Abstract Background: High-throughput data generation is developing in the cancer area and offers a better opportunity of understanding molecular pathways involved in the progression of tumors. Meta-analysis of gene expression based on data integration makes it possible to determine changes in gene expression with more accuracy. This approach and downstream analysis were utilized for colorectal cancer to identify promising biomarkers and drug targets. Materials and Methods: First, a systematic search was performed in the Gene Expression Omnibus (GEO) database. Meta-analysis was used to obtain differentially expressed (DE) genes from the NetworkAnalyst database. Moreover, microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and transcription factors (TFs) enriched with DE genes were determined by the Enrichr database. The integrated DE genes-miRNAs-lncRNAs-TFