Next Article in Journal
The Pathogenic Diagnosis in Pediatric Diabetology: Next Generation Sequencing and Precision Therapy
Next Article in Special Issue
Laboratory Findings in Children with Excess Body Weight in Romania
Previous Article in Journal
Distributions of Platelet-Derived Growth Factor Receptor-α Positive Cells and Interstitial Cells of Cajal in the Colon of Rats with Diabetes Mellitus Type 2
Previous Article in Special Issue
Adiponectin/Leptin Ratio as an Index to Determine Metabolic Risk in Patients after Kidney Transplantation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Bioinformatics and Next-Generation Data Analysis for Identification of Genes and Molecular Pathways Involved in Subjects with Diabetes and Obesity

by
Prashanth Ganekal
1,
Basavaraj Vastrad
2,
Satish Kavatagimath
3,
Chanabasayya Vastrad
4,* and
Shivakumar Kotrashetti
4
1
Department of General Medicine, Basaveshwara Medical College, Chitradurga 577501, Karnataka, India
2
Department of Pharmaceutical Chemistry, K.L.E. College of Pharmacy, Gadag 582101, Karnataka, India
3
Department of Pharmacognosy, K.L.E. College of Pharmacy, Belagavi 590010, Karnataka, India
4
Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karnataka, India
*
Author to whom correspondence should be addressed.
Medicina 2023, 59(2), 309; https://doi.org/10.3390/medicina59020309
Submission received: 21 December 2022 / Revised: 19 January 2023 / Accepted: 29 January 2023 / Published: 7 February 2023

Abstract

:
Background and Objectives: A subject with diabetes and obesity is a class of the metabolic disorder. The current investigation aimed to elucidate the potential biomarker and prognostic targets in subjects with diabetes and obesity. Materials and Methods: The next-generation sequencing (NGS) data of GSE132831 was downloaded from Gene Expression Omnibus (GEO) database. Functional enrichment analysis of DEGs was conducted with ToppGene. The protein–protein interactions network, module analysis, target gene–miRNA regulatory network and target gene–TF regulatory network were constructed and analyzed. Furthermore, hub genes were validated by receiver operating characteristic (ROC) analysis. A total of 872 DEGs, including 439 up-regulated genes and 433 down-regulated genes were observed. Results: Second, functional enrichment analysis showed that these DEGs are mainly involved in the axon guidance, neutrophil degranulation, plasma membrane bounded cell projection organization and cell activation. The top ten hub genes (MYH9, FLNA, DCTN1, CLTC, ERBB2, TCF4, VIM, LRRK2, IFI16 and CAV1) could be utilized as potential diagnostic indicators for subjects with diabetes and obesity. The hub genes were validated in subjects with diabetes and obesity. Conclusion: This investigation found effective and reliable molecular biomarkers for diagnosis and prognosis by integrated bioinformatics analysis, suggesting new and key therapeutic targets for subjects with diabetes and obesity.

1. Introduction

Diabetes mellitus and obesity are major metabolic or endocrine disorders and are dramatically increasing throughout the globe [1]. The prevalence of obesity and type 2 diabetes mellitus is considerably higher [2]. Diabetes mellitus and obesity are linked with progression of cardiovascular diseases [3], hypertension [4], and neurological and neuropsychiatric disorders [5] and asthma [6]. Till today, there is no cure for diabetes mellitus and obesity, and treatment and mediation tailored to clinical features are endorsed. Genetic and environmental factors are two initial contributors to these disorders [7]. Exploration of the molecular mechanisms of diabetes mellitus and obesity will develop the considerate of its pathogenesis and has key implications for designing new therapy.
Molecular mechanisms of subject with diabetes and obesity have been increasingly studied. Previous investigations showed that genes and signaling pathways are associated with diabetes mellitus and obesity. Key genes such as ENPP1 [8] and FTO [9] were responsible for development of diabetes mellitus and obesity. Recent investigations showed that PI3K/AKT pathway [10] and TLR pathway [11] as a potential target for diabetes mellitus and obesity. However, certain key genes and pathways associated with diabetes mellitus and obesity have not been completely investigated. Further studies are necessary to elucidate these essential genes and pathways to provide novel therapeutic targets for the treatment of diabetes and obesity.
In recent years, the analysis of biological information, known as bioinformatics, has attracted a great deal of attention and sustained breakthroughs in the search for biomarkers for various diseases [12,13,14]. With the gradual advancement of next-generation sequencing (NGS) technology, bioinformatics has become increasingly essential in molecular pathogenesis, performing a major role in elucidating diseases mechanisms and finding novel targets for diseases treatment and patient prognosis [15]. With the wide function of NGS, a huge amount of data hasbeen generated, and most of the data have been deposited and stored in public databases. NGS data analyses have been carried out on diabetes and obesity in recent years [16], and hundreds of differentially expressed genes (DEGs) have been obtained. Bioinformatics methods combining with NGS techniques will be innovative.
Therefore, in this investigation, we downloaded the next-generation sequencing (NGS)data GSE132831, provided by Osinski et al. [17], from Gene Expression Omnibus (GEO, http://www.ncbi.nlm.nih.gov/geo/, accessed on 11 June 2020) [18] database to identify the differentially expressed genes (DEGs) between diabetes mellitus and obesity samples and normal control samples. With the identified DEGs, we performed Gene Ontology (GO) and pathway enrichment analyses to investigate the functions and pathways enriched by the DEGs. Additionally, we constructed a protein–protein interaction (PPI) network and modules screened out some important gene nodes to perform clustering analysis. Furthermore, we constructed a target gene–miRNA regulatory network and target gene–TF regulatory network based on these key genes to investigate the potential relationships between genes and subject with diabetes and obesity. Finally, hub genes were validated by using receiver operating characteristic (ROC) curve analysis. The research design of this study was shown in Figure 1. These results might provide novel ideas for future investigation and treatment of diabetes mellitus and obesity by exploring prognostic markers and therapeutic targets in diabetes mellitus and obesity.

2. Materials and Methods

2.1. RNA Sequencing Data

The NGS data GSE132831 was downloaded from the GEO database, which was based on the platform of GPL1857 Illumina NextSeq 500 (Homo sapiens). This dataset, including samples of 104 diabetic obese and samples of 120 normal control, was deposited by Osinski et al. [17].

2.2. Identification of DEGs

The limma R/Bioconductor software package was used to perform the identification of DEGs between samples of diabetic obese and normal control in R software [19]. The cutoff criteria were |logFC| > 1.112 for up-regulated genes, |logFC| <−0.64 for down-regulated genes, and a p-value < 0.05. The significance of p value measures how likely it is that any observed difference between two groups (diabetes mellitus and obesity samples and normal control samples). The significance of log FC looks only at genes which vary wildly amongst other genes.

2.3. GO and Pathway Enrichment Analyses of DEGs

The ToppGene (ToppFun) (https://toppgene.cchmc.org/enrichment.jsp, accessed on 11 June 2020) [20] bioinformatics resources was utilized to distinguish and enrich the biological attributes, such as biological processes (BP), cellular components (CC), molecular functions (MF) and pathways, of identified DEGs (Up- and down-regulated genes separately). Moreover, GO (http://geneontology.org/, accessed on 11 June 2020) [21] and REACTOME (https://reactome.org/, accessed on 11 June 2020) [22] pathway enrichment analyses were used to identify the significant GO terms and pathways. p < 0.05 was set as the cutoff criterion for significant enrichment.

2.4. Protein–Protein Interaction (PPI) Network and Module Analysis

The IID interactome (http://iid.ophid.utoronto.ca/, accessed on 11 June 2020) [23] is an online database containing known and predicted PPI networks. In this investigation, a PPI network of identified DEGs in dataset was identified using the IID interactome database (combined score >0.4) and subsequently visualized using Cytoscape (http://www.cytoscape.org/, accessed on 11 June 2020) software (version 3.8.2) [24]. The regulatory relationship between genes were analyzed through topological property of computing network including the node degree [25], betweenness centrality [26], stress centrality [27] and closeness centrality [28] by using the Network Analyzer app within Cytoscape. The PEWCC1 (http://apps.cytoscape.org/apps/PEWCC1, accessed on 11 June 2020) [29] program within Cytoscape was used to detect modules of the PPI network. The GO and pathway enrichment analysis of the identified modules was then performed using the ToppGene database.

2.5. Target Gene–miRNARegulatory Network

miRNet database (https://www.mirnet.ca/, accessed on 11 June 2020) [30] is a bioinformatics platform for predicting target gene–miRNA pairs. In the present study, the target genes were predicted using 14 miRNA databases: TarBase, miRTarBase, miRecords, miRanda (S mansoni only), miR2Disease, HMDD, PhenomiR, SM2miR, PharmacomiR, EpimiR, starBase, TransmiR, ADmiRE, and TAM 2.0. In this study, miRNAs were considered the targeted miRNAs of hub genes based on these miRNA databases. The target gene–miRNA regulatory network was depicted and visualized using Cytoscape software.

2.6. Target Gene–TF Regulatory Network

NetworkAnalyst database (https://www.networkanalyst.ca/, accessed on 11 June 2020) [31] is a bioinformatics platform for predicting target gene–TF pairs. In the present study, the target genes were predicted using ChEA TF database. In this study, TFs were considered the targeted TFs of hub genes based on this TF database. The target gene–TF regulatory network was depicted and visualized using Cytoscape software.

2.7. Receiver Operating Characteristic (ROC) Analysis

A ROC analysis is a technique for visualizing, construct and determining classifiers based on their achievement. A diagnostic test was firstly performed in order to measure the diagnostic value of candidate biomarkers in subject with diabetes and obesity. Sensitivity and specificity of each biomarker in this diagnostic test were determined. ROC curves were retrieved by plotting the sensitivity, against the specificity using the pROC in R software [32]. Area under the ROC curve (AUC) was determined to predict the efficiency of this diagnostic test. A test with AUC bigger than 0.9 is assigned great efficiency, 0.7–0.9, modest efficiency and 0.5–0.7, small efficiency.

3. Results

3.1. Identification of DEGs

The DEGs were screened by “limma” package (p-value < 0.05, and |logFC| > 1.112 for up-regulated genes and |logFC| <−0.64 for down-regulated genes). The GSE132831 dataset contained 872 DEGs, including 439 up-regulated genes and 433 down-regulated genes. DEGs are listed in Table S1. The volcano plot is presented in Figure 2. The heat map DEGs is shown in Figure 3.

3.2. GO and Pathway Enrichment Analyses of DEGs

To gain in-depth and comprehensive biological characteristics of these DEGs, GO functional annotation and REACTOME pathway enrichment analysis were performed through online analytical tool ToppGene. The BP was mainly enriched in plasma membrane bounded cell projection organization, neurogenesis, cell activation and secretion (Table S2). The CC was mainly enriched in neuron projection, golgi apparatus, secretory granule and secretory vesicle (Table S2). The MF was significantly enriched in drug binding, ribonucleotide binding, signaling receptor binding and molecular transducer activity (Table S2). Result of REACTOME enrichment analysis showed that top pathways were axon guidance, extracellular matrix organization, neutrophil degranulation and innate immune system (Table S3).

3.3. Protein–Protein Interaction (PPI) Network and Module Analysis

To find the hub genes in the DEGs, Network Analyzer, a plug-in Cytoscape was performed. All the genes and edges were determined. IID interactome mapped 872 DEGs into a PPI network containing 3894 nodes and 7142 edges (Figure 4). Hub genes with the high node degree, betweenness centrality, stress centrality and closeness centrality are listed in Table S4. MYH9 (Degree 231; Betweenness 0.083106; Stress 11909200; Closeness 0.348923), FLNA (Degree 196; Betweenness 0.07999; Stress 10927852; Closeness 0.35285), DCTN1 (Degree 168; Betweenness 0.080808; Stress 7748054; Closeness 0.330668), CLTC (Degree 158; Betweenness 0.071579; Stress 7687192; Closeness 0.351161), ERBB2 (Degree 158; Betweenness 0.069347; Stress 7857692; Closeness 0.327216), TCF4 (Degree 186; Betweenness 0.080443; Stress 7982798; Closeness 0.320102), VIM (Degree 146; Betweenness 0.062238; Stress 9673084; Closeness 0.327078), LRRK2 (Degree 114; Betweenness 0.046449; Stress 5881270; Closeness 0.333305), IFI16 (Degree 91; Betweenness 0.035934; Stress 2890936; Closeness 0.294671) and CAV1 (Degree 74; Betweenness 0.034501; Stress 3642486; Closeness 0.323593). Then, PEWCC1 was used to find clusters in the network. Module1 contained 16 nodes and 39 edges (Figure 5A). Module1 was associated with including axon guidance, signaling by NGF, plasma membrane bounded cell projection organization and neurogenesis. Module2contained 13 nodes and 24 edges (Figure 5B). Module 2 was associated with an innate immune system.

3.4. Target Gene–miRNA Regulatory Network

The target gene–miRNA regulatory network included 2520 nodes (miRNAs: 2224; gene: 296) and 15485 edges (Figure 6). The nodes with degrees were listed in Table S5. We discovered that MYH9 was targeted by 116 miRNAs (ex; hsa-mir-4329); ERBB2 was targeted by 73miRNAs (ex; hsa-mir-4315); MYO18A was targeted by 71miRNAs (ex; hsa-mir-1299); SEC16A was targeted by 66 miRNAs (ex; hsa-mir-4779); PLCG1 was targeted by 56 miRNAs (ex; hsa-mir-3685); CCNB1 was targeted by 84miRNAs (ex; hsa-mir-6134); CAV1 was targeted by 58miRNAs (ex; hsa-mir-4459); VIM was targeted by 30miRNAs (ex; hhsa-mir-6124); HAP1 was targeted by 22miRNAs (ex; hsa-mir-9500); MAD2L1 was targeted by 17miRNAs (ex; hsa-mir-1297).

3.5. Target Gene–TF Regulatory Network

The target gene–TF regulatory networkincluded 487 nodes (TFs: 195; gene: 292) and 7094 edges (Figure 7). The nodes with degrees were listed in Table S5. We discovered that that CLTC was targeted by 59 TFs (ex; SMARCA4); MYH9 was targeted by 53 TFs (ex; TCF7); NOTCH1 was targeted by 45 TFs (ex; MYB); SHC1 was targeted by 42 TFs (ex; E2F4); KIFC3 was targeted by 42 TFs (ex; CUX1); TCF4 was targeted by 50 TFs (ex; NANOG); VIM was targeted by 43 TFs (ex; GFI1B); CAV1 was targeted by 36 TFs (ex; GATA4); FBL was targeted by 33 TFs (ex; HIF1A); TUBA1A was targeted by 32 TFs (ex; CLOCK).

