (65); its expression is decreased in obesity and in impaired glucose tolerance, both of which are related to the development of T2DM.

The functional assay for candidate genes using ClueGO and CluePedia in GO terms or KEGG pathways identified several molecular mechanisms, widely known to underlie the pathogenesis of T2DM. A number of vital processes/signaling pathways and key factors connected with the pathogenesis of T2DM were identified from the functional enrichment analyses. In the upregulated group, a number of the corresponding encoded proteins were distributed in the extracellular and cytoplasmic layers. In particular, it was identified that the majority of BPs were associated with ‘inflammatory response’, ‘cellular response to cytokine stimulus’ and ‘lipid metabolic process’. In previous years, a number of studies have suggested that T2DM may be a chronic inflammatory response modulated by inflammatory factors (6669). Immune cell infiltration and high levels of cytokines were observed in the pancreas islets of T2DM (70,71), which caused differing degrees of impairment to pancreatic β-cell activity, resulting in β-cell failure (72,73). Lipotoxic effects lead to impaired insulin secretion and apoptosis of β-cells, which can give rise to the β-cell functional loss in the pathogenesis of T2DM (74). In the downregulated group, more corresponding encoded proteins were distributed in the plasma membrane and nucleus layers, and BPs were related to the ‘positive regulation of cell death’ and ‘positive regulation of cell proliferation’. Previous studies demonstrated that glucotoxic conditions are used to elevate β-cell proliferation and neogenesis, and the inhibition of apoptosis and death lead to insulin release defects, which is typical of diabetes (75,76); β-cell death is the major cause of T2DM. According to protein subcellular localization, the composition and biological value of proteins could change; analyzing PPIs may help identify the signaling pathways. The present study identified three interactions among 83 candidate genes, including SERPINA3 and CTRB1 in the extracellular matrix, SERPING1 and thymosin b4 X-linked (TMSB4X) in the cytoplasm, and SYT1 and UNC13B in the cytoplasm, which were shared between BPs, including protein input into the cytoplasm and cell-cell signaling, which might indicate that their expression was altered by the signaling cascades of the extracellular-plasma membrane-cytoplasm or nucleus and alteration in cell development. Takahashi et al (77) identified that SERPINA3 levels were significantly increased in T2DM. The rs7202877 locus for CTRB1 and CTRB2, a known diabetes risk locus, might be able to prevent diabetes via the incretin pathway (78). SERPINA5 inhibits activated protein C (APC). APC has a potential preventative role for islet β-cell damage and diabetes (79). A previous study observed that the plasma levels of APC were notably decreased in T2DM (80). SERPINA5 may be involved in T2DM via inhibited APC expression. TMSB4X is involved in cell proliferation, migration and differentiation, and its level increased in diabetic membranes (81).

The enriched KEGG pathways of candidate genes involved ‘MAPK signaling pathway’, ‘Ras signaling pathway’, ‘PI3K-Akt signaling pathway’, ‘Rap1 signaling pathway’ and ‘purine metabolism’. Previous studies demonstrated that p38 MAPK and ERK signaling were activated to inhibit obesity and associated T2DM (82,83). The Ras/Raf/ERK signaling pathway may control β-cell proliferation, and Ras is essential for normal β-cell development and function (84). Saltiel and Kahn (75) demonstrated that any obstacles in the PI3K/Akt insulin signaling pathway result in islet β-cells insulin resistance and lead to β-cell function reduction. Previous studies demonstrated that the Rap1 pathway may yield targets for β-cell dysfunction therapy in diabetes (8588). The pathway enrichment results for candidate genes in the present study identified the MAPK. Ras, PI3K-Akt and Rap1 signaling pathways in diabetes. RASGRF1 is mainly involved in the MAPK and Ras signaling pathways (89). Suppressing the expression of Rasgrf1 may contribute to insufficient insulin secretion (90), which due to insulin resistance, causes T2DM. In addition, EFNA3 in the extracellular matrix was enriched in the PI3K-Akt, MAPK, Rap1 and Ras signaling pathways. EFNA3 is an upstream gene of the MAPK signaling pathway and the PI3K-Akt signaling pathway (91). Therefore, it was hypothesized that a low expression of EFNA3 may regulate β-cell proliferation by activating Ras/Raf/MEK/ERK. FHIT, a protein product involved in purine metabolism that participates in the T2DM pathway, is expressed in the pancreas (92). Its single-nucleotide polymorphism (rs3845971) was related to an intensified risk of T2DM (93). FHIT increases adenosine-diphosphate in the purine metabolism pathway (94). Therefore, FHIT may induce β-cell apoptosis in the pancreas due to T2DM.

miRNA-target gene interaction networks were constructed from 12 miRNAs. HMMR, ubiquitin like 3, ornithine decarboxylase 1, muscleblind like splicing regulator 3 and procollagen C-endopeptidase enhancer 2 (PCOLCE2) were regulated by hsa-miR-192-5p. Connective tissue growth factor (CTGF), family with sequence similarity 105, member A (FAM105A), MyoD family inhibitor domain containing, soluble carrier family 22 member 3, IGFBP3, CKS2 and EF-hand domain family member D2 were regulated by hsa-miR-124-3p. Protein phosphatase 1 regulatory inhibitor subunit 1 A (PPP1R1A), C-C motif chemokine receptor 4 (CCR4), SYT1, LMBR1 domain containing 2, C-type lectin domain containing 11A, NLR family pyrin domain containing 13, SERPINA3, protein phosphatase, Mg2+/Mn2+ dependent 1E (PPM1E), GIPC PDZ domain containing family member 2, NCF4, SERPING1, SIX homebox 6 CTRB1, CEL, cell adhesion molecular L1 like (CHL1), SFTPD and PLXNA1 were regulated by hsa-miR-335-5p. It was observed that hsa-miR-192-5p, hsa-miR-124-3p and hsa-miR-335-5p appeared to regulate the majority of the candidate genes identified in T2DM in the present study. It was previously identified that the downregulation of miR-192-5p usually occurs in the more extreme stages of diabetes (95). PCOLCE2, a collagen-related gene, is significantly reduced in T2DM (96). Zhu et al (97), identified that the expression level of hsa-miR-124-3p is decreased in patients with T2DM [9 high-body mass index (BMI) and 1 low-BMI] CTGF expression, a vital adjudicator of progressive pancreatic fibrosis, is elevated in T2DM (98). FAM105A is reported to be associated with T2DM (36). miR-335-5p expression was increased by islets in a diabetic Goto-Kakizaki-rat model (99). PPP1R1A has previously been identified as a potential participant and experimentally validated in the pathogenesis of islet dysfunction in T2DM (100). The ratio of CXCR3 to CCR4 receptor expression was positively correlated with the duration of T1D (r=0.947; P=0.0004) (101). The expression of SERPINA3 was increased significantly in T2DM and can be used for the early detection of T2DM (77). PPM1E is a potential drug target for diabetic therapies (102). The CTRB1/2 locus influences the susceptibility and treatment for diabetes via the incretin pathway (78). It was previously identified that mutations for the highly polymorphic CEL gene can be a rare cause of monogenic diabetes (103). CHL1 affects insulin secretion in INS-1 cells and has been identified as being potentially involved in T2DM (104). SFTPD expression was decreased in patients with T2DM (65). Therefore, has-miR-8485, has-miR-1277-3p, has-miR-190a-3p, has-miR-5011-3p, has-let-7a-5p, has-let-7b-5p, has-miR-98-5p, has-miR-7106-5p and has-miR-26b-5p may also be involved in T2DM by potentially regulating the expression of various candidate genes, such as CTGF, PCK1, PPP1RA, PCOLCE2, FAM105A, TRAF3PI2 and neuraminidase 3.

A TF-target gene regulatory network was constructed, from which 10 TFs were identified and Smad5 was a potential target for T2DM treatment (105). The Forkhead box class O/Bcl6/cyclin D2 pathway connects nutrient and growth factor status to cell cycle control in pancreatic β-cells, and should therefore be considered for its therapeutic potential in diabetes (106). Notably, 8 of these transcription regulatory factors, SUZ12, EZH2, ZNF580, KLF9, MAZ, ATF1, SSRP1, WRNIP1, CHD1 were shown to be involved in the development of T2DM by modulating the expression of various candidate genes such as ankyrin repeat domain 23 (ANKRD23), transmembrane protein 37 (TMEM37), PPP1R1A, PCK1, CTGF, ISG1, SSRP1, WRNIP1 and CHD1, which have not been previously reported to be dysregulated in T2DM. The present study predicted that these TFs might play key roles in the occurrence and development of T2DM. This result provided preliminary evidence that a lower expression of TMEM37 could reflect a decrease in β-cell numbers in T2DM (107). ANKRD23, a diabetes-related ankyrin repeat protein, was identified as a novel gene that is upregulated in the hearts of KKA(y) mice, a T2DM and insulin-resistant animal model (108). Solimena et al (110) also identified that ANKRD23, PPP1R1A and TMEM37 were enriched in β-cells and downregulated in T2DM. TMEM37 prohibits Ca2+-influx and insulin secretion in β-cells (109).

The present study has some limitations. The number of samples was relatively inadequate, although the combining of multiple datasets can compensate for missing or unreliable information in any single dataset. Additionally, the present results are preliminary and descriptive. Integrative analysis of gene profiling data cannot entirely exclude false positive results. Furthermore, the present study only discussed mRNA expression and did not refer to the protein expression of the factors identified. Due to post-transcription regulatory events, protein expression levels may or may not correlate with mRNA levels. However, the alteration of protein structure, function and interaction is the underlying mechanism of a number of diseases, including diabetes (110112). Therefore, some experiments, such as reverse transcription-quantitative PCR (113,114), western blotting (115), cross-linking immunoprecipitation (116) or functional experimental validation, are needed to validate key genes, TFs and miRNAs and relevant proteins in T2DM development. Despite these limitations, the present study still provided insight for understanding the complicated underlying molecular mechanisms of T2DM.

Overall, the bioinformatics analysis of the present study identified potential markers that may play a potential role in the occurrence, development and treatment of T2DM. A total of 83 candidate genes were selected, and ISG15, PCK1, SYT1, IGFBP3, TIMM44 and TRAF3IP2 could be the core genes of T2DM. Certain key BPs such as ‘inflammatory response’, ‘cellular responses to cytokine stimulus’, ‘lipid metabolic process’, ‘positive regulation of cell death’ and ‘positive regulation of cell proliferation’, and certain signaling pathways associated with the PI3K-Akt, MAPK, Rap1 and Ras signaling pathways were identified to be involved in T2DM. The present study also identified miRNAs, including hsa-miR-192-5p, hsa-miR-124-5p and hsa-miR-335-5p, and TFs, including Smad5 and Bcl6, that might be potential targets for the diagnosis and treatment of T2DM. In addition, has-miR-8485, has-miR-1277-3p, has-miR-190a-3p, has-miR-5011-3p, has-let-7a-5p, has-let-7b-5p, has-miR-98-5p, has-miR-7106-5p and has-miR-26b-5p, and TFs SUZ12, EZH2, ZNF580, KLF9, MAZ, ATF1, SSRP1, WRNIP1 and CHD1 have not been previously identified to be related to T2DM, to the best of the authors’ knowledge, while they and their target genes may serve as diagnostic indicators for patients with T2DM. To obtain more reliable correlation results, it is necessary to validate the predicted results with a series of verification experiments. The present study identified candidate genes for T2DM development, which might be redefined as pathogenic genes for T2DM diagnosis and therapy. The experimental results could provide insight for future genomic individualized treatment of T2DM and help to identify the underlying molecular mechanisms that lead to T2DM.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by The National Natural Science Foundation of China (grant no. c010201), Startup Funding for Specialized Professorship provided by The Shanghai Jiao Tong University (grant no. WF220441502), The National Key Research and Development Program (grant no. 2017YFC1308605), The Fundamental Research Funds for The Central Universities Key Grant (grant no. CQDXWL-2014-Z002), a grant (grant no. YZYN-15-04) from The Basic Scientific Research Special of The Central Public Welfare Research Institutes of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, The Applied Basic Research Foundation of Yunnan Province of China (grant no. 2016FB143), The National Natural Science Foundation of China (grant no. 81503289) and The PUMC Youth Fund (grant no. 2017350013).