3.6. Receiver Operating Characteristic (ROC) Analysis

To identify new potential biomarkers for diabetes and obesity, ROC curves of data derived from healthy controls and patients with diabetes and obesity was analyzed using the R package. The AUC calculated to assess the discriminatory ability of hub genes (Figure 8). Validated by ROC curves, we found that hub genes had high sensitivity and specificity, including MYH9, FLNA, DCTN1, CLTC, ERBB2, TCF4, VIM, LRRK2, IFI16 and CAV1, and AUC values more than 0.7. This analysis demonstrated that the hub genes had a diagnostic role.

4. Discussion

A NGS investigation is an ideal way to comprehensively investigate diabetes mellitus and obesity. In this investigation, we collected NGS dataset from the GEO database, and a total of 872 DEGs, including 439 up-regulated genes and 433 down-regulated genes, were found. Altered expression of XIST (X inactive specific transcript) [33] and SELL (selectin L) [34] are associated with prognosis ofdiabetes. S100A9 and S100A8 are associated with the prognosis of diabetes mellitus and obesity [35]. IL1R2 [36] and SPINK5 [37] plays an important role in the diabetes mellitus and obesity.
GO term and REACTOME enrichment analyzes were accomplished to examine interactions between the DEGs. The altered expression of genes including ERBB2 [38], DACT1 [39], ARAP1 [40], MYH9 [41], INPPL1 [42], SARM1 [43], NOTCH1 [44], ROBO1 [45], MAPK8IP1 [46], ANK1 [47], SARM1 [43], SREBF2 [48], SIK1 [49], PASK (PAS domain-containing serine/threonine kinase) [50], NOS2 [51], OAS3 [52], KL (klotho) [53], PECAM1 [54], S100A12 [55], S100P [56], BATF3 [57], PLEK (pleckstrin) [58], ALOX5 [59], ARG1 [60], CXCL8 [61], CXCR1 [62], PTAFR (platelet-activating factor receptor) [63], PYGL (glycogen phosphorylase L) [64], TCF4 [65], CAMP (cathelicidin antimicrobial peptide) [66], RUNX2 [67], PLA2G2A [68], GCG (glucagon) [69], RARRES2 [70] and HAP1 [71] in diabetes mellitus was reported to be an independent prognostic factors. ACHE (acetylcholinesterase) [72], FGFR3 [73], VLDLR (very-low-density lipoprotein receptor) [74], SHC1 [75], HDAC6 [76], CHRNA2 [77], CASR (calcium-sensing receptor) [78], ELK1 [79], TYK2 [80], CIITA (class II major histocompatibility complex transactivator) [81], ZAP70 [82], GPT (glutamic-pyruvic transaminase) [83], CHI3L1 [84], AIF1 [85], MMP9 [86], ITGB2 [87], CFD (complement factor D) [88], C3AR1 [89], LGALS1 [90], CD14 [91], TIMP1 [92], TLR2 [93], LTF (lactotransferrin) [94], BRCA2 [95] and IGFBP3 [96] are a potential prognostic markers in obesity. Sun et al. [97] reported that TRPM2 was significantly regulated in diabetes and obesity. Findings were implied by Richter et al. [98], Suchkova et al. [99], Qureshi et al. [100], Wang et al. [101], Wang et al. [102], Aoki-Suzuki et al. [103], Ohno et al. [104], Richter et al. [98], Rahman and Copeland [105], Congiu et al. [106], Ji et al. [107], Wollmer et al. [108], Yamazaki et al. [109], Bardien et al. [110], Comella Bolla et al. [111], Horvath et al. [112], Watanabe et al. [113], Kushima et al. [114], Grünblatt et al. [115], and Sato and Kawata [116] when they found that TAOK2, ACAP3, PLXNA3, PLXNA4, DCTN1, NTNG2, LRP4, AGRN (agrin), TAOK2, POLG (DNA polymerase gamma, catalytic subunit), KCNK2, OPRK1, ABCA2, ABCA7, LRRK2, CD200, PAK3, PADI2, EPHB1, CHAT (choline O-acetyltransferase) and SLC18A1 plays a substantial role in the patients with neurological and neuropsychiatric disorders. Studies showed that biomarkers include PLD2 [117], FLNA (filamin A) [118], SMURF1 [119], LINGO1 [120], CACNA1H [121], NLRP6 [122], NLRC3 [123], CXCR2 [124] and C5AR1 [125] plays an important role in progression of hypertension. Sauzeau et al. [126], Xu et al. [127], Hirota et al. [128], Alharatani et al. [129], Beitelshees et al. [130], Zhu et al. [131], Gil-Cayuela et al. [132], Liu et al. [133], Xie et al. [134], Kroupis et al. [135], López-Mejías et al. [136], Gremmel et al. [137] Yamada and Guo [138], Petri et al. [139], DeFilippis et al. [140], Rocca et al. [141] and Tur et al. [142] found that genes include VAV2, RASAL1, LIF (LIF interleukin 6 family cytokine), CTNND1, CACNA1C, MAP3K10, NRBP2, TRPM4, LILRB2, FCGR2A, PIK3CG, SELPLG (selectin P ligand), PRDX4, FPR2, PLG (plasminogen), SELENOM (selenoprotein M) and NCAM1 were a diagnostic markers of cardiovascular diseasesand could be used as therapeutic targets. Accumulating evidence shows that ITGB4 [143], SEMA3D [144], FCAR (Fc fragment of IgA receptor) [145], KIT (KIT proto-oncogene, receptor tyrosine kinase) [146], PGLYRP1 [147], IL17RB [148], BIRC5 [149] and PTGS1 [150] are associated with prognosis in asthma. Studies showed that GRK2 [151], ADCY3 [152], FASN (fatty acid synthase) [153], DGKD (diacylglycerol kinase delta) [154], DGKQ (diacylglycerol kinase theta) [154], IP6K1 [155], ANXA1 [156], SUCNR1 [157], PRNP (prion protein) [158], CXCR4 [159], CAV1 [160], LCN2 [161], AQP9 [162], NMU (neuromedin U) [163], NPY1R [164], FFAR2 [165], OSM (oncostatin M) [166] and TREM1 [167] might be a potential markers for diabetes mellitus and obesity. Researchers have shown that UNC13B [168], PFKFB3 [169], FCN1 [170] and SLC11A1 [171] were diagnostic markers for type 1 diabetes. DEGs involved in GO terms and pathways were more likely related to diabetes mellitus and obesity, and DEGs also involved in neurological and neuropsychiatric disorders, hypertension, cardiovascular diseases and asthma.
As known, dynamic networks analysis and disease gene association were criteria for progression of various diseases [172,173]. Protein–protein interaction (PPI) network and its module can be regarded as key to the understanding of progression of diabetes mellitus and obesity, and might also lead to novel therapeutic way. MYH9 [41,174,175,176], ERBB2 [38,177,178,179,180], TCF4 [65,181], VIM (vimentin) [182,183], LRRK2 [184,185] and CAV1 [161,186,187,188,189,190,191,192] have been implicated as a principal mediator of diabetes mellitus. VIM (vimentin) binds to insulin-responsive aminopeptidas, a major cargo protein of glucose transporter type 4, and decreases the glucose tolerance [182]. IFI16 [193], ERBB2 [194], VIM (vimentin) [182,195] and CAV1 [160,196,197,198,199] are crucial factors for advancement of obesity. IFI16 showed adipogenesis, an enhanced inflammatory state and damaged insulin-stimulated glucose uptake in adipose tissue [193]. Motor protein MYH9 bindsto actin and producesmechanical force through magnesium-dependent hydrolysis of ATP, and it generatesthe contraction of striated and smooth muscles [200]. ErbB2 is a receptor tyrosine kinase family whose activity in cells depends on dimerization with another ligand-binding ErbB receptor, and associated with progression of various diseases [201]. TCF4 is a member of the basic helix–loop–helix (bHLH) family of transcription factors that have a key role in a various diseases [202]. VIM (vimentin) is an intermediate filament (IF) protein and plays an important role in epithelial–mesenchymal transition (EMT), a process that occurs during the development of various diseases [203]. LRRK2 is an enigmatic protein and has been one of the central molecules in a number of human diseases [204]. CAV1 is a cell surface protein shownto play a key role in insulin resistance [205]. IFI16 is an innate immune sensor for intracellular DNA and is associated with DNA damage in various diseases [206]. We identified novel targets including CLTC (clathrin heavy chain), TNS2, PLCG1 and NIFK (nucleolar protein interacting with the FHA domain of MKI67) for specific therapy of diabetes mellitus and obesity. Further investigation is needed to validate these results and investigate the roles of these biomarkers in diabetes mellitus and obesity.
In the present investigation, NGS data analysis revealed that the mechanism of occurrence of diabetes mellitus and obesity might be related to the expression of miRNA and TF. To validate the accuracy of the target genes, miRNAs and TFs identified by target gene–miRNA regulatory network and target gene–TF regulatory network analysis. Yan et al. [207], Wang et al. [208], Yan et al. [209] and Guo et al. [210] showed that expression and prognosis of hsa-mir-4329, hsa-mir-3685, hsa-mir-6124, hsa-mir-1297 and SMARCA4 are associated with the risk of cardiovascular diseases. Several studies have shown that biomarkers including hsa-mir-1299 [211], hsa-mir-4779 [212] and hsa-mir-4459 [213] might be predictive biomarkers for the efficacy of diabetes mellitus treatment. TCF7 was revealed and regarded as diagnostic biomarker in type 1 diabetes mellitus [214]. Transcription factor MYB was involved in asthma [215]. MYB might be associated with diabetes and obesity. E2F4 [216] and CLOCK [217] are associated with prognosis in patients with diabetes mellitus and obesity. CUX1 [218], NANOG [219], GATA4 [220] and HIF1A [221] plays a vital role in the patients with obesity. Novel targets include MYO18A, SEC16A, CCNB1, MAD2L1, hsa-mir-4315, hsa-mir-6134, hsa-mir-9500, KIFC3, FBL (fibrillarin), TUBA1A and GFI1B might have crucial biologic functions in the pathogenesis of patients with diabetes mellitus and obesity. This result indicated that our identified biomarkers are involved in the pathological progression of diabetes and obesity, its associated complications beingneurological and neuropsychiatric disorders, hypertension, cardiovascular diseases and asthma, thus warranting further exploration.
However, there are some limitations in this investigation. For instance, the NGS data were obtained from the GEO database and were not given by the authors. Therefore, further research should be conducted to verify whether these target genes can be used in the clinical treatment of diabetes mellitus and obesity.

5. Conclusions

Using a bioinformatics analysis of NGS dataset GSE132831, we identified the genes of diabetes and obesity. We found that DEGs in patients were enriched for pathways mainly involved in the axon guidance, neutrophil degranulation, plasma membrane-bounded cell projection organization, and cell activation. Focusing on the key genes and corresponding pathways involved in diabetes and obesity could provide new insights for diabetes mellitus and obesity treatment. Hub genes including MYH9, FLNA, DCTN1, CLTC, ERBB2, TCF4, VIM, LRRK2, IFI16 and CAV1 were identified as potential novel biomarkers for diabetes and obesity. The validation of hub genes was demonstrated by ROC analysis. Further investigation isurgently demanded to validate the hub genes, and further molecular mechanisms would be uncovered. All the output will lay the foundation for finding a possible therapeutic strategy to treat diabetes mellitus and obesity.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/medicina59020309/s1, Table S1: The statistical metrics for key differentially expressed genes (DEGs).; Table S2: The enriched GO terms of the up and down regulated differentially expressed genes; Table S3: The enriched pathway terms of the up and down regulated differentially expressed genes; Table S4: Topology table for up and down regulated genes; Table S5: miRNA—target gene and TF—target gene interaction.

Author Contributions

P.G., methodology and validation; B.V., writing—original draft, and review and editing; S.K. (Satish Kavatagimath) formal analysis and resources; C.V., software, and investigation; S.K. (Shivakumar Kotrashetti) supervision and validation. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets supporting the conclusions of this article are available in the GEO (Gene Expression Omnibus) (https://www.ncbi.nlm.nih.gov/geo/, accessed on 11 June 2020) repository. [(GSE132831) (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE132831, accessed on 11 June 2020)].

Acknowledgments

The authors thank Stephane Le Crom, IBENS, Genomic Core Facility, Paris, France, very much, and also the author who deposited their expression profiling by high throughput sequencing dataset, GSE132831, into the public GEO database.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hossain, P.; Kawar, B.; El Nahas, M. Obesity and Diabetes in the Developing World—A Growing Challenge. N. Engl. J. Med. 2007, 356, 213–215. [Google Scholar] [CrossRef]
  2. Daousi, C.; Casson, I.F.; Gill, G.V.; MacFarlane, I.A.; Wilding, J.P.; Pinkney, J.H. Prevalence of obesity in type 2 diabetes in secondary care: Association with cardiovascular risk factors. Postgrad Med. J. 2006, 82, 280–284. [Google Scholar] [CrossRef] [PubMed]
  3. Scherer, P.E.; Hill, J.A. Obesity, Diabetes, and Cardiovascular Diseases: A Compendium. Circ. Res. 2016, 118, 1703–1705. [Google Scholar] [CrossRef]
  4. Babu, G.R.; Murthy, G.V.S.; Ana, Y.; Patel, P.; Deepa, R.; Neelon, S.E.B.; Kinra, S.; Reddy, K.S. Association of obesity with hypertension and type 2 diabetes mellitus in India: A meta-analysis of observational studies. World J. Diabetes 2018, 9, 40–52. [Google Scholar] [CrossRef]
  5. Naseer, M.; Bibi, F.; Alqahtani, M.H.; Chaudhary, A.G.; Azhar, E.I.; Kamal, M.A.; Yasir, M. Role of Gut Microbiota in Obesity, Type 2 Diabetes and Alzheimer’s Disease. CNS Neurol. Disord. Drug Targets 2014, 13, 305–311. [Google Scholar] [CrossRef] [PubMed]
  6. Medina-Remón, A.; Kirwan, R.; Lamuela-Raventós, R.M.; Estruch, R. Dietary patterns and the risk of obesity, type 2 diabetes mellitus, cardiovascular diseases, asthma, and neurodegenerative diseases. Crit. Rev. Food Sci. Nutr. 2018, 58, 262–296. [Google Scholar] [CrossRef] [PubMed]
  7. Romao, I.; Roth, J. Genetic and Environmental Interactions in Obesity and Type 2 Diabetes. J. Am. Diet. Assoc. 2008, 108 (Suppl. 1), S24–S28. [Google Scholar] [CrossRef]
  8. Meyre, D.; Bouatia-Naji, N.; Tounian, A.; Samson, C.; Lecoeur, C.; Vatin, V.; Ghoussaini, M.; Wachter, C.; Hercberg, S.; Charpentier, G.; et al. Variants of ENPP1 are associated with childhood and adult obesity and increase the risk of glucose intolerance and type 2 diabetes. Nat. Genet. 2005, 37, 863–867. [Google Scholar] [CrossRef]
  9. Ramya, K.; Radha, V.; Ghosh, S.; Majumder, P.P.; Mohan, V. Genetic Variations in the FTO Gene Are Associated with Type 2 Diabetes and Obesity in South Indians (CURES-79). Diabetes Technol. Ther. 2011, 13, 33–42. [Google Scholar] [CrossRef]
  10. Huang, X.; Liu, G.; Guo, J.; Su, Z. The PI3K/AKT pathway in obesity and type 2 diabetes. Int. J. Biol. Sci. 2018, 14, 1483–1496. [Google Scholar] [CrossRef] [Green Version]
  11. Aamir, K.; Khan, H.U.; Sethi, G.; Hossain, M.A.; Arya, A. Wnt signaling mediates TLR pathway and promote unrestrained adipogenesis and metaflammation: Therapeutic targets for obesity and type 2 diabetes. Pharmacol. Res. 2020, 152, 104602. [Google Scholar] [CrossRef]
  12. Kumar, S.U.; Kumar, D.T.; Siva, R.; Doss, C.G.P.; Zayed, H. Integrative Bioinformatics Approaches to Map Potential Novel Genes and Pathways Involved in Ovarian Cancer. Front. Bioeng. Biotechnol. 2019, 7, 391. [Google Scholar] [CrossRef]
  13. Udhaya Kumar, S.; Thirumal Kumar, D.; Bithia, R.; Sankar, S.; Magesh, R.; Sidenna, M.; George Priya Doss, C.; Zayed, H. Analysis of Differentially Expressed Genes and Molecular Pathways in Familial Hypercholesterolemia Involved in Atherosclerosis: A Systematic and Bioinformatics Approach. Front. Genet. 2020, 11, 734. [Google Scholar] [CrossRef]
  14. Fu, D.; Zhang, B.; Yang, L.; Huang, S.; Xin, W. Development of an Immune-Related Risk Signature for Predicting Prognosis in Lung Squamous Cell Carcinoma. Front. Genet. 2020, 11, 978. [Google Scholar] [CrossRef]
  15. Li, X.; Liao, Z.; Deng, Z.; Chen, N.; Zhao, L. Combining bulk and single-cell RNA-sequencing data to reveal gene expression pattern of chondrocytes in the osteoarthritic knee. Bioengineered 2021, 12, 997–1007. [Google Scholar] [CrossRef]
  16. Prashanth, G.; Vastrad, B.; Tengli, A.; Vastrad, C.; Kotturshetti, I. Investigation of candidate genes and mechanisms underlying obesity associated type 2 diabetes mellitus using bioinformatics analysis and screening of small drug molecules. BMC Endocr. Disord. 2021, 21, 80. [Google Scholar] [CrossRef]
  17. Osinski, C.; Le Gléau, L.; Poitou, C.; de Toro-Martin, J.; Genser, L.; Fradet, M.; Soula, H.A.; Leturque, A.; Blugeon, C.; Jourdren, L.; et al. Type 2 diabetes is associated with impaired jejunal enteroendocrine GLP-1 cell lineage in human obesity. Int. J. Obes. 2020, 45, 170–183. [Google Scholar] [CrossRef]
  18. Clough, E.; Barrett, T. The Gene Expression Omnibus Database. Methods Mol. Biol. 2016, 1418, 93–110. [Google Scholar]
  19. Ritchie, M.E.; Phipson, B.; Wu, D.; Hu, Y.; Law, C.W.; Shi, W.; Smyth, G.K. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015, 43, e47. [Google Scholar] [CrossRef]
  20. Chen, J.; Bardes, E.E.; Aronow, B.J.; Jegga, A.G. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 2009, 37, W305–W311. [Google Scholar] [CrossRef]
  21. Thomas, P.D. The Gene Ontology and the Meaning of Biological Function. Methods Mol. Biol. 2017, 1446, 15–24. [Google Scholar] [PubMed] [Green Version]
  22. Fabregat, A.; Jupe, S.; Matthews, L.; Sidiropoulos, K.; Gillespie, M.; Garapati, P.; Haw, R.; Jassal, B.; Korninger, F.; May, B.; et al. The Reactome Pathway Knowledgebase. Nucleic Acids Res. 2018, 46, D649–D655. [Google Scholar] [CrossRef] [PubMed]
  23. Kotlyar, M.; Pastrello, C.; Malik, Z.; Jurisica, I. IID 2018 update: Context-specific physical protein–protein interactions in human, model organisms and domesticated species. Nucleic Acids Res. 2019, 47, D581–D589. [Google Scholar] [CrossRef] [PubMed]
  24. Shannon, P.; Markiel, A.; Ozier, O.; Baliga, N.S.; Wang, J.T.; Ramage, D.; Amin, N.; Schwikowski, B.; Ideker, T. Cytoscape: A software environment for integrated models of Biomolecular Interaction Networks. Genome Res. 2003, 13, 2498–2504. [Google Scholar] [CrossRef] [PubMed]
  25. Przulj, N.; Wigle, D.A.; Jurisica, I. Functional topology in a network of protein interactions. Bioinformatics 2004, 20, 340–348. [Google Scholar] [CrossRef]
  26. Nguyen, T.-P.; Liu, W.-C.; Jordán, F. Inferring pleiotropy by network analysis: Linked diseases in the human PPI network. BMC Syst. Biol. 2011, 5, 179. [Google Scholar] [CrossRef]
  27. Shi, Z.; Zhang, B. Fast network centrality analysis using GPUs. BMC Bioinform. 2011, 12, 149. [Google Scholar] [CrossRef]
  28. Fadhal, E.; Gamieldien, J.; Mwambene, E.C. Protein interaction networks as metric spaces: A novel perspective on distribution of hubs. BMC Syst. Biol. 2014, 8, 6. [Google Scholar] [CrossRef]
  29. Zaki, N.; Efimov, D.; Berengueres, J. Protein complex detection using interaction reliability assessment and weighted clustering coefficient. BMC Bioinform. 2013, 14, 163. [Google Scholar] [CrossRef]
  30. Fan, Y.; Xia, J. miRNet—Functional Analysis and Visual Exploration of miRNA–Target Interactions in a Network Context. Methods Mol. Biol. 2018, 1819, 215–233. [Google Scholar] [CrossRef]
  31. Zhou, G.; Soufan, O.; Ewald, J.; Hancock, R.E.W.; Basu, N.; Xia, J. NetworkAnalyst 3.0: A visual analytics platform for comprehensive gene expression profiling and meta-analysis. Nucleic Acids Res. 2019, 47, W234–W241. [Google Scholar] [CrossRef] [Green Version]
  32. Robin, X.; Turck, N.; Hainard, A.; Tiberti, N.; Lisacek, F.; Sanchez, J.-C.; Müller, M. pROC: An open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinform. 2011, 12, 77. [Google Scholar] [CrossRef]
  33. Sohrabifar, N.; Ghaderian, S.M.H.; Parsa, S.A.; Ghaedi, H.; Jafari, H. Variation in the expression level of MALAT1, MIAT and XIST lncRNAs in coronary artery disease patients with and without type 2 diabetes mellitus. Arch. Physiol. Biochem. 2020, 128, 1308–1315. [Google Scholar] [CrossRef]
  34. Stavarachi, M.; Panduru, N.M.; Serafinceanu, C.; Moţa, E.; Moţa, M.; Cimponeriu, D.; Ion, D.A. Investigation of P213S SELL gene polymorphism in type 2 diabetes mellitus and related end stage renal disease. A case-control study. Romanian J. Morphol. Embryol. 2011, 52 (Suppl. 3), 995–998. [Google Scholar]
  35. Lylloff, L.; Bathum, L.; Madsbad, S.; Grundtvig, J.L.G.; Nordgaard-Lassen, I.; Fenger, M. S100A8/A9 (Calprotectin), Interleukin-6, and C-Reactive Protein in Obesity and Diabetes before and after Roux-en-Y Gastric Bypass Surgery. Obes. Facts 2017, 10, 386–395. [Google Scholar] [CrossRef]
  36. Gagné-Ouellet, V.; Guay, S.-P.; Boucher-Lafleur, A.-M.; Bouchard, L.; Laprise, C. DNA methylation signature of interleukin 1 receptor type II in asthma. Clin. Epigenet. 2015, 7, 80. [Google Scholar] [CrossRef]
  37. Martínez-Aguilar, N.E.; Del Río-Navarro, B.E.; Navarro-Olivos, E.; García-Ortíz, H.; Orozco, L.; Jiménez-Morales, S. SPINK5 and ADRB2 haplotypes are risk factors for asthma in Mexican pediatric patients. J. Asthma 2015, 52, 232–239. [Google Scholar] [CrossRef]
  38. Muhammad, I.F.; Borné, Y.; Bao, X.; Melander, O.; Orho-Melander, M.; Nilsson, P.M.; Nilsson, J.; Engström, G. Circulating HER2/ErbB2 Levels Are Associated With Increased Incidence of Diabetes: A Population-Based Cohort Study. Diabetes Care 2019, 42, 1582–1588. [Google Scholar] [CrossRef]
  39. Yu, C.-Y.; Yang, C.-Y.; Rui, Z.-L. MicroRNA-125b-5p improves pancreatic β-cell function through inhibiting JNK signaling pathway by targeting DACT1 in mice with type 2 diabetes mellitus. Life Sci. 2019, 224, 67–75. [Google Scholar] [CrossRef]
  40. Kulzer, J.R.; Stitzel, M.L.; Morken, M.A.; Huyghe, J.R.; Fuchsberger, C.; Kuusisto, J.; Laakso, M.; Boehnke, M.; Collins, F.S.; Mohlke, K.L. A Common Functional Regulatory Variant at a Type 2 Diabetes Locus Upregulates ARAP1 Expression in the Pancreatic Beta Cell. Am. J. Hum. Genet. 2014, 94, 186–197. [Google Scholar] [CrossRef]
  41. Freedman, B.I.; Langefeld, C.D.; Lu, L.; Divers, J.; Comeau, M.E.; Kopp, J.; Winkler, C.A.; Nelson, G.W.; Johnson, R.C.; Palmer, N.D.; et al. Differential Effects of MYH9 and APOL1 Risk Variants on FRMD3 Association with Diabetic ESRD in African Americans. PLoS Genet. 2011, 7, e1002150. [Google Scholar] [CrossRef] [PubMed]
  42. Marion, E.; Kaisaki, P.J.; Pouillon, V.; Gueydan, C.; Levy, J.C.; Bodson, A.; Krzentowski, G.; Daubresse, J.-C.; Mockel, J.; Behrends, J.; et al. The Gene INPPL1, Encoding the Lipid Phosphatase SHIP2, Is a Candidate for Type 2 Diabetes In Rat and Man. Diabetes 2002, 51, 2012–2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Cheng, Y.; Liu, J.; Luan, Y.; Liu, Z.; Lai, H.; Zhong, W.; Yang, Y.; Yu, H.; Feng, N.; Wang, H.; et al. Sarm1 Gene Deficiency Attenuates Diabetic Peripheral Neuropathy in Mice. Diabetes 2019, 68, 2120–2130. [Google Scholar] [CrossRef] [PubMed]
  44. DiNicolantonio, J.J.; Mccarty, M. Autophagy-induced degradation of Notch1, achieved through intermittent fasting, may promote beta cell neogenesis: Implications for reversal of type 2 diabetes. Open Heart 2019, 6, e001028. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, J.; Hou, W.; Guan, T.; Tang, L.; Zhu, X.; Li, Y.; Hou, S.; Zhang, J.; Chen, H.; Huang, Y. Slit2/Robo1 signaling is involved in angiogenesis of glomerular endothelial cells exposed to a diabetic-like environment. Angiogenesis 2018, 21, 237–249. [Google Scholar] [CrossRef]