Availability of data and materials

The datasets used or analyzed during the present study are included in this article.

Authors' contributions

HL and GL designed and conceived the experiments. YLu, GL and YLi collected and analyzed the data. HL and YL wrote the manuscript, and all authors reviewed the manuscript.

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 

Zheng Y, Ley SH and Hu FB: Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat Rev Endocrinol. 14:88–98. 2018. View Article : Google Scholar : PubMed/NCBI

2 

International Diabetes Federation: IDF Diabetes Atlas. (8th). IDF. (Brussels, Belgium). 2017.

3 

Ohn JH, Kwak SH, Cho YM, Lim S, Jang HC, Park KS and Cho NH: 10-year trajectory of β-cell function and insulin sensitivity in the development of type 2 diabetes: A community-based prospective cohort study. Lancet Diabetes Endocrinol. 4:27–34. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Ashcroft FM, Rohm M, Clark A and Brereton MF: Is type 2 diabetes a glycogen storage disease of pancreatic β cells? Cell Metab. 26:17–23. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Almaça J, Weitz J, Rodriguez-Diaz R, Pereira E and Caicedo A: The pericyte of the pancreatic islet regulates capillary diameter and local blood flow. Cell Metab. 27:630–644.e4. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Wei FJ, Cai CY, Yu P, Lv J, Ling C, Shi WT, Jiao HX, Chang BC, Yang FH, Tian Y, et al: Quantitative candidate gene association studies of metabolic traits in Han Chinese type 2 diabetes patients. Genet Mol Res. 14:15471–15481. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Chen J, Meng Y, Zhou J, Zhuo M, Ling F, Zhang Y, Du H and Wang X: Identifying candidate genes for type 2 diabetes mellitus and obesity through gene expression profiling in multiple tissues or cells. J Diabetes Res. 2013:9704352013. View Article : Google Scholar : PubMed/NCBI

8 

Lynch CJ and Adams SH: Branched-chain amino acids in metabolic signalling and insulin resistance. Nat Rev Endocrinol. 10:723–736. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Xue A, Wu Y, Zhu Z, Zhang F, Kemper KE, Zheng Z, Yengo L, Lloyd-Jones LR, Sidorenko J, Wu Y, et al: eQTLGen Consortium: Genome-wide association analyses identify 143 risk variants and putative regulatory mechanisms for type 2 diabetes. Nat Commun. 9:29412018. View Article : Google Scholar : PubMed/NCBI

10 

Lawlor N, Khetan S, Ucar D and Stitzel ML: Genomics of Islet (Dys)function and Type 2 Diabetes. Trends Genet. 33:244–255. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Lee I, Blom UM, Wang PI, Shim JE and Marcotte EM: Prioritizing candidate disease genes by network-based boosting of genome-wide association data. Genome Res. 21:1109–1121. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Lawlor N, George J, Bolisetty M, Kursawe R, Sun L, Sivakamasundari V, Kycia I, Robson P and Stitzel ML: Single-cell transcriptomes identify human islet cell signatures and reveal cell-type-specific expression changes in type 2 diabetes. Genome Res. 27:208–222. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Segerstolpe Å, Palasantza A, Eliasson P, Andersson EM, Andréasson AC, Sun X, Picelli S, Sabirsh A, Clausen M, Bjursell MK, et al: Single-cell transcriptome profiling of human pancreatic islets in health and type 2 diabetes. Cell Metab. 24:593–607. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Zeggini E, Scott LJ, Saxena R, Voight BF, Marchini JL, Hu T, de Bakker PI, Abecasis GR, Almgren P, Andersen G, et al Wellcome Trust Case Control Consortium, : Meta-analysis of genome-wide association data and large-scale replication identifies additional susceptibility loci for type 2 diabetes. Nat Genet. 40:638–645. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Maruthur NM, Gribble MO, Bennett WL, Bolen S, Wilson LM, Balakrishnan P, Sahu A, Bass E, Kao WH and Clark JM: The pharmacogenetics of type 2 diabetes: A systematic review. Diabetes Care. 37:876–886. 2014. View Article : Google Scholar : PubMed/NCBI

16 

van de Bunt M, Manning Fox JE, Dai X, Barrett A, Grey C, Li L, Bennett AJ, Johnson PR, Rajotte RV, Gaulton KJ, et al: Transcript expression data from human islets links regulatory signals from genome-wide association studies for type 2 diabetes and glycemic traits to their downstream effectors. PLoS Genet. 11:e10056942015. View Article : Google Scholar : PubMed/NCBI

17 

Bonnefond A and Froguel P: Rare and common genetic events in type 2 diabetes: What should biologists know? Cell Metab. 21:357–368. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Ndiaye FK, Ortalli A, Canouil M, Huyvaert M, Salazar-Cardozo C, Lecoeur C, Verbanck M, Pawlowski V, Boutry R, Durand E, et al: Expression and functional assessment of candidate type 2 diabetes susceptibility genes identify four new genes contributing to human insulin secretion. Mol Metab. 6:459–470. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Pellegrino M, Sciambi A, Treusch S, Durruthy-Durruthy R, Gokhale K, Jacob J, Chen TX, Geis JA, Oldham W, Matthews J, et al: High-throughput single-cell DNA sequencing of acute myeloid leukemia tumors with droplet microfluidics. Genome Res. 28:1345–1352. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Zhang P, Xia JH, Zhu J, Gao P, Tian YJ, Du M, Guo YC, Suleman S, Zhang Q, Kohli M, et al: High-throughput screening of prostate cancer risk loci by single nucleotide polymorphisms sequencing. Nat Commun. 9:20222018. View Article : Google Scholar : PubMed/NCBI

21 

Nepal C, O'Rourke CJ, Oliveira DVNP, Taranta A, Shema S, Gautam P, Calderaro J, Barbour A, Raggi C, Wennerberg K, et al: Genomic perturbations reveal distinct regulatory networks in intrahepatic cholangiocarcinoma. Hepatology. 68:949–963. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Fuchsberger C, Flannick J, Teslovich TM, Mahajan A, Agarwala V, Gaulton KJ, Ma C, Fontanillas P, Moutsianas L, McCarthy DJ, et al: The genetic architecture of type 2 diabetes. Nature. 536:41–47. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Thul PJ, Åkesson L, Wiking M, Mahdessian D, Geladaki A, Ait Blal H, Alm T, Asplund A, Björk L, Breckels LM, et al: A subcellular map of the human proteome. Science. 356:8062017. View Article : Google Scholar : PubMed/NCBI

24 

Zhu H, Bilgin M and Snyder M: Proteomics. Annu Rev Biochem. 72:783–812. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Larance M and Lamond AI: Multidimensional proteomics for cell biology. Nat Rev Mol Cell Biol. 16:269–280. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Stuart LM, Boulais J, Charriere GM, Hennessy EJ, Brunet S, Jutras I, Goyette G, Rondeau C, Letarte S, Huang H, et al: A systems biology analysis of the Drosophila phagosome. Nature. 445:95–101. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Zhao L, Chen Y, Bajaj AO, Eblimit A, Xu M, Soens ZT, Wang F, Ge Z, Jung SY, He F, et al: Integrative subcellular proteomic analysis allows accurate prediction of human disease-causing genes. Genome Res. 26:660–669. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Hunt KK, Karakas C, Ha MJ, Biernacka A, Yi M, Sahin AA, Adjapong O, Hortobagyi GN, Bondy M, Thompson P, et al: Cytoplasmic Cyclin E Predicts Recurrence in Patients with Breast Cancer. Clin Cancer Res. 23:2991–3002. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Mezzanzanica D, Fabbi M, Bagnoli M, Staurengo S, Losa M, Balladore E, Alberti P, Lusa L, Ditto A, Ferrini S, et al: Subcellular localization of activated leukocyte cell adhesion molecule is a molecular predictor of survival in ovarian carcinoma patients. Clin Cancer Res. 14:1726–1733. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Zhu W, Yang L and Du Z: Layered functional network analysis of gene expression in human heart failure. PLoS One. 4:e62882009. View Article : Google Scholar : PubMed/NCBI

31 

Du ZP, Wu BL, Wang SH, Shen JH, Lin XH, Zheng CP, Wu ZY, Qiu XY, Zhan XF, Xu LY, et al: Shortest path analyses in the protein-protein interaction network of NGAL (neutrophil gelatinase-associated lipocalin) overexpression in esophageal squamous cell carcinoma. Asian Pac J Cancer Prev. 15:6899–6904. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Diao B, Liu Y, Zhang Y, Liu Q, Lu WJ and Xu G: Functional network analysis with the subcellular location and gene ontology information in human allergic asthma. Genet Test Mol Biomarkers. 16:1287–1292. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Lee S, Zhang C, Kilicarslan M, Piening BD, Bjornson E, Hallström BM, Groen AK, Ferrannini E, Laakso M, Snyder M, et al: Integrated Network Analysis Reveals an Association between Plasma Mannose Levels and Insulin Resistance. Cell Metab. 24:172–184. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Alvarez MJ, Shen Y, Giorgi FM, Lachmann A, Ding BB, Ye BH and Califano A: Functional characterization of somatic mutations in cancer using network-based inference of protein activity. Nat Genet. 48:838–847. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Krell J, Stebbing J, Frampton AE, Carissimi C, Harding V, De Giorgio A, Fulci V, Macino G, Colombo T and Castellano L: The role of TP53 in miRNA loading onto AGO2 and in remodelling the miRNA-mRNA interaction network. Lancet. 385 (Suppl 1):S152015. View Article : Google Scholar : PubMed/NCBI

36 

Marselli L, Thorne J, Dahiya S, Sgroi DC, Sharma A, Bonner-Weir S, Marchetti P and Weir GC: Gene expression profiles of Beta-cell enriched tissue obtained by laser capture microdissection from subjects with type 2 diabetes. PLoS One. 5:e114992010. View Article : Google Scholar : PubMed/NCBI

37 

Keshava Prasad TS, Goel R, Kandasamy K, Keerthikumar S, Kumar S, Mathivanan S, Telikicherla D, Raju R, Shafreen B, Venugopal A, et al: Human Protein Reference Database--2009 update. Nucleic Acids Res 37 (Database). D767–D772. 2009. View Article : Google Scholar