  46. Waeber, G.; Delplanque, J.; Bonny, C.; Mooser, V.; Steinmann, M.; Widmann, C.; Maillard, A.; Miklossy, J.; Dina, C.; Hani, E.H.; et al. The gene MAPK8IP1, encoding islet-brain-1, is a candidate for type 2 diabetes. Nat. Genet. 2000, 24, 291–295. [Google Scholar] [CrossRef]
  47. Sun, L.; Zhang, X.; Wang, T.; Chen, M.; Qiao, H. Association of ANK1 variants with new-onset type 2 diabetes in a Han Chinese population from northeast China. Exp. Ther. Med. 2017, 14, 3184–3190. [Google Scholar] [CrossRef]
  48. Galavi, H.; Noorzehi, N.; Saravani, R.; Sargazi, S.; Mollashahee-Kohkan, F.; Shahraki, H. Association study of SREBF-2 gene polymorphisms and the risk of type 2 diabetes in a sample of Iranian population. Gene 2018, 660, 145–150. [Google Scholar] [CrossRef]
  49. Song, D.; Yin, L.; Wang, C.; Wen, X. Adenovirus-mediated expression of SIK1 improves hepatic glucose and lipid metabolism in type 2 diabetes mellitus rats. PLoS ONE 2019, 14, e0210930. [Google Scholar] [CrossRef]
  50. Da Silva Xavier, G.; Farhan, H.; Kim, H.; Caxaria, S.; Johnson, P.; Hughes, S.; Bugliani, M.; Marselli, L.; Marchetti, P.; Birzele, F.; et al. Per-arnt-sim (PAS) domain-containing protein kinase is downregulated in human islets in type 2 diabetes and regulates glucagon secretion. Diabetologia 2011, 54, 819–827. [Google Scholar] [CrossRef]
  51. Chen, F.; Li, Y.-M.; Yang, L.-Q.; Zhong, C.-G.; Zhuang, Z.-X. Association of NOS2 and NOS3 gene polymorphisms with susceptibility to type 2 diabetes mellitus and diabetic nephropathy in the Chinese Han population. IUBMB Life 2016, 68, 516–525. [Google Scholar] [CrossRef]
  52. Park, S.; Liu, M.; Kang, S. Alcohol Intake Interacts with CDKAL1, HHEX, and OAS3 Genetic Variants, Associated with the Risk of Type 2 Diabetes by Lowering Insulin Secretion in Korean Adults. Alcohol. Clin. Exp. Res. 2018, 42, 2326–2336. [Google Scholar] [CrossRef]
  53. Donate-Correa, J.; Martín-Núñez, E.; Ferri, C.; Hernández-Carballo, C.; Tagua, V.G.; Delgado-Molinos, A.; López-Castillo, Á.; Rodríguez-Ramos, S.; Cerro-López, P.; López-Tarruella, V.C.; et al. FGF23 and Klotho Levels are Independently Associated with Diabetic Foot Syndrome in Type 2 Diabetes Mellitus. J. Clin. Med. 2019, 8, 448. [Google Scholar] [CrossRef]
  54. Završnik, M.; Kariž, S.; Makuc, J.; Šeruga, M.; Cilenšek, I.; Petrovič, D. PECAM-1 Leu125Val (rs688) Polymorphism and Diabetic Nephropathy in Caucasians with Type 2 Diabetes Mellitus. Anal. Cell. Pathol. 2016, 2016, 3152967. [Google Scholar] [CrossRef]
  55. Dong, N.; Shi, H.; Xu, B.; Cai, Y. Increased Plasma S100A12 Levels Are Associated with Diabetic Retinopathy and Prognostic Biomarkers of Macrovascular Events in Type 2 Diabetic Patients. Investig. Opthalmology Vis. Sci. 2015, 56, 4177–4185. [Google Scholar] [CrossRef]
  56. Afarideh, M.; Esteghamati, V.Z.; Ganji, M.; Heidari, B.; Esteghamati, S.; Lavasani, S.; Ahmadi, M.; Tafakhori, A.; Nakhjavani, M.; Esteghamati, A. Associations of Serum S100B and S100P With the Presence and Classification of Diabetic Peripheral Neuropathy in Adults with Type 2 Diabetes: A Case-Cohort Study. Can. J. Diabetes 2019, 43, 336–344.e2. [Google Scholar] [CrossRef]
  57. Ferris, S.T.; Carrero, J.A.; Mohan, J.F.; Calderon, B.; Murphy, K.M.; Unanue, E.R. A Minor Subset of Batf3-Dependent Antigen-Presenting Cells in Islets of Langerhans Is Essential for the Development of Autoimmune Diabetes. Immunity 2014, 41, 657–669. [Google Scholar] [CrossRef]
  58. Ding, Y.; Kantarci, A.; Badwey, J.A.; Hasturk, H.; Malabanan, A.; Van Dyke, T.E. Phosphorylation of Pleckstrin Increases Proinflammatory Cytokine Secretion by Mononuclear Phagocytes in Diabetes Mellitus. J. Immunol. 2007, 179, 647–654. [Google Scholar] [CrossRef]
  59. Nejatian, N.; Häfner, A.-K.; Shoghi, F.; Badenhoop, K.; Penna-Martinez, M. 5-Lipoxygenase (ALOX5): Genetic susceptibility to type 2 diabetes and vitamin D effects on monocytes. J. Steroid Biochem. Mol. Biol. 2019, 187, 52–57. [Google Scholar] [CrossRef]
  60. Shah, S.F.A.; Iqbal, T.; Naveed, N.; Akram, S.; Rafiq, M.A.; Hussain, S. ARG1 single nucleotide polymorphisms rs2781666 and rs2781665 confer risk of Type 2 diabetes mellitus. EXCLI J. 2018, 17, 847–855. [Google Scholar] [CrossRef]
  61. Da Silva, B.R.; Cirelli, T.; Nepomuceno, R.; Theodoro, L.H.; Orrico, S.R.; Cirelli, J.A.; Barros, S.P.; Scarel-Caminaga, R.M. Functional haplotype in the Interleukin8 (CXCL8) gene is associated with type 2 Diabetes Mellitus and Periodontitis in Brazilian population. Diabetes Metab. Syndr. 2020, 14, 1665–1672. [Google Scholar] [CrossRef] [PubMed]
  62. Gond, D.P.; Singh, S.; Agrawal, N. Testing an association between TLR4 and CXCR1 gene polymorphisms with susceptibility to urinary tract infection in type 2 diabetes in north Indian population. Gene 2018, 641, 196–202. [Google Scholar] [CrossRef] [PubMed]
  63. Yamaguchi, M.; Matsui, M.; Higa, R.; Yamazaki, Y.; Ikari, A.; Miyake, M.; Miwa, M.; Ishii, S.; Sugatani, J.; Shimizu, T. A platelet-activating factor (PAF) receptor deficiency exacerbates diet-induced obesity but PAF/PAF receptor signaling does not contribute to the development of obesity-induced chronic inflammation. Biochem. Pharmacol. 2015, 93, 482–495. [Google Scholar] [CrossRef] [PubMed]
  64. Nagy, L.; Docsa, T.; Szántó, M.; Brunyánszki, A.; Hegedűs, C.; Márton, J.; Kónya, B.; Virág, L.; Somsák, L.; Gergely, P.; et al. Glycogen Phosphorylase Inhibitor N-(3,5-Dimethyl-Benzoyl)-N’-(β-D-Glucopyranosyl)Urea Improves Glucose Tolerance under Normoglycemic and Diabetic Conditions and Rearranges Hepatic Metabolism. PLoS ONE 2013, 8, e69420. [Google Scholar] [CrossRef] [PubMed]
  65. Boj, S.F.; van Es, J.H.; Huch, M.; Li, V.S.; José, A.; Hatzis, P.; Mokry, M.; Haegebarth, A.; Born, M.V.D.; Chambon, P.; et al. Diabetes Risk Gene and Wnt Effector Tcf7l2/TCF4 Controls Hepatic Response to Perinatal and Adult Metabolic Demand. Cell 2012, 151, 1595–1607. [Google Scholar] [CrossRef]
  66. Patrick, C.; Wang, G.-S.; Lefebvre, D.E.; Crookshank, J.A.; Sonier, B.; Eberhard, C.; Mojibian, M.; Kennedy, C.R.; Brooks, S.P.; Kalmokoff, M.L.; et al. Promotion of Autoimmune Diabetes by Cereal Diet in the Presence or Absence of Microbes Associated with Gut Immune Activation, Regulatory Imbalance, and Altered Cathelicidin Antimicrobial Peptide. Diabetes 2013, 62, 2036–2047. [Google Scholar] [CrossRef]
  67. Zhang, G.; Li, H.; Zhao, W.; Li, M.; Tian, L.; Ju, W.; Li, X. miR-205 regulates bone turnover in elderly female patients with type 2 diabetes mellitus through targeted inhibition of Runx2. Exp. Ther. Med. 2020, 20, 1557–1565. [Google Scholar] [CrossRef]
  68. Khajeniazi, S.; Marjani, A.; Shakeri, R.; Hakimi, S. Polymorphism of Secretary PLA2G2A Gene Associated with Its Serum Level in Type2 Diabetes Mellitus Patients in Northern Iran. Endocrine Metab. Immune Disord. Drug Targets 2019, 19, 1192–1197. [Google Scholar] [CrossRef]
  69. Li, L.; Gao, K.; Zhao, J.; Feng, T.; Yin, L.; Wang, J.; Wang, C.; Li, C.; Wang, Y.; Wang, Q.; et al. Glucagon gene polymorphism modifies the effects of smoking and physical activity on risk of type 2 diabetes mellitus in Han Chinese. Gene 2014, 534, 352–355. [Google Scholar] [CrossRef]
  70. Zhao, K.; Ding, W.; Zhang, Y.; Ma, K.; Wang, D.; Hu, C.; Liu, J.; Zhang, X. Variants in the RARRES2 gene are associated with serum chemerin and increase the risk of diabetic kidney disease in type 2 diabetes. Int. J. Biol. Macromol. 2020, 165, 1574–1580. [Google Scholar] [CrossRef]
  71. Gong, Y.-J.; Feng, Y.; Cao, Y.-Y.; Zhao, J.; Wu, W.; Zheng, Y.-Y.; Wu, J.-R.; Li, X.; Yang, G.-Z.; Zhou, X. Huntingtin-associated protein 1 plays an essential role in the pathogenesis of type 2 diabetes by regulating the translocation of GLUT4 in mouse adipocytes. BMJ Open Diabetes Res. Care 2020, 8, e001199. [Google Scholar] [CrossRef]
  72. Shenhar-Tsarfaty, S.; Sherf-Dagan, S.; Berman, G.; Webb, M.; Raziel, A.; Keidar, A.; Goitein, D.; Sakran, N.; Zwang, E.; Shapira, I.; et al. Obesity-related acetylcholinesterase elevation is reversed following laparoscopic sleeve gastrectomy. Int. J. Obes. 2019, 43, 297–305. [Google Scholar] [CrossRef]
  73. Saint-Laurent, C.; Garcia, S.; Sarrazy, V.; Dumas, K.; Authier, F.; Sore, S.; Tran, A.; Gual, P.; Gennero, I.; Salles, J.-P.; et al. Early postnatal soluble FGFR3 therapy prevents the atypical development of obesity in achondroplasia. PLoS ONE 2018, 13, e0195876. [Google Scholar] [CrossRef]
  74. Kim, O.Y.; Lee, S.-M.; Chung, J.H.; Do, H.J.; Moon, J.; Shin, M.-J. Arginase I and the very low-density lipoprotein receptor are associated with phenotypic biomarkers for obesity. Nutrition 2012, 28, 635–639. [Google Scholar] [CrossRef]
  75. Feigelson, H.S.; Teras, L.R.; Diver, W.R.; Tang, W.; Patel, A.V.; Stevens, V.L.; Calle, E.E.; Thun, M.J.; Bouzyk, M. Genetic variation in candidate obesity genes ADRB2, ADRB3, GHRL, HSD11B1, IRS1, IRS2, and SHC1 and risk for breast cancer in the Cancer Prevention Study II. Breast Cancer Res. 2008, 10, R57. [Google Scholar] [CrossRef]
  76. Lieber, A.D.; Beier, U.H.; Xiao, H.; Wilkins, B.J.; Jiao, J.; Li, X.S.; Schugar, R.C.; Strauch, C.M.; Wang, Z.; Brown, J.M.; et al. Loss of HDAC6 alters gut microbiota and worsens obesity. FASEB J. 2019, 33, 1098–1109. [Google Scholar] [CrossRef]
  77. Kim, J. Association of CHRNA2 polymorphisms with overweight/obesity and clinical characteristics in a Korean population. Clin. Chem. Lab. Med. 2008, 46, 1085–1089. [Google Scholar] [CrossRef]
  78. Mattar, P.; Sanhueza, S.; Yuri, G.; Briones, L.; Perez-Leighton, C.; Rudich, A.; Lavandero, S.; Cifuentes, M. Calcium-Sensing Receptor in Adipose Tissue: Possible Association with Obesity-Related Elevated Autophagy. Int. J. Mol. Sci. 2020, 21, 7617. [Google Scholar] [CrossRef]
  79. Pang, L.; You, L.; Ji, C.-B.; Shi, C.; Chen, L.; Yang, L.; Huang, F.; Zhou, Y.; Zhang, J.; Chen, X.; et al. miR-1275 inhibits adipogenesis via ELK1 and its expression decreases in obese subjects. J. Mol. Endocrinol. 2016, 57, 33–43. [Google Scholar] [CrossRef]
  80. Derecka, M.; Gornicka, A.; Koralov, S.B.; Szczepanek, K.; Morgan, M.; Raje, V.; Sisler, J.; Zhang, Q.; Otero, D.; Cichy, J.; et al. Tyk2 and Stat3 Regulate Brown Adipose Tissue Differentiation and Obesity. Cell Metab. 2012, 16, 814–824. [Google Scholar] [CrossRef]
  81. Deng, T.; Lyon, C.J.; Minze, L.J.; Lin, J.; Zou, J.; Liu, J.Z.; Ren, Y.; Yin, Z.; Hamilton, D.J.; Reardon, P.R.; et al. Class II Major Histocompatibility Complex Plays an Essential Role in Obesity-Induced Adipose Inflammation. Cell Metab. 2013, 17, 411–422. [Google Scholar] [CrossRef] [PubMed]
  82. Torres, K.; Torres, A.; Chrościcki, A.; Maciejewski, R.; Radej, S.; Roliński, J.; Pietrzyk, Ł.; Wallner, G. Evaluation of lymphocytes CD4+ and CD8+ and expression of ZAP-70 kinase on CD3+ and CD19+ lymphocytes in obese patients undergoing laparoscopic cholecystectomy. Surg. Endosc. 2013, 27, 872–879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Nakamura, S.; Takezawa, Y.; Nakajima, Y.; Maeda, T. Elevation of glutamic pyruvic transaminase and .GAMMA.-glutamyl transpeptidase in obesity. Tohoku J. Exp. Med. 1980, 132, 473–478. [Google Scholar] [CrossRef] [PubMed]