38 

Chatr-Aryamontri A, Breitkreutz BJ, Oughtred R, Boucher L, Heinicke S, Chen D, Stark C, Breitkreutz A, Kolas N, O'Donnell L, et al: The BioGRID interaction database: 2015 update. Nucleic Acids Res. 43(D1): D470–D478. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Kerrien S, Aranda B, Breuza L, Bridge A, Broackes-Carter F, Chen C, Duesbury M, Dumousseau M, Feuermann M, Hinz U, et al: The IntAct molecular interaction database in 2012. Nucleic Acids Res. 40(D1): D841–D846. 2012. View Article : Google Scholar : PubMed/NCBI

40 

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(D1): D447–D452. 2015. View Article : Google Scholar : PubMed/NCBI

41 

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

42 

Pontén F, Jirström K and Uhlen M: The Human Protein Atlas--a tool for pathology. J Pathol. 216:387–393. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Barsky A, Gardy JL, Hancock RE and Munzner T: Cerebral: A Cytoscape plugin for layout of and interaction with biological networks using subcellular localization annotation. Bioinformatics. 23:1040–1042. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Bindea G, Galon J and Mlecnik B: CluePedia Cytoscape plugin: Pathway insights using integrated experimental and in silico data. Bioinformatics. 29:661–663. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Bindea G, Mlecnik B, Hackl H, Charoentong P, Tosolini M, Kirilovsky A, Fridman WH, Pagès F, Trajanoski Z and Galon J: ClueGO: A Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 25:1091–1093. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Gene Ontology Consortium: Gene Ontology Consortium: Going forward. Nucleic Acids Res. 43(D1): D1049–D1056. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Kanehisa M, Furumichi M, Tanabe M, Sato Y and Morishima K: KEGG: New perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 45(D1): D353–D361. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Fan Y, Siklenka K, Arora SK, Ribeiro P, Kimmins S and Xia J: miRNet - dissecting miRNA-target interactions and functional associations through network-based visual analysis. Nucleic Acids Res 44 (W1). W135–41. 2016. View Article : Google Scholar

49 

Chou CH, Chang NW, Shrestha S, Hsu SD, Lin YL, Lee WH, Yang CD, Hong HC, Wei TY, Tu SJ, et al: miRTarBase 2016: Updates to the experimentally validated miRNA-target interactions database. Nucleic Acids Res. 44(D1): D239–D247. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Xiao F, Zuo Z, Cai G, Kang S, Gao X and Li T: miRecords: An integrated resource for microRNA-target interactions. Nucleic Acids Res 37 (Database). D105–D110. 2009. View Article : Google Scholar

51 

Xia J, Gill EE and Hancock RE: NetworkAnalyst for statistical, visual and network-based meta-analysis of gene expression data. Nat Protoc. 10:823–844. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Wang S, Sun H, Ma J, Zang C, Wang C, Wang J, Tang Q, Meyer CA, Zhang Y and Liu XS: Target analysis by integration of transcriptome and ChIP-seq data with BETA. Nat Protoc. 8:2502–2515. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Yoshikawa A, Imagawa A, Nakata S, Fukui K, Kuroda Y, Miyata Y, Sato Y, Hanafusa T, Matsuoka TA, Kaneto H, et al: Interferon stimulated gene 15 has an anti-apoptotic effect on MIN6 cells. Endocr J. 61:883–890. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Rees SD, Britten AC, Bellary S, O'Hare JP, Kumar S, Barnett AH and Kelly MA: The promoter polymorphism −232C/G of the PCK1 gene is associated with type 2 diabetes in a UK-resident South Asian population. BMC Med Genet. 10:832009. View Article : Google Scholar : PubMed/NCBI

55 

Gustavsson N and Han W: Calcium-sensing beyond neurotransmitters: Functions of synaptotagmins in neuroendocrine and endocrine secretion. Biosci Rep. 29:245–259. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Marshall C, Hitman GA, Partridge CJ, Clark A, Ma H, Shearer TR and Turner MD: Evidence that an isoform of calpain-10 is a regulator of exocytosis in pancreatic beta-cells. Mol Endocrinol. 19:213–224. 2005. View Article : Google Scholar : PubMed/NCBI

57 

Rajpathak SN, He M, Sun Q, Kaplan RC, Muzumdar R, Rohan TE, Gunter MJ, Pollak M, Kim M, Pessin JE, et al: Insulin-like growth factor axis and risk of type 2 diabetes in women. Diabetes. 61:2248–2254. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Drogan D, Schulze MB, Boeing H and Pischon T: Insulin-like growth factor 1 and insulin-like growth factor-binding protein 3 in relation to the risk of type 2 diabetes mellitus: Results from the EPIC-Potsdam study. Am J Epidemiol. 183:553–560. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Wang Y, Katayama A, Terami T, Han X, Nunoue T, Zhang D, Teshigawara S, Eguchi J, Nakatsuka A, Murakami K, et al: Translocase of inner mitochondrial membrane 44 alters the mitochondrial fusion and fission dynamics and protects from type 2 diabetes. Metabolism. 64:677–688. 2015. View Article : Google Scholar : PubMed/NCBI

60 

Venkatesan B, Valente AJ, Das NA, Carpenter AJ, Yoshida T, Delafontaine JL, Siebenlist U and Chandrasekar B: CIKS (Act1 or TRAF3IP2) mediates high glucose-induced endothelial dysfunction. Cell Signal. 25:359–371. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Bergholdt R, Brorsson C, Palleja A, Berchtold LA, Fløyel T, Bang-Berthelsen CH, Frederiksen KS, Jensen LJ, Størling J and Pociot F: Identification of novel type 1 diabetes candidate genes by integrating genome-wide association data, protein-protein interactions, and human pancreatic islet gene expression. Diabetes. 61:954–962. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Kokkola T, Suuronen T, Molnár F, Määttä J, Salminen A, Jarho EM and Lahtela-Kakkonen M: AROS has a context-dependent effect on SIRT1. FEBS Lett. 588:1523–1528. 2014. View Article : Google Scholar : PubMed/NCBI

63 

Haigis MC and Sinclair DA: Mammalian sirtuins: Biological insights and disease relevance. Annu Rev Pathol. 5:253–295. 2010. View Article : Google Scholar : PubMed/NCBI

64 

Zhou CH, Zhang MX, Zhou SS, Li H, Gao J, Du L and Yin XX: SIRT1 attenuates neuropathic pain by epigenetic regulation of mGluR1/5 expressions in type 2 diabetic rats. Pain. 158:130–139. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Ortega FJ, Pueyo N, Moreno-Navarrete JM, Sabater M, Rodriguez-Hermosa JI, Ricart W, Tinahones FJ and Fernandez-Real JM: The lung innate immune gene surfactant protein-D is expressed in adipose tissue and linked to obesity status. Int J Obes (Lond) (2005). 37:1532–1538. 2013. View Article : Google Scholar

66 

Crook M: Type 2 diabetes mellitus: A disease of the innate immune system? An update. Diabet Med. 21:203–207. 2004. View Article : Google Scholar : PubMed/NCBI

67 

Yang W, Li Y, Wang JY, Han R and Wang L: Circulating levels of adipose tissue-derived inflammatory factors in elderly diabetes patients with carotid atherosclerosis: A retrospective study. Cardiovasc Diabetol. 17:752018. View Article : Google Scholar : PubMed/NCBI

68 

Prattichizzo F, De Nigris V, Spiga R, Mancuso E, La Sala L, Antonicelli R, Testa R, Procopio AD, Olivieri F and Ceriello A: Inflammageing and metaflammation: The yin and yang of type 2 diabetes. Ageing Res Rev. 41:1–17. 2018. View Article : Google Scholar : PubMed/NCBI

69 

Naidoo V, Naidoo M and Ghai M: Cell- and tissue-specific epigenetic changes associated with chronic inflammation in insulin resistance and type 2 diabetes mellitus. Scand J Immunol. 88:e127232018. View Article : Google Scholar : PubMed/NCBI

70 

Ehses JA, Perren A, Eppler E, Ribaux P, Pospisilik JA, Maor-Cahn R, Gueripel X, Ellingsgaard H, Schneider MK, Biollaz G, et al: Increased number of islet-associated macrophages in type 2 diabetes. Diabetes. 56:2356–2370. 2007. View Article : Google Scholar : PubMed/NCBI

71 

Böni-Schnetzler M, Thorne J, Parnaud G, Marselli L, Ehses JA, Kerr-Conte J, Pattou F, Halban PA, Weir GC and Donath MY: Increased interleukin (IL)-1beta messenger ribonucleic acid expression in beta -cells of individuals with type 2 diabetes and regulation of IL-1beta in human islets by glucose and autostimulation. J Clin Endocrinol Metab. 93:4065–4074. 2008. View Article : Google Scholar : PubMed/NCBI

72 

Donath MY, Böni-Schnetzler M, Ellingsgaard H and Ehses JA: Islet inflammation impairs the pancreatic beta-cell in type 2 diabetes. Physiology (Bethesda). 24:325–331. 2009.PubMed/NCBI

73 

Marchetti P, Lupi R, Del Guerra S, Bugliani M, D'Aleo V, Occhipinti M, Boggi U, Marselli L and Masini M: Goals of treatment for type 2 diabetes: Beta-cell preservation for glycemic control. Diabetes Care. 32 (Suppl 2):S178–S183. 2009. View Article : Google Scholar : PubMed/NCBI

74 

Cnop M: Fatty acids and glucolipotoxicity in the pathogenesis of Type 2 diabetes. Biochem Soc Trans. 36:348–352. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Saltiel AR and Kahn CR: Insulin signalling and the regulation of glucose and lipid metabolism. Nature. 414:799–806. 2001. View Article : Google Scholar : PubMed/NCBI

76 

Wajchenberg BL: beta-cell failure in diabetes and preservation by clinical treatment. Endocr Rev. 28:187–218. 2007. View Article : Google Scholar : PubMed/NCBI

77 

Takahashi E, Unoki-Kubota H, Shimizu Y, Okamura T, Iwata W, Kajio H, Yamamoto-Honda R, Shiga T, Yamashita S, Tobe K, et al: Proteomic analysis of serum biomarkers for prediabetes using the Long-Evans Agouti rat, a spontaneous animal model of type 2 diabetes mellitus. J Diabetes Investig. 8:661–671. 2017. View Article : Google Scholar : PubMed/NCBI

78 

Hart LM, Fritsche A, Nijpels G, van Leeuwen N, Donnelly LA, Dekker JM, Alssema M, Fadista J, Carlotti F, Gjesing AP, et al: The CTRB1/2 locus affects diabetes susceptibility and treatment via the incretin pathway. Diabetes. 62:3275–3281. 2013. View Article : Google Scholar : PubMed/NCBI

79 

Xue M and Jackson CJ: Activated protein C and its potential applications in prevention of islet β-cell damage and diabetes. Vitam Horm. 95:323–363. 2014. View Article : Google Scholar : PubMed/NCBI

80 

Matsumoto K, Yano Y, Gabazza EC, Araki R, Bruno NE, Suematsu M, Akatsuka H, Katsuki A, Taguchi O, Adachi Y, et al: Inverse correlation between activated protein C generation and carotid atherosclerosis in Type 2 diabetic patients. Diabet Med. 24:1322–1328. 2007. View Article : Google Scholar : PubMed/NCBI

81 

Wang JY, Lu Q, Tao Y, Jiang YR and Jonas JB: Intraocular expression of thymosin β4 in proliferative diabetic retinopathy. Acta Ophthalmol. 89:e396–e403. 2011. View Article : Google Scholar : PubMed/NCBI