  84. Teitsdottir, U.D.; Arnardottir, E.S.; Bjornsdottir, E.; Gislason, T.; Petersen, P.H. Obesity modulates the association between sleep apnea treatment and CHI3L1 levels but not CHIT1 activity in moderate to severe OSA: An observational study. Sleep Breath. 2018, 22, 1101–1109. [Google Scholar] [CrossRef] [PubMed]
  85. Parikh, D.; Riascos-Bernal, D.F.; Egaña-Gorroño, L.; Jayakumar, S.; Almonte, V.; Chinnasamy, P.; Sibinga, N.E.S. Allograft inflammatory factor-1-like is not essential for age dependent weight gain or HFD-induced obesity and glucose insensitivity. Sci. Rep. 2020, 10, 3594. [Google Scholar] [CrossRef]
  86. Allott, E.H.; Lysaght, J.; Cathcart, M.C.; Donohoe, C.L.; Cummins, R.; McGarrigle, S.A.; Kay, E.; Reynolds, J.V.; Pidgeon, G.P. MMP9 expression in oesophageal adenocarcinoma is upregulated with visceral obesity and is associated with poor tumour differentiation. Mol. Carcinog. 2013, 52, 144–154. [Google Scholar] [CrossRef]
  87. Awaya, T.; Yokosaki, Y.; Yamane, K.; Usui, H.; Kohno, N.; Eboshida, A. Gene-environment Association of an ITGB2 Sequence Variant With Obesity in Ethnic Japanese. Obesity 2008, 16, 1463–1466. [Google Scholar] [CrossRef]
  88. Mathews, J.A.; Wurmbrand, A.P.; Ribeiro, L.; Neto, F.L.; Shore, S.A. Induction of IL-17A Precedes Development of Airway Hyperresponsiveness during Diet-Induced Obesity and Correlates with Complement Factor D. Front. Immunol. 2014, 5, 440. [Google Scholar] [CrossRef]
  89. Cero, C.; Razzoli, M.; Han, R.; Sahu, B.S.; Patricelli, J.; Guo, Z.; Zaidman, N.A.; Miles, J.M.; O’Grady, S.M.; Bartolomucci, A. The neuropeptide TLQP-21 opposes obesity via C3aR1-mediated enhancement of adrenergic-induced lipolysis. Mol. Metab. 2016, 6, 148–158. [Google Scholar] [CrossRef]
  90. Mukherjee, R.; Kim, S.W.; Park, T.; Choi, M.S.; Yun, J.W. Targeted inhibition of galectin 1 by thiodigalactoside dramatically reduces body weight gain in diet-induced obese rats. Int. J. Obes. 2015, 39, 1349–1358. [Google Scholar] [CrossRef]
  91. Leite, F.; Leite, Â.; Santos, A.; Lima, M.; Barbosa, J.; Cosentino, M.; Ribeiro, L. Predictors of Subclinical Inflammatory Obesity: Plasma Levels of Leptin, Very Low-Density Lipoprotein Cholesterol and CD14 Expression of CD16+ Monocytes. Obes. Facts 2017, 10, 308–322. [Google Scholar] [CrossRef]
  92. Andrade, V.L.; Petruceli, E.; Belo, V.A.; Andrade-Fernandes, C.M.; Russi, C.V.C.; Bosco, A.A.; Tanus-Santos, J.E.; Sandrim, V.C. Evaluation of plasmatic MMP-8, MMP-9, TIMP-1 and MPO levels in obese and lean women. Clin. Biochem. 2012, 45, 412–415. [Google Scholar] [CrossRef]
  93. Martínez-García, M.; Ojeda-Ojeda, M.; Rodríguez-Martín, E.; Insenser, M.; Moncayo, S.; Álvarez-Blasco, F.; Luque-Ramírez, M.; Escobar-Morreale, H.F. TLR2 and TLR4 Surface and Gene Expression in White Blood Cells after Fasting and Oral Glucose, Lipid and Protein Challenges: Influence of Obesity and Sex Hormones. Biomolecules 2020, 10, 111. [Google Scholar] [CrossRef]
  94. Jamka, M.; Kaczmarek, N.; Mądry, E.; Krzyżanowska-Jankowska, P.; Bajerska, J.; Kręgielska-Narożna, M.; Bogdański, P.; Walkowiak, J. Metabolic Health in Obese Subjects—Is There a Link to Lactoferrin and Lactoferrin Receptor-Related Gene Polymorphisms? Nutrients 2020, 12, 2843. [Google Scholar] [CrossRef]
  95. Peplonska, B.; Bukowska, A.; Wieczorek, E.; Przybek, M.; Zienolddiny, S.; Reszka, E. Rotating night work, lifestyle factors, obesity and promoter methylation in BRCA1 and BRCA2 genes among nurses and midwives. PLoS ONE 2017, 12, e0178792. [Google Scholar] [CrossRef]
  96. Moreno-Santos, I.; Castellano-Castillo, D.; Lara, M.F.; Fernandez-Garcia, J.C.; Tinahones, F.J.; Macias-Gonzalez, M. IGFBP-3 Interacts with the Vitamin D Receptor in Insulin Signaling Associated with Obesity in Visceral Adipose Tissue. Int. J. Mol. Sci. 2017, 18, 2349. [Google Scholar] [CrossRef]
  97. Sun, L.; Liu, Y.-L.; Ye, F.; Xie, J.-W.; Zeng, J.-W.; Qin, L.; Xue, J.; Wang, Y.-T.; Guo, K.-M.; Ma, M.-M.; et al. Free fatty acid-induced H2O2 activates TRPM2 to aggravate endothelial insulin resistance via Ca2+-dependent PERK/ATF4/TRB3 cascade in obese mice. Free. Radic. Biol. Med. 2019, 143, 288–299. [Google Scholar] [CrossRef]
  98. Richter, M.; Murtaza, N.; Scharrenberg, R.; White, S.H.; Johanns, O.; Walker, S.; Yuen, R.K.C.; Schwanke, B.; Bedürftig, B.; Henis, M.; et al. Altered TAOK2 activity causes autism-related neurodevelopmental and cognitive abnormalities through RhoA signaling. Mol. Psychiatry 2019, 24, 1329–1350. [Google Scholar] [CrossRef]
  99. Suchkova, I.O.; Borisova, E.V.; Patkin, E.L. Length Polymorphism and Methylation Status of UPS29 Minisatellite of the ACAP3 Gene as Molecular Biomarker of Epilepsy. Sex Differences in Seizure Types and Symptoms. Int. J. Mol. Sci. 2020, 21, E9206. [Google Scholar] [CrossRef]
  100. Qureshi, M.; Hatem, M.; Alroughani, R.; Jacob, S.P.; Al-Temaimi, R.A. PLXNA3 Variant rs5945430 is Associated with Severe Clinical Course in Male Multiple Sclerosis Patients. NeuroMolecular Med. 2017, 19, 286–292. [Google Scholar] [CrossRef]
  101. Wang, H.; Sun, F.-R.; Tan, L.; Zhang, W.; Wang, Z.-X.; Jiang, T.; Yu, J.-T.; Tan, L. Association study of the PLXNA4 gene with the risk of Alzheimer’s disease. Ann. Transl. Med. 2016, 4, 108. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, N.; Ma, Q.; Peng, P.; Yu, Y.; Xu, S.; Wang, G.; Ying, Z.; Wang, H. Autophagy and Ubiquitin-Proteasome System Coordinate to Regulate the Protein Quality Control of Neurodegenerative Disease-Associated DCTN1. Neurotox. Res. 2020, 37, 48–57. [Google Scholar] [CrossRef] [PubMed]
  103. Aoki-Suzuki, M.; Yamada, K.; Meerabux, J.; Iwayama-Shigeno, Y.; Ohba, H.; Iwamoto, K.; Takao, H.; Toyota, T.; Suto, Y.; Nakatani, N.; et al. A family-based association study and gene expression analyses of netrin-G1 and -G2 genes in schizophrenia. Biol. Psychiatry 2005, 57, 382–393. [Google Scholar] [CrossRef] [PubMed]
  104. Ohno, K.; Ohkawara, B.; Ito, M. Agrin-LRP4-MuSK signaling as a therapeutic target for myasthenia gravis and other neuromuscular disorders. Expert Opin. Ther. Targets 2017, 21, 949–958. [Google Scholar] [CrossRef]
  105. Rahman, S.; Copeland, W.C. POLG-related disorders and their neurological manifestations. Nat. Rev. Neurol. 2019, 15, 40–52. [Google Scholar] [CrossRef]
  106. Congiu, C.; Minelli, A.; Bonvicini, C.; Bortolomasi, M.; Sartori, R.; Maj, C.; Scassellati, C.; Maina, G.; Trabucchi, L.; Segala, M.; et al. The role of the potassium channel gene KCNK2 in major depressive disorder. Psychiatry Res. 2015, 225, 489–492. [Google Scholar] [CrossRef]
  107. Ji, H.; Wang, Y.; Liu, G.; Xu, X.; Dai, D.; Chen, Z.; Zhou, D.; Zhou, X.; Han, L.; Li, Y.; et al. OPRK1 promoter hypermethylation increases the risk of Alzheimer’s disease. Neurosci. Lett. 2015, 606, 24–29. [Google Scholar] [CrossRef]
  108. Wollmer, M.A.; Kapaki, E.; Hersberger, M.; Muntwyler, J.; Brunner, F.; Tsolaki, M.; Akatsu, H.; Kosaka, K.; Michikawa, M.; Molyva, D.; et al. Ethnicity-dependent genetic association of ABCA2 with sporadic Alzheimer’s disease. Am. J. Med. Genet. Part B: Neuropsychiatr. Genet. 2006, 141, 534–536. [Google Scholar] [CrossRef]
  109. Yamazaki, K.; Yoshino, Y.; Mori, T.; Yoshida, T.; Ozaki, Y.; Sao, T.; Mori, Y.; Ochi, S.; Iga, J.-I.; Ueno, S.-I. Gene Expression and Methylation Analysis of ABCA7 in Patients with Alzheimer’s Disease. J. Alzheimer Dis. 2017, 57, 171–181. [Google Scholar] [CrossRef]
  110. Bardien, S.; Lesage, S.; Brice, A.; Carr, J. Genetic characteristics of leucine-rich repeat kinase 2 (LRRK2) associated Parkinson’s disease. Park. Relat. Disord. 2011, 17, 501–508. [Google Scholar] [CrossRef]
  111. Bolla, A.C.; Valente, T.; Miguez, A.; Brito, V.; Gines, S.; Solà, C.; Straccia, M.; Canals, J.M. CD200 is up-regulated in R6/1 transgenic mouse model of Huntington’s disease. PLoS ONE 2019, 14, e0224901. [Google Scholar] [CrossRef]
  112. Horvath, G.A.; Tarailo-Graovac, M.; Bartel, T.; Race, S.; Van Allen, M.I.; Blydt-Hansen, I.; Ross, C.J.; Wasserman, W.W.; Connolly, M.B.; van Karnebeek, C.D.M. Improvement of Self-Injury with Dopamine and Serotonin Replacement Therapy in a Patient with a Hemizygous PAK3 Mutation: A New Therapeutic Strategy for Neuropsychiatric Features of an Intellectual Disability Syndrome. J. Child Neurol. 2018, 33, 106–113. [Google Scholar] [CrossRef] [Green Version]
  113. Watanabe, Y.; Nunokawa, A.; Kaneko, N.; Arinami, T.; Ujike, H.; Inada, T.; Iwata, N.; Kunugi, H.; Itokawa, M.; Otowa, T.; et al. A two-stage case–control association study of PADI2 with schizophrenia. J. Hum. Genet. 2009, 54, 430–432. [Google Scholar] [CrossRef]
  114. Kushima, I.; Nakamura, Y.; Aleksic, B.; Ikeda, M.; Ito, Y.; Shiino, T.; Okochi, T.; Fukuo, Y.; Ujike, H.; Suzuki, M.; et al. Resequencing and Association Analysis of the KALRN and EPHB1 Genes and Their Contribution to Schizophrenia Susceptibility. Schizophr. Bull. 2012, 38, 552–560. [Google Scholar] [CrossRef]
  115. Grünblatt, E.; Reif, A.; Jungwirth, S.; Galimberti, D.; Weber, H.; Scarpini, E.; Sauer, C.; Wichart, I.; Rainer, M.K.; Huber, K.; et al. Genetic variation in the choline O-acetyltransferase gene in depression and Alzheimer’s disease: The VITA and Milano studies. J. Psychiatr. Res. 2011, 45, 1250–1256. [Google Scholar] [CrossRef]
  116. Sato, D.X.; Kawata, M. Positive and balancing selection on SLC18A1 gene associated with psychiatric disorders and human-unique personality traits. Evol. Lett. 2018, 2, 499–510. [Google Scholar] [CrossRef]
  117. Kundu, A.; Ramakrishnan, P.; Rajendran, A.; Dharwar, N.V.; Anbarasu, A. Analysis of non-synonymous single-nucleotide polymorphisms and population variability of PLD2 gene associated with hypertension. Int. J. Bioinform. Res. Appl. 2013, 9, 227–241. [Google Scholar] [CrossRef]
  118. Jain, M.; Mann, T.D.; Stulić, M.; Rao, S.P.; Kirsch, A.; Pullirsch, D.; Strobl, X.; Rath, C.; Reissig, L.; Moreth, K.; et al. RNA editing of Filamin A pre- mRNA regulates vascular contraction and diastolic blood pressure. EMBO J. 2018, 37, e94813. [Google Scholar] [CrossRef]
  119. Baptista, R.; Marques, C.; Catarino, S.; Enguita, F.J.; Costa, M.C.; Matafome, P.; Zuzarte, M.; Castro, G.; Reis, A.; Monteiro, P.; et al. MicroRNA-424(322) as a new marker of disease progression in pulmonary arterial hypertension and its role in right ventricular hypertrophy by targeting SMURF1. Cardiovasc. Res. 2018, 114, 53–64. [Google Scholar] [CrossRef]
  120. Fu, Q.-L.; Hu, B.; Li, X.; Shao, Z.; Shi, J.-B.; Wu, W.; So, K.-F.; Mi, S. LINGO-1 negatively regulates TrkB phosphorylation after ocular hypertension. Eur. J. Neurosci. 2010, 31, 1091–1097. [Google Scholar] [CrossRef]
  121. Scholl, U.I.; Stölting, G.; Nelson-Williams, C.; Vichot, A.A.; Choi, M.; Loring, E.; Prasad, M.L.; Goh, G.; Carling, T.; Juhlin, C.C.; et al. Recurrent gain of function mutation in calcium channel CACNA1H causes early-onset hypertension with primary aldosteronism. Elife 2015, 4, e06315. [Google Scholar] [CrossRef] [PubMed]
  122. Glorioso, N.; Herrera, V.L.; Didishvili, T.; Ortu, M.F.; Zaninello, R.; Fresu, G.; Argiolas, G.; Troffa, C.; Ruiz-Opazo, N. Sex-Specific Effects of NLRP6/AVR and ADM Loci on Susceptibility to Essential Hypertension in a Sardinian Population. PLoS ONE 2013, 8, e77562. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Zha, L.; Zhou, J.; Li, T.; Luo, H.; Zhang, M.; Li, S.; Yu, Z. NLRC3 inhibits MCT-induced pulmonary hypertension in rats via attenuating PI3K activation. J. Cell. Physiol. 2019, 234, 15963–15976. [Google Scholar] [CrossRef] [PubMed]
  124. Zhang, Y.; Teng, F.; Han, X.; Li, P.; Yan, X.; Guo, S.; Li, H. Selective blocking of CXCR2 prevents and reverses atrial fibrillation in spontaneously hypertensive rats. J. Cell. Mol. Med. 2020, 24, 11272–11282. [Google Scholar] [CrossRef] [PubMed]
  125. Weiss, S.; Rosendahl, A.; Czesla, D.; Meyer-Schwesinger, C.; Stahl, R.A.K.; Ehmke, H.; Kurts, C.; Zipfel, P.F.; Köhl, J.; Wenzel, U.O. The complement receptor C5aR1 contributes to renal damage but protects the heart in angiotensin II-induced hypertension. Am. J. Physiol. Physiol. 2016, 310, F1356–F1365. [Google Scholar] [CrossRef]
  126. Sauzeau, V.; Jerkic, M.; López-Novoa, J.M.; Bustelo, X.R. Loss of Vav2 Proto-Oncogene Causes Tachycardia and Cardiovascular Disease in Mice. Mol. Biol. Cell 2007, 18, 943–952. [Google Scholar] [CrossRef]
  127. Xu, X.; Tan, X.; Tampe, B.; Nyamsuren, G.; Liu, X.; Maier, L.S.; Sossalla, S.; Kalluri, R.; Zeisberg, M.; Hasenfuss, G.; et al. Epigenetic balance of aberrant Rasal1 promoter methylation and hydroxymethylation regulates cardiac fibrosis. Cardiovasc. Res. 2015, 105, 279–291. [Google Scholar] [CrossRef]
  128. Hirota, H.; Izumi, M.; Hamaguchi, T.; Sugiyama, S.; Murakami, E.; Kunisada, K.; Fujio, Y.; Oshima, Y.; Nakaoka, Y.; Yamauchi-Takihara, K. Circulating interleukin-6 family cytokines and their receptors in patients with congestive heart failure. Heart Vessel. 2004, 19, 237–241. [Google Scholar] [CrossRef]
  129. Alharatani, R.; Ververi, A.; Beleza-Meireles, A.; Ji, W.; Mis, E.; Patterson, Q.T.; Griffin, J.N.; Bhujel, N.; Chang, C.A.; Dixit, A.; et al. Novel truncating mutations in CTNND1 cause a dominant craniofacial and cardiac syndrome. Hum. Mol. Genet. 2020, 29, 1900–1921. [Google Scholar] [CrossRef]
  130. Beitelshees, A.L.; Navare, H.; Wang, D.; Gong, Y.; Wessel, J.; Moss, J.I.; Langaee, T.Y.; Cooper-DeHoff, R.M.; Sadee, W.; Pepine, C.J.; et al. CACNA1C Gene Polymorphisms, Cardiovascular Disease Outcomes, and Treatment Response. Circ. Cardiovasc. Genet. 2009, 2, 362–370. [Google Scholar] [CrossRef]
  131. Zhu, J.; Chen, T.; Yang, L.; Li, Z.; Wong, M.M.; Zheng, X.; Pan, X.; Zhang, L.; Yan, H. Regulation of MicroRNA-155 in Atherosclerotic Inflammatory Responses by Targeting MAP3K10. PLoS ONE 2012, 7, e46551. [Google Scholar] [CrossRef]
  132. Gil-Cayuela, C.; López, A.; Martinez-Dolz, L.; Juanatey, J.R.G.; Lago, F.; Roselló-Lletí, E.; Rivera, M.; Portolés, M. The altered expression of autophagy-related genes participates in heart failure: NRBP2 and CALCOCO2 are associated with left ventricular dysfunction parameters in human dilated cardiomyopathy. PLoS ONE 2019, 14, e0215818. [Google Scholar] [CrossRef]
  133. Liu, H.; El Zein, L.; Kruse, M.; Guinamard, R.; Beckmann, A.; Bozio, A.; Kurtbay, G.; Mégarbané, A.; Ohmert, I.; Blaysat, G.; et al. Gain-of-Function Mutations in TRPM4 Cause Autosomal Dominant Isolated Cardiac Conduction Disease. Circ. Cardiovasc. Genet. 2010, 3, 374–385. [Google Scholar] [CrossRef]
  134. Xie, M.; Hu, C.; Li, D.; Li, S. MicroRNA-377 Alleviates Myocardial Injury Induced by Hypoxia/Reoxygenation via Downregulating LILRB2 Expression. Dose-Response 2020, 18, 1559325820936124. [Google Scholar] [CrossRef]
  135. Kroupis, C.; Theodorou, M.; Chaidaroglou, A.; Dalamaga, M.; Oliveira, S.C.; Cokkinos, D.V.; Degiannis, D.; Manginas, A. The Association Between a CommonFCGR2APolymorphism and C-Reactive Protein and Coronary Artery Disease Revisited. Genet. Test. Mol. Biomark. 2010, 14, 839–846. [Google Scholar] [CrossRef]
  136. López-Mejías, R.; Genre, F.; García-Bermúdez, M.; Ubilla, B.; Castañeda, S.; Llorca, J.; González-Juanatey, C.; Corrales, A.; Miranda-Filloy, J.A.; Pina, T.; et al. Lack of Association between ABO, PPAP2B, ADAMST7, PIK3CG, and EDNRA and Carotid Intima-Media Thickness, Carotid Plaques, and Cardiovascular Disease in Patients with Rheumatoid Arthritis. Mediat. Inflamm. 2014, 2014, 756279. [Google Scholar] [CrossRef]
  137. Koppensteiner, R.; Kaider, A.; Eichelberger, B.; Mannhalter, C.; Panzer, S.; Gremmel, T. Impact of variables of the P-selectin–P-selectin glycoprotein ligand-1 axis on leukocyte-platelet interactions in cardiovascular disease. Thromb. Haemost. 2015, 113, 806–812. [Google Scholar] [CrossRef]
  138. Yamada, S.; Guo, X. Peroxiredoxin 4 (PRDX4): Its critical in vivo roles in animal models of metabolic syndrome ranging from atherosclerosis to nonalcoholic fatty liver disease. Pathol. Int. 2018, 68, 91–101. [Google Scholar] [CrossRef]
  139. Petri, M.H.; Laguna-Fernández, A.; Gonzalez-Diez, M.; Paulsson-Berne, G.; Hansson, G.K.; Bäck, M. The role of the FPR2/ALX receptor in atherosclerosis development and plaque stability. Cardiovasc. Res. 2015, 105, 65–74. [Google Scholar] [CrossRef]
  140. DeFilippis, A.P.; Chernyavskiy, I.; Amraotkar, A.R.; Trainor, P.J.; Kothari, S.; Ismail, I.; Hargis, C.W.; Korley, F.K.; Leibundgut, G.; Tsimikas, S.; et al. Circulating levels of plasminogen and oxidized phospholipids bound to plasminogen distinguish between atherothrombotic and non-atherothrombotic myocardial infarction. J. Thromb. Thrombolysis 2016, 42, 61–76. [Google Scholar] [CrossRef]
  141. Rocca, C.; Pasqua, T.; Boukhzar, L.; Anouar, Y.; Angelone, T. Progress in the emerging role of selenoproteins in cardiovascular disease: Focus on endoplasmic reticulum-resident selenoproteins. Cell. Mol. Life Sci. 2019, 76, 3969–3985. [Google Scholar] [CrossRef] [PubMed]
  142. Tur, M.K.; Etschmann, B.; Benz, A.; Leich, E.; Waller, C.; Schuh, K.; Rosenwald, A.; Ertl, G.; Kienitz, A.; Haaf, A.T.; et al. The 140-kD Isoform of CD56 (NCAM1) Directs the Molecular Pathogenesis of Ischemic Cardiomyopathy. Am. J. Pathol. 2013, 182, 1205–1218. [Google Scholar] [CrossRef] [PubMed]
  143. Yu, P.-F.; Pang, L.-L.; Mao, Q.-S.; Zou, S.-C.; Shi, Y.; Lin, D.-J. Dose dependency PM2.5 aggravated airway inflammation in asthmatic mice via down-regulating expression of ITGB4. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 1688–1697. [Google Scholar] [PubMed]
  144. McGeachie, M.J.; Wu, A.C.; Tse, S.M.; Clemmer, G.L.; Sordillo, J.; Himes, B.E.; Lasky-Su, J.; Chase, R.P.; Martinez, F.D.; Weeke, P.; et al. CTNNA3 and SEMA3D: Promising loci for asthma exacerbation identified through multiple genome-wide association studies. J. Allergy Clin. Immunol. 2015, 136, 1503–1510. [Google Scholar] [CrossRef]
  145. Jasek, M.; Obojski, A.; Mańczak, M.; Wiśniewski, A.; Winiarska, B.; Małolepszy, J.; Jutel, M.; Łuszczek, W.; Kuśnierczyk, P. Are Single Nucleotide Polymorphisms of the Immunoglobulin A Fc Receptor Gene Associated with Allergic Asthma? Int. Arch. Allergy Immunol. 2004, 135, 325–331. [Google Scholar] [CrossRef]
  146. Cahill, K.N.; Katz, H.R.; Cui, J.; Lai, J.; Kazani, S.; Crosby-Thompson, A.; Garofalo, D.; Castro, M.; Jarjour, N.; DiMango, E.; et al. KIT Inhibition by Imatinib in Patients with Severe Refractory Asthma. N. Engl. J. Med. 2017, 376, 1911–1920. [Google Scholar] [CrossRef]
  147. Banskar, S.; Detzner, A.A.; Juarez-Rodriguez, M.D.; Hozo, I.; Gupta, D.; Dziarski, R. The Pglyrp1-Regulated Microbiome Enhances Experimental Allergic Asthma. J. Immunol. 2019, 203, 3113–3125. [Google Scholar] [CrossRef]
  148. Hunninghake, G.M.; Chu, J.-H.; Sharma, S.S.; Cho, M.H.; Himes, B.E.; Rogers, A.J.; Murphy, A.; Carey, V.J.; Raby, B.A. The CD4+ T-cell transcriptome and serum IgE in asthma: IL17RB and the role of sex. BMC Pulm. Med. 2011, 11, 17. [Google Scholar] [CrossRef]
  149. Ungvári, I.; Hadadi, E.; Virág, V.; Bikov, A.; Nagy, A.; Semsei, A.F.; Gálffy, G.; Tamási, L.; Horvath, I.; Szalai, C. Implication of BIRC5 in asthma pathogenesis. Int. Immunol. 2012, 24, 293–301. [Google Scholar] [CrossRef]
  150. Yucesoy, B.; Kashon, M.L.; Johnson, V.J.; Lummus, Z.L.; Fluharty, K.; Gautrin, D.; Cartier, A.; Boulet, L.-P.; Sastre, J.; Quirce, S.; et al. Genetic variants in TNFα, TGFB1, PTGS1 and PTGS2 genes are associated with diisocyanate-induced asthma. J. Immunotoxicol. 2016, 13, 119–126. [Google Scholar] [CrossRef]
  151. Vila-Bedmar, R.; Cruces-Sande, M.; Lucas, E.; Willemen, H.L.D.M.; Heijnen, C.J.; Kavelaars, A.; Mayor, F.; Murga, C. Reversal of diet-induced obesity and insulin resistance by inducible genetic ablation of GRK2. Sci. Signal. 2015, 8, ra73. [Google Scholar] [CrossRef]
  152. Grarup, N.; Moltke, I.; Andersen, M.K.; Dalby, M.; Vitting-Seerup, K.; Kern, T.; Mahendran, Y.; Jørsboe, E.; Larsen, C.V.L.; Dahl-Petersen, I.; et al. Loss-of-function variants in ADCY3 increase risk of obesity and type 2 diabetes. Nat. Genet. 2018, 50, 172–174. [Google Scholar] [CrossRef]
  153. Berndt, J.; Kovacs, P.; Ruschke, K.; Klöting, N.; Fasshauer, M.; Schön, M.R.; Körner, A.; Stumvoll, M.; Blüher, M. Fatty acid synthase gene expression in human adipose tissue: Association with obesity and type 2 diabetes. Diabetologia 2007, 50, 1472–1480. [Google Scholar] [CrossRef]
  154. Mannerås-Holm, L.; Kirchner, H.; Björnholm, M.; Chibalin, A.V.; Zierath, J.R. mRNA expression of diacylglycerol kinase isoforms in insulin-sensitive tissues: Effects of obesity and insulin resistance. Physiol. Rep. 2015, 3, e12372. [Google Scholar] [CrossRef]
  155. Ghoshal, S.; Zhu, Q.; Asteian, A.; Lin, H.; Xu, H.; Ernst, G.; Barrow, J.C.; Xu, B.; Cameron, M.D.; Kamenecka, T.M.; et al. TNP [N2-(m-Trifluorobenzyl), N6-(p-nitrobenzyl)purine] ameliorates diet induced obesity and insulin resistance via inhibition of the IP6K1 pathway. Mol. Metab. 2016, 5, 903–917. [Google Scholar] [CrossRef]
  156. Pietrani, N.T.; Ferreira, C.N.; Rodrigues, K.F.; Perucci, L.O.; Carneiro, F.S.; Bosco, A.A.; Oliveira, M.C.; Pereira, S.S.; Teixeira, A.L.; Alvarez-Leite, J.I.; et al. Proresolving protein Annexin A1: The role in type 2 diabetes mellitus and obesity. Biomed. Pharmacother. 2018, 103, 482–489. [Google Scholar] [CrossRef]
  157. Van Diepen, J.A.; Robben, J.H.; Hooiveld, G.J.; Carmone, C.; Alsady, M.; Boutens, L.; Bekkenkamp-Grovenstein, M.; Hijmans, A.; Engelke, U.F.H.; Wevers, R.A.; et al. SUCNR1-mediated chemotaxis of macrophages aggravates obesity-induced inflammation and diabetes. Diabetologia 2017, 60, 1304–1313. [Google Scholar] [CrossRef]
  158. De Brito, G.; Lupinacci, F.C.; Beraldo, F.H.; Santos, T.G.; Roffé, M.; Lopes, M.H.; de Lima, V.C.; Martins, V.R.; Hajj, G.N. Loss of prion protein is associated with the development of insulin resistance and obesity. Biochem. J. 2017, 474, 2981–2991. [Google Scholar] [CrossRef]