82 

Sidarala V and Kowluru A: The regulatory roles of mitogen-activated protein kinase (MAPK) pathways in health and diabetes: Lessons learned from the pancreatic β-cell. Recent Pat Endocr Metab Immune Drug Discov. 10:76–84. 2017. View Article : Google Scholar : PubMed/NCBI

83 

Pang Y, Zhu H, Xu J, Yang L, Liu L and Li J: β-arrestin-2 is involved in irisin induced glucose metabolism in type 2 diabetes via p38 MAPK signaling. Exp Cell Res. 360:199–204. 2017. View Article : Google Scholar : PubMed/NCBI

84 

Stewart AF, Hussain MA, García-Ocaña A, Vasavada RC, Bhushan A, Bernal-Mizrachi E and Kulkarni RN: Human β-cell proliferation and intracellular signaling: Part 3. Diabetes. 64:1872–1885. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Fraenkel M, Ketzinel-Gilad M, Ariav Y, Pappo O, Karaca M, Castel J, Berthault MF, Magnan C, Cerasi E, Kaiser N, et al: mTOR inhibition by rapamycin prevents beta-cell adaptation to hyperglycemia and exacerbates the metabolic state in type 2 diabetes. Diabetes. 57:945–957. 2008. View Article : Google Scholar : PubMed/NCBI

86 

Baeder WL, Sredy J, Sehgal SN, Chang JY and Adams LM: Rapamycin prevents the onset of insulin-dependent diabetes mellitus (IDDM) in NOD mice. Clin Exp Immunol. 89:174–178. 1992. View Article : Google Scholar : PubMed/NCBI

87 

Zhang CL, Katoh M, Shibasaki T, Minami K, Sunaga Y, Takahashi H, Yokoi N, Iwasaki M, Miki T and Seino S: The cAMP sensor Epac2 is a direct target of antidiabetic sulfonylurea drugs. Science. 325:607–610. 2009. View Article : Google Scholar : PubMed/NCBI

88 

Sabbatini ME, Chen X, Ernst SA and Williams JA: Rap1 activation plays a regulatory role in pancreatic amylase secretion. J Biol Chem. 283:23884–23894. 2008. View Article : Google Scholar : PubMed/NCBI

89 

Manyes L, Arribas M, Gomez C, Calzada N, Fernandez-Medarde A and Santos E: Transcriptional profiling reveals functional links between RasGrf1 and Pttg1 in pancreatic beta cells. BMC Genomics. 15:10192014. View Article : Google Scholar : PubMed/NCBI

90 

Hoffmann A and Spengler D: Transient neonatal diabetes mellitus gene Zac1 impairs insulin secretion in mice through Rasgrf1. Mol Cell Biol. 32:2549–2560. 2012. View Article : Google Scholar : PubMed/NCBI

91 

Yang Y, Fu Q, Wang X, Liu Y, Zeng Q, Li Y, Gao S, Bao L, Liu S, Gao D, et al: Comparative transcriptome analysis of the swimbladder reveals expression signatures in response to low oxygen stress in channel catfish, Ictalurus punctatus. Physiol Genomics. 50:636–647. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Dong Q, Ginsberg HN and Erlanger BF: Overexpression of the A1 adenosine receptor in adipose tissue protects mice from obesity-related insulin resistance. Diabetes Obes Metab. 3:360–366. 2001. View Article : Google Scholar : PubMed/NCBI

93 

Rampersaud E, Damcott CM, Fu M, Shen H, McArdle P, Shi X, Shelton J, Yin J, Chang YP, Ott SH, et al: Identification of novel candidate genes for type 2 diabetes from a genome-wide association scan in the Old Order Amish: Evidence for replication from diabetes-related quantitative traits and from independent populations. Diabetes. 56:3053–3062. 2007. View Article : Google Scholar : PubMed/NCBI

94 

Straub SG and Sharp GW: Glucose-stimulated signaling pathways in biphasic insulin secretion. Diabetes Metab Res Rev. 18:451–463. 2002. View Article : Google Scholar : PubMed/NCBI

95 

Ma X, Lu C, Lv C, Wu C and Wang Q: The expression of miR-192 and its significance in diabetic nephropathy patients with different urine albumin creatinine ratio. J Diabetes Res. 2016:67894022016. View Article : Google Scholar : PubMed/NCBI

96 

Wu C, Chen X, Shu J and Lee CT: Whole-genome expression analyses of type 2 diabetes in human skin reveal altered immune function and burden of infection. Oncotarget. 8:34601–34609. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Zhu Z, Yin J, Li DC and Mao ZQ: Role of microRNAs in the treatment of type 2 diabetes mellitus with Roux-en-Y gastric bypass. Braz J Med Biol Res. 50:e58172017. View Article : Google Scholar : PubMed/NCBI

98 

Pivovarova O, Fisher E, Dudziak K, Ilkavets I, Dooley S, Slominsky P, Limborska S, Weickert MO, Spranger J, Fritsche A, et al: A polymorphism within the connective tissue growth factor (CTGF) gene has no effect on non-invasive markers of beta-cell area and risk of type 2 diabetes. Dis Markers. 31:241–246. 2011. View Article : Google Scholar : PubMed/NCBI

99 

Salunkhe VA, Ofori JK, Gandasi NR, Salo SA, Hansson S, Andersson ME, Wendt A, Barg S, Esguerra JLS and Eliasson L: MiR-335 overexpression impairs insulin secretion through defective priming of insulin vesicles. Physiol Rep. 52017. PubMed/NCBI

100 

Taneera J, Fadista J, Ahlqvist E, Atac D, Ottosson-Laakso E, Wollheim CB and Groop L: Identification of novel genes for glucose metabolism based upon expression pattern in human islets and effect on insulin secretion and glycemia. Hum Mol Genet. 24:1945–1955. 2015. View Article : Google Scholar : PubMed/NCBI