  159. Dang, Z.; Avolio, E.; Thomas, A.C.; Faulkner, A.; Beltrami, A.P.; Cervellin, C.; Carrizzo, A.; Maciag, A.; Gu, Y.; Ciaglia, E.; et al. Transfer of a human gene variant associated with exceptional longevity improves cardiac function in obese type 2 diabetic mice through induction of the SDF-1/CXCR4 signalling pathway. Eur. J. Heart Fail. 2020, 22, 1568–1581. [Google Scholar] [CrossRef]
  160. Catalan, V.; Gomez-Ambrosi, J.; Rodríguez, A.; Silva, C.; Rotellar, F.; Gil, M.J.; Cienfuegos, J.; Salvador, J.; Frühbeck, G. Expression of caveolin-1 in human adipose tissue is upregulated in obesity and obesity-associated type 2 diabetes mellitus and related to inflammation. Clin. Endocrinol. 2008, 68, 213–219. [Google Scholar] [CrossRef]
  161. Elkhidir, A.E.; Eltaher, H.B.; Mohamed, A.O. Association of lipocalin-2 level, glycemic status and obesity in type 2 diabetes mellitus. BMC Res. Notes 2017, 10, 285. [Google Scholar] [CrossRef] [PubMed]
  162. Catalán, V.; Gómez-Ambrosi, J.; Pastor, C.; Rotellar, F.; Silva, C.; Rodríguez, A.; Gil, M.J.; Cienfuegos, J.A.; Salvador, J.; Vendrell, J.; et al. Influence of Morbid Obesity and Insulin Resistance on Gene Expression Levels of AQP7 in Visceral Adipose Tissue and AQP9 in Liver. Obes. Surg. 2008, 18, 695–701. [Google Scholar] [CrossRef] [PubMed]
  163. Ingallinella, P.; Peier, A.M.; Pocai, A.; Di Marco, A.; Desai, K.; Zytko, K.; Qian, Y.; Du, X.; Cellucci, A.; Monteagudo, E.; et al. PEGylation of Neuromedin U yields a promising candidate for the treatment of obesity and diabetes. Bioorganic Med. Chem. 2012, 20, 4751–4759. [Google Scholar] [CrossRef] [PubMed]
  164. Wittrisch, S.; Klöting, N.; Mörl, K.; Chakaroun, R.; Blüher, M.; Beck-Sickinger, A.G. NPY1R-targeted peptide-mediated delivery of a dual PPARα/γ agonist to adipocytes enhances adipogenesis and prevents diabetes progression. Mol. Metab. 2020, 31, 163–180. [Google Scholar] [CrossRef]
  165. Bódis, K.; Kahl, S.; Simon, M.-C.; Zhou, Z.; Sell, H.; Knebel, B.; Tura, A.; Strassburger, K.; Burkart, V.; Müssig, K.; et al. Reduced expression of stearoyl-CoA desaturase-1, but not free fatty acid receptor 2 or 4 in subcutaneous adipose tissue of patients with newly diagnosed type 2 diabetes mellitus. Nutr. Diabetes 2018, 8, 49. [Google Scholar] [CrossRef]
  166. Sánchez-Infantes, D.; White, U.A.; Elks, C.M.; Morrison, R.F.; Gimble, J.M.; Considine, R.V.; Ferrante, A.W.; Ravussin, E.; Stephens, J.M. Oncostatin M Is Produced in Adipose Tissue and Is Regulated in Conditions of Obesity and Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2014, 99, E217–E225. [Google Scholar] [CrossRef]
  167. Subramanian, S.; Pallati, P.K.; Sharma, P.; Agrawal, D.K.; Nandipati, K.C. Significant association of TREM-1 with HMGB1, TLRs and RAGE in the pathogenesis of insulin resistance in obese diabetic populations. Am. J. Transl. Res. 2017, 9, 3224–3244. [Google Scholar]
  168. Trégouet, D.-A.; Groop, P.-H.; McGinn, S.; Forsblom, C.; Hadjadj, S.; Marre, M.; Parving, H.-H.; Tarnow, L.; Telgmann, R.; Godefroy, T.; et al. G/T Substitution in Intron 1 of the UNC13B Gene Is Associated with Increased Risk of Nephropathy in Patients With Type 1 Diabetes. Diabetes 2008, 57, 2843–2850. [Google Scholar] [CrossRef]
  169. Nomoto, H.; Pei, L.; Montemurro, C.; Rosenberger, M.; Furterer, A.; Coppola, G.; Nadel, B.; Pellegrini, M.; Gurlo, T.; Butler, P.C.; et al. Activation of the HIF1α/PFKFB3 stress response pathway in beta cells in type 1 diabetes. Diabetologia 2020, 63, 149–161. [Google Scholar] [CrossRef]
  170. Anjosa, Z.P.; Santos, M.M.S.; Rodrigues, N.J.; DE Lacerda, G.A.N.; Araujo, J.; Silva, J.D.A.; Tavares, N.D.A.C.; Guimarães, R.L.; Crovella, S.; Brandão, L.A.C. Polymorphism in ficolin-1 (FCN1) gene is associated with an earlier onset of type 1 diabetes mellitus in children and adolescents from northeast Brazil. J. Genet. 2016, 95, 1031–1034. [Google Scholar] [CrossRef]
  171. Paccagnini, D.; Sieswerda, L.; Rosu, V.; Masala, S.; Pacifico, A.; Gazouli, M.; Ikonomopoulos, J.; Ahmed, N.; Zanetti, S.; Sechi, L.A. Linking Chronic Infection and Autoimmune Diseases: Mycobacterium avium Subspecies paratuberculosis, SLC11A1 Polymorphisms and Type-1 Diabetes Mellitus. PLoS ONE 2009, 4, e7109. [Google Scholar] [CrossRef]
  172. Cinaglia, P.; Cannataro, M. Network alignment and motif discovery in dynamic networks. Netw. Model. Anal. Health Inform. Bioinform. 2022, 11, 38. [Google Scholar] [CrossRef]
  173. Cinaglia, P.; Guzzi, P.H.; Veltri, P. INTEGRO: An algorithm for data-integration and disease-gene association. In Proceedings of the 2018 IEEE International Conference on Bioinformatics and Biomedicine (BIBM), Madrid, Spain, 3–6 December 2018; pp. 2076–2081. [Google Scholar] [CrossRef]
  174. Freedman, B.I.; Hicks, P.J.; Bostrom, M.A.; Comeau, M.E.; Divers, J.; Bleyer, A.J.; Kopp, J.B.; Winkler, C.A.; Nelson, G.W.; Langefeld, C.D.; et al. Non-muscle myosin heavy chain 9 gene MYH9 associations in African Americans with clinically diagnosed type 2 diabetes mellitus-associated ESRD. Nephrol. Dial. Transplant. 2009, 24, 3366–3371. [Google Scholar] [CrossRef]
  175. Zhao, H.; Ma, L.; Yan, M.; Wang, Y.; Zhao, T.; Zhang, H.; Liu, P.; Liu, Y.; Li, P. Association between MYH9 and APOL1 Gene Polymorphisms and the Risk of Diabetic Kidney Disease in Patients with Type 2 Diabetes in a Chinese Han Population. J. Diabetes Res. 2018, 2018, 5068578. [Google Scholar] [CrossRef]
  176. Ling, C.; Cai, C.; Chang, B.; Shi, W.; Wei, F.; Yu, P.; Chen, L.; Li, W. MYH9 gene polymorphisms may be associated with cerebrovascular blood flow in patients with type 2 diabetes. Genet. Mol. Res. 2015, 14, 1008–1016. [Google Scholar] [CrossRef]
  177. Huang, Y.; Han, X.; Chang, T.; Li, F.-F.; Chen, X.; She, Y.-Q. Serum ErbB2 concentration positively correlated to the glycemic variations in newly diagnosed Type 2 diabetic patients. Sci. Rep. 2022, 12, 4940. [Google Scholar] [CrossRef]
  178. De Kay, J.T.; Carver, J.; Shevenell, B.; Kosta, A.M.; Tsibulnikov, S.; Certo, E.; Sawyer, D.B.; Ryzhov, S.; Robich, M.P. Decreased expression of ErbB2 on left ventricular epicardial cells in patients with diabetes mellitus. Cell. Signal. 2022, 96, 110360. [Google Scholar] [CrossRef]
  179. Akhtar, S.; Yousif, M.H.M.; Chandrasekhar, B.; Benter, I.F. Activation of EGFR/ERBB2 via Pathways Involving ERK1/2, P38 MAPK, AKT and FOXO Enhances Recovery of Diabetic Hearts from Ischemia-Reperfusion Injury. PLoS ONE 2012, 7, e39066. [Google Scholar] [CrossRef]
  180. Akhtar, S.; Yousif, M.; Dhaunsi, G.S.; Sarkhouh, F.; Chandrasekhar, B.; Attur, S.; Benter, I.F. Activation of ErbB2 and Downstream Signalling via Rho Kinases and ERK1/2 Contributes to Diabetes-Induced Vascular Dysfunction. PLoS ONE 2013, 8, e67813. [Google Scholar] [CrossRef]
  181. Wei, H.; Qu, H.; Wang, H.; Ji, B.; Ding, Y.; Liu, D.; Duan, Y.; Liang, H.; Peng, C.; Xiao, X.; et al. 1,25-Dihydroxyvitamin-D3 prevents the development of diabetic cardiomyopathy in type 1 diabetic rats by enhancing autophagy via inhibiting the β-catenin/TCF4/GSK-3β/mTOR pathway. J. Steroid Biochem. Mol. Biol. 2017, 168, 71–90. [Google Scholar] [CrossRef]
  182. Kim, S.; Kim, I.; Cho, W.; Oh, G.T.; Park, Y.M. Vimentin Deficiency Prevents High-Fat Diet-Induced Obesity and Insulin Resistance in Mice. Diabetes Metab. J. 2020, 45, 97–108. [Google Scholar] [CrossRef] [PubMed]
  183. Roefs, M.M.; Carlotti, F.; Jones, K.; Wills, H.; Hamilton, A.; Verschoor, M.; Durkin, J.M.W.; Perez, L.G.; Brereton, M.F.; McCulloch, L.; et al. Increased vimentin in human α- and β-cells in type 2 diabetes. J. Endocrinol. 2017, 233, 217–227. [Google Scholar] [CrossRef] [PubMed]
  184. Yang, S.; Xia, C.; Li, S.; Du, L.; Zhang, L.; Hu, Y. Mitochondrial dysfunction driven by the LRRK2-mediated pathway is associated with loss of Purkinje cells and motor coordination deficits in diabetic rat model. Cell Death Dis. 2014, 5, e1217. [Google Scholar] [CrossRef] [PubMed]
  185. Huang, M.-H.; Liu, Y.-F.; Nfor, O.N.; Hsu, S.-Y.; Lin, W.-Y.; Chang, Y.-S.; Liaw, Y.-P. Interactive Association Between Intronic Polymorphism (rs10506151) of the LRRK2 Gene and Type 2 Diabetes on Neurodegenerative Diseases. Pharm. Pers. Med. 2021, 14, 839–847. [Google Scholar] [CrossRef] [PubMed]
  186. Reinbothe, T.; Alkayyali, S.; Ahlqvist, E.; Tuomi, T.; Isomaa, B.; Lyssenko, V.; Renström, E. The human L-type calcium channel Cav1.3 regulates insulin release and polymorphisms in CACNA1D associate with type 2 diabetes. Diabetologia 2013, 56, 340–349. [Google Scholar] [CrossRef]
  187. Bonds, J.A.; Shetti, A.; Bheri, A.; Chen, Z.; Disouky, A.; Tai, L.; Mao, M.; Head, B.P.; Bonini, M.G.; Haus, J.M.; et al. Depletion of Caveolin-1 in Type 2 Diabetes Model Induces Alzheimer’s Disease Pathology Precursors. J. Neurosci. 2019, 39, 8576–8583. [Google Scholar] [CrossRef]
  188. Luo, M.; Xu, C.; Luo, Y.; Wang, G.; Wu, J.; Wan, Q. Circulating miR-103 family as potential biomarkers for type 2 diabetes through targeting CAV-1 and SFRP4. Acta Diabetol. 2020, 57, 309–322. [Google Scholar] [CrossRef]
  189. Oh, Y.S.; Lee, T.S.; Cheon, G.J.; Jang, I.-S.; Jun, H.-S.; Park, S.C. Modulation of Insulin Sensitivity and Caveolin-1 Expression by Orchidectomy in a Nonobese Type 2 Diabetes Animal Model. Mol. Med. 2011, 17, 4–11. [Google Scholar] [CrossRef]
  190. Oh, Y.S.; Khil, L.-Y.; Cho, K.A.; Ryu, S.J.; Ha, M.K.; Cheon, G.J.; Lee, T.S.; Yoon, J.-W.; Jun, H.-S.; Park, S.C. A potential role for skeletal muscle caveolin-1 as an insulin sensitivity modulator in ageing-dependent non-obese type 2 diabetes: Studies in a new mouse model. Diabetologia 2008, 51, 1025–1034. [Google Scholar] [CrossRef]
  191. Elçioğlu, K.H.; Kabasakal, L.; Çetinel, S.; Conturk, G.; Sezen, S.F.; Ayanoğlu-Dülger, G. Changes in caveolin-1 expression and vasoreactivity in the aorta and corpus cavernosum of fructose and streptozotocin-induced diabetic rats. Eur. J. Pharmacol. 2010, 642, 113–120. [Google Scholar] [CrossRef]
  192. Carrillo-Sepulveda, M.A.; Matsumoto, T. Phenotypic Modulation of Mesenteric Vascular Smooth Muscle Cells from Type 2 Diabetic Rats is Associated with Decreased Caveolin-1 Expression. Cell. Physiol. Biochem. 2014, 34, 1497–1506. [Google Scholar] [CrossRef]
  193. Stadion, M.; Schwerbel, K.; Graja, A.; Baumeier, C.; Rödiger, M.; Jonas, W.; Wolfrum, C.; Staiger, H.; Fritsche, A.; Häring, H.-U.; et al. Increased Ifi202b/IFI16 expression stimulates adipogenesis in mice and humans. Diabetologia 2018, 61, 1167–1179. [Google Scholar] [CrossRef] [Green Version]
  194. Mousa, U.; Onur, H.; Utkan, G. Is obesity always a risk factor for all breast cancer patients? c-erbB2 expression is significantly lower in obese patients with early stage breast cancer. Clin. Transl. Oncol. 2012, 14, 923–930. [Google Scholar] [CrossRef]
  195. Roh, E.; Yoo, H.J. The Role of Adipose Tissue Lipolysis in Diet-Induced Obesity: Focus on Vimentin. Diabetes Metab. J. 2021, 45, 43–45. [Google Scholar] [CrossRef]
  196. Abaj, F.; Saeedy, S.A.G.; Mirzaei, K. Mediation role of body fat distribution (FD) on the relationship between CAV1 rs3807992 polymorphism and metabolic syndrome in overweight and obese women. BMC Med. Genom. 2021, 14, 202. [Google Scholar] [CrossRef]
  197. Razani, B.; Combs, T.P.; Wang, X.B.; Frank, P.G.; Park, D.S.; Russell, R.G.; Li, M.; Tang, B.; Jelicks, L.A.; Scherer, P.E.; et al. Caveolin-1-deficient Mice Are Lean, Resistant to Diet-induced Obesity, and Show Hypertriglyceridemia with Adipocyte Abnormalities. J. Biol. Chem. 2002, 277, 8635–8647. [Google Scholar] [CrossRef]
  198. Pandey, V.; Vijayakumar, M.V.; Ajay, A.K.; Malvi, P.; Bhat, M.K. Diet-induced obesity increases melanoma progression: Involvement of Cav-1 and FASN. Int. J. Cancer 2012, 130, 497–508. [Google Scholar] [CrossRef]
  199. Czikora, I.; Feher, A.; Lucas, R.; Fulton, D.J.R.; Bagi, Z. Caveolin-1 prevents sustained angiotensin II-induced resistance artery constriction and obesity-induced high blood pressure. Am. J. Physiol. Circ. Physiol. 2015, 308, H376–H385. [Google Scholar] [CrossRef]
  200. Pecci, A.; Ma, X.; Savoia, A.; Adelstein, R.S. MYH9: Structure, functions and role of non-muscle myosin IIA in human disease. Gene 2018, 664, 152–167. [Google Scholar] [CrossRef]
  201. Holbro, T.; Beerli, R.R.; Maurer, F.; Koziczak, M.; Barbas, C.F.; Hynes, N.E. The ErbB2/ErbB3 heterodimer functions as an oncogenic unit: ErbB2 requires ErbB3 to drive breast tumor cell proliferation. Proc. Natl. Acad. Sci. USA 2003, 100, 8933–8938. [Google Scholar] [CrossRef]
  202. Forrest, M.P.; Hill, M.J.; Quantock, A.J.; Martin-Rendon, E.; Blake, D.J. The emerging roles of TCF4 in disease and development. Trends Mol. Med. 2014, 20, 322–331. [Google Scholar] [CrossRef] [PubMed]
  203. Battaglia, R.; Delic, S.; Herrmann, H.; Snider, N.T. Vimentin on the move: New developments in cell migration. F1000Research 2018, 7, 1796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Berwick, D.C.; Heaton, G.R.; Azeggagh, S.; Harvey, K. LRRK2 Biology from structure to dysfunction: Research progresses, but the themes remain the same. Mol. Neurodegener. 2019, 14, 49. [Google Scholar] [CrossRef] [PubMed]
  205. Cao, H.; Alston, L.; Ruschman, J.; Hegele, R.A. Heterozygous CAV1 frameshift mutations (MIM 601047) in patients with atypical partial lipodystrophy and hypertriglyceridemia. Lipids Health Dis. 2008, 7, 3. [Google Scholar] [CrossRef] [PubMed]
  206. Jønsson, K.L.; Laustsen, A.; Krapp, C.; Skipper, K.A.; Thavachelvam, K.; Hotter, D.; Egedal, J.H.; Kjolby, M.; Mohammadi, P.; Prabakaran, T.; et al. IFI16 is required for DNA sensing in human macrophages by promoting production and function of cGAMP. Nat. Commun. 2017, 8, 14391. [Google Scholar] [CrossRef]
  207. Yan, Y.; Shi, R.; Yu, X.; Sun, C.; Zang, W.; Tian, H. Identification of atrial fibrillation-associated microRNAs in left and right atria of rheumatic mitral valve disease patients. Genes Genet. Syst. 2019, 94, 23–34. [Google Scholar] [CrossRef]
  208. Wang, S.; Min, J.; Yu, Y.; Yin, L.; Wang, Q.; Shen, H.; Yang, J.; Zhang, P.; Xiao, J.; Wang, Z. Differentially expressed miRNAs in circulating exosomes between atrial fibrillation and sinus rhythm. J. Thorac. Dis. 2019, 11, 4337–4348. [Google Scholar] [CrossRef]
  209. Yan, Y.; Song, D.; Zhang, X.; Hui, G.; Wang, J. GEO Data Sets Analysis Identifies COX-2 and Its Related Micro RNAs as Biomarkers for Non-Ischemic Heart Failure. Front. Pharmacol. 2020, 11, 1155. [Google Scholar] [CrossRef]
  210. Guo, X.; Wang, X.; Wang, Y.; Zhang, C.; Quan, X.; Zhang, Y.; Jia, S.; Ma, W.; Fan, Y.; Wang, C. Variants in the SMARCA4 gene was associated with coronary heart disease susceptibility in Chinese han population. Oncotarget 2017, 8, 7350–7356. [Google Scholar] [CrossRef]
  211. Matsha, T.E.; Kengne, A.P.; Hector, S.; Mbu, D.L.; Yako, Y.Y.; Erasmus, R.T. MicroRNA profiling and their pathways in South African individuals with prediabetes and newly diagnosed type 2 diabetes mellitus. Oncotarget 2018, 9, 30485–30498. [Google Scholar] [CrossRef]
  212. Tavano, F.; Fontana, A.; Mazza, T.; Gioffreda, D.; Biagini, T.; Palumbo, O.; Carella, M.; Andriulli, A. Early-Onset Diabetes as Risk Factor for Pancreatic Cancer: miRNA Expression Profiling in Plasma Uncovers a Role for miR-20b-5p, miR-29a, and miR-18a-5p in Diabetes of Recent Diagnosis. Front. Oncol. 2020, 10, 1567. [Google Scholar] [CrossRef]
  213. Ding, L.; Ai, D.; Wu, R.; Zhang, T.; Jing, L.; Lu, J.; Zhong, L. Identification of the differential expression of serum microRNA in type 2 diabetes. Biosci. Biotechnol. Biochem. 2016, 80, 461–465. [Google Scholar] [CrossRef] [Green Version]
  214. Erlich, H.A.; Valdes, A.M.; Julier, C.; Mirel, D.; Noble, J.A.; the Type I Diabetes Genetics Consortium. Evidence for association of the TCF7 locus with type I diabetes. Genes Immun. 2009, 10 (Suppl. 1), S54–S59. [Google Scholar] [CrossRef]
  215. Binia, A.; Van Stiphout, N.; Liang, L.; Michel, S.; Bhavsar, P.K.; Chung, K.F.; Brightling, C.E.; Barnes, P.J.; Kabesch, M.; Bush, A.; et al. A Polymorphism Affecting MYB Binding within the Promoter of the PDCD4 Gene is Associated with Severe Asthma in Children. Hum. Mutat. 2013, 34, 1131–1139. [Google Scholar] [CrossRef]
  216. Goldfine, A.B.; Crunkhorn, S.; Costello, M.; Gami, H.; Landaker, E.J.; Niinobe, M.; Yoshikawa, K.; Lo, D.; Warren, A.; Jimenez-Chillaron, J.; et al. Necdin and E2F4 Are Modulated by Rosiglitazone Therapy in Diabetic Human Adipose and Muscle Tissue. Diabetes 2006, 55, 640–650. [Google Scholar] [CrossRef]
  217. Rakshit, K.; Matveyenko, A.V. Induction of Core Circadian Clock Transcription Factor Bmal1 Enhances β-Cell Function and Protects Against Obesity-Induced Glucose Intolerance. Diabetes 2020, 70, 143–154. [Google Scholar] [CrossRef]
  218. Stratigopoulos, G.; LeDuc, C.A.; Cremona, M.L.; Chung, W.K.; Leibel, R.L. Cut-like Homeobox 1 (CUX1) Regulates Expression of the Fat Mass and Obesity-associated and Retinitis Pigmentosa GTPase Regulator-interacting Protein-1-like (RPGRIP1L) Genes and Coordinates Leptin Receptor Signaling. J. Biol. Chem. 2011, 286, 2155–2170. [Google Scholar] [CrossRef]
  219. Sedaghat, F.; Cheraghpour, M.; Hosseini, S.A.; Pourvali, K.; Teimoori-Toolabi, L.; Mehrtash, A.; Talaei, R.; Zand, H. Hypomethylation of NANOG promoter in colonic mucosal cells of obese patients: A possible role of NF-κB. Br. J. Nutr. 2019, 122, 499–508. [Google Scholar] [CrossRef]
  220. Patankar, J.V.; Chandak, P.G.; Obrowsky, S.; Pfeifer, T.; Diwoky, C.; Uellen, A.; Sattler, W.; Stollberger, R.; Hoefler, G.; Heinemann, A.; et al. Loss of intestinal GATA4 prevents diet-induced obesity and promotes insulin sensitivity in mice. Am. J. Physiol. Endocrinol. Metab. 2011, 300, E478–E488. [Google Scholar] [CrossRef]
  221. Drareni, K.; Ballaire, R.; Barilla, S.; Mathew, M.J.; Toubal, A.; Fan, R.; Liang, N.; Chollet, C.; Huang, Z.; Kondili, M.; et al. GPS2 Deficiency Triggers Maladaptive White Adipose Tissue Expansion in Obesity via HIF1A Activation. Cell Rep. 2018, 24, 2957–2971.e6. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Overview of data analysis methodology.
Figure 1. Overview of data analysis methodology.
Medicina 59 00309 g001
Figure 2. Volcano plot of differentially expressed genes. Genes with a significant change of more than two-fold were selected. Green dot represented up-regulated significant genes and red dot represented down-regulated significant genes.
Figure 2. Volcano plot of differentially expressed genes. Genes with a significant change of more than two-fold were selected. Green dot represented up-regulated significant genes and red dot represented down-regulated significant genes.
Medicina 59 00309 g002
Figure 3. Heat map of differentially expressed genes. Legend on the top left indicate log fold change ingenes. (A1–A120 = non-diabetic obese samples; B1–B104 = diabetic obese samples.)
Figure 3. Heat map of differentially expressed genes. Legend on the top left indicate log fold change ingenes. (A1–A120 = non-diabetic obese samples; B1–B104 = diabetic obese samples.)
Medicina 59 00309 g003
Figure 4. PPI network of DEGs. The PPI network of DEGs was constructed using Cytoscap. Up-regulated genes are marked in green; down-regulated genes are marked in red.
Figure 4. PPI network of DEGs. The PPI network of DEGs was constructed using Cytoscap. Up-regulated genes are marked in green; down-regulated genes are marked in red.
Medicina 59 00309 g004
Figure 5. Modules of isolated form PPI of DEGs. (A) The most significant module was obtained from PPI network with 16 nodes and 39 edges for up-regulated genes. (B) The most significant module was obtained from PPI network with 13 nodes and 24 edges for down-regulated genes. Up-regulated genes are marked in green; down-regulated genes are marked in red.
Figure 5. Modules of isolated form PPI of DEGs. (A) The most significant module was obtained from PPI network with 16 nodes and 39 edges for up-regulated genes. (B) The most significant module was obtained from PPI network with 13 nodes and 24 edges for down-regulated genes. Up-regulated genes are marked in green; down-regulated genes are marked in red.
Medicina 59 00309 g005
Figure 6. Target gene–miRNA regulatory network between target genes. The black-color diamond nodes represent the key miRNAs; up-regulated genes are marked in green; down-regulated genes are marked in orange.
Figure 6. Target gene–miRNA regulatory network between target genes. The black-color diamond nodes represent the key miRNAs; up-regulated genes are marked in green; down-regulated genes are marked in orange.
Medicina 59 00309 g006
Figure 7. Target gene–TF regulatory network between target genes. The gray-color triangle nodes represent the key TFs; up-regulated genes are marked in green; down-regulated genes are marked in red.
Figure 7. Target gene–TF regulatory network between target genes. The gray-color triangle nodes represent the key TFs; up-regulated genes are marked in green; down-regulated genes are marked in red.
Medicina 59 00309 g007
Figure 8. ROC curve analyses of hub genes. (A) MYH9 (B) FLNA (C) DCTN1 (D) CLTC (E) ERBB2 (F) TCF4 (G) VIM (H) LRRK2 (I) IFI16 (J) CAV1.
Figure 8. ROC curve analyses of hub genes. (A) MYH9 (B) FLNA (C) DCTN1 (D) CLTC (E) ERBB2 (F) TCF4 (G) VIM (H) LRRK2 (I) IFI16 (J) CAV1.
Medicina 59 00309 g008
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ganekal, P.; Vastrad, B.; Kavatagimath, S.; Vastrad, C.; Kotrashetti, S. Bioinformatics and Next-Generation Data Analysis for Identification of Genes and Molecular Pathways Involved in Subjects with Diabetes and Obesity. Medicina 2023, 59, 309. https://doi.org/10.3390/medicina59020309

AMA Style

Ganekal P, Vastrad B, Kavatagimath S, Vastrad C, Kotrashetti S. Bioinformatics and Next-Generation Data Analysis for Identification of Genes and Molecular Pathways Involved in Subjects with Diabetes and Obesity. Medicina. 2023; 59(2):309. https://doi.org/10.3390/medicina59020309

Chicago/Turabian Style

Ganekal, Prashanth, Basavaraj Vastrad, Satish Kavatagimath, Chanabasayya Vastrad, and Shivakumar Kotrashetti. 2023. "Bioinformatics and Next-Generation Data Analysis for Identification of Genes and Molecular Pathways Involved in Subjects with Diabetes and Obesity" Medicina 59, no. 2: 309. https://doi.org/10.3390/medicina59020309

Article Metrics

Back to TopTop