101 

Aso Y, Matsuura H, Momobayashi A, Inukai Y, Sugawara N, Nakano T, Yamamoto R, Wakabayashi S, Takebayashi K and Inukai T: Profound reduction in T-helper (Th) 1 lymphocytes in peripheral blood from patients with concurrent type 1 diabetes and Graves' disease. Endocr J. 53:377–385. 2006. View Article : Google Scholar : PubMed/NCBI

102 

Voss M, Paterson J, Kelsall IR, Martín-Granados C, Hastie CJ, Peggie MW and Cohen PT: Ppm1E is an in cellulo AMP-activated protein kinase phosphatase. Cell Signal. 23:114–124. 2011. View Article : Google Scholar : PubMed/NCBI

103 

Torsvik J, Johansson S, Johansen A, Ek J, Minton J, Raeder H, Ellard S, Hattersley A, Pedersen O, Hansen T, et al: Mutations in the VNTR of the carboxyl-ester lipase gene (CEL) are a rare cause of monogenic diabetes. Hum Genet. 127:55–64. 2010. View Article : Google Scholar : PubMed/NCBI

104 

Taneera J, Lang S, Sharma A, Fadista J, Zhou Y, Ahlqvist E, Jonsson A, Lyssenko V, Vikman P, Hansson O, et al: A systems genetics approach identifies genes and pathways for type 2 diabetes in human islets. Cell Metab. 16:122–134. 2012. View Article : Google Scholar : PubMed/NCBI

105 

Anhê FF, Lellis-Santos C, Leite AR, Hirabara SM, Boschero AC, Curi R, Anhê GF and Bordin S: Smad5 regulates Akt2 expression and insulin-induced glucose uptake in L6 myotubes. Mol Cell Endocrinol. 319:30–38. 2010. View Article : Google Scholar : PubMed/NCBI

106 

Glauser DA and Schlegel W: The FoxO/Bcl-6/cyclin D2 pathway mediates metabolic and growth factor stimulation of proliferation in Min6 pancreatic beta-cells. J Recept Signal Transduct Res. 29:293–298. 2009. View Article : Google Scholar : PubMed/NCBI

107 

Rechsteiner MP, Floros X, Boehm BO, Marselli L, Marchetti P, Stoffel M, Moch H and Spinas GA: Automated assessment of β-cell area and density per islet and patient using TMEM27 and BACE2 immunofluorescence staining in human pancreatic β-cells. PLoS One. 9:e989322014. View Article : Google Scholar : PubMed/NCBI

108 

Ikeda K, Emoto N, Matsuo M and Yokoyama M: Molecular identification and characterization of a novel nuclear protein whose expression is up-regulated in insulin-resistant animals. J Biol Chem. 278:3514–3520. 2003. View Article : Google Scholar : PubMed/NCBI

109 

Solimena M, Schulte AM, Marselli L, Ehehalt F, Richter D, Kleeberg M, Mziaut H, Knoch KP, Parnis J, Bugliani M, et al: Systems biology of the IMIDIA biobank from organ donors and pancreatectomised patients defines a novel transcriptomic signature of islets from individuals with type 2 diabetes. Diabetologia. 61:641–657. 2018. View Article : Google Scholar : PubMed/NCBI

110 

Scott EM, Carter AM and Findlay JB: The application of proteomics to diabetes. Diab Vasc Dis Res. 2:54–60. 2005. View Article : Google Scholar : PubMed/NCBI

111 

Khan AR and Awan FR: Mining of protein based biomarkers for type 2 diabetes mellitus. Pak J Pharm Sci. 25:889–901. 2012.PubMed/NCBI

112 

Okada S, List EO, Sankaran S and Kopchick JJ: Plasma protein biomarkers correlated with the development of diet-induced Type 2 diabetes in mice. Clin Proteomics. 6:6–17. 2010. View Article : Google Scholar : PubMed/NCBI

113 

Bustin SA and Mueller R: Real-time reverse transcription PCR (qRT-PCR) and its potential use in clinical diagnosis. Clin Sci (Lond). 109:365–379. 2005. View Article : Google Scholar : PubMed/NCBI

114 

Guénin S, Mauriat M, Pelloux J, Van Wuytswinkel O, Bellini C and Gutierrez L: Normalization of qRT-PCR data: The necessity of adopting a systematic, experimental conditions-specific, validation of references. J Exp Bot. 60:487–493. 2009. View Article : Google Scholar : PubMed/NCBI

115 

Kim B: Western Blot Techniques. Methods Mol Biol. 1606:133–139. 2017. View Article : Google Scholar : PubMed/NCBI

116 

Ule J, Hwang HW and Darnell RB: The future of cross-linking and immunoprecipitation (CLIP). Cold Spring Harb Perspect Biol. 10:a0322432018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2020
Volume 22 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
Lu Y, Li Y, Li G and Lu H: Identification of potential markers for type 2 diabetes mellitus via bioinformatics analysis. Mol Med Rep 22: 1868-1882, 2020
APA
Lu, Y., Li, Y., Li, G., & Lu, H. (2020). Identification of potential markers for type 2 diabetes mellitus via bioinformatics analysis. Molecular Medicine Reports, 22, 1868-1882. https://doi.org/10.3892/mmr.2020.11281
MLA
Lu, Y., Li, Y., Li, G., Lu, H."Identification of potential markers for type 2 diabetes mellitus via bioinformatics analysis". Molecular Medicine Reports 22.3 (2020): 1868-1882.
Chicago
Lu, Y., Li, Y., Li, G., Lu, H."Identification of potential markers for type 2 diabetes mellitus via bioinformatics analysis". Molecular Medicine Reports 22, no. 3 (2020): 1868-1882. https://doi.org/10.3892/mmr.2020.11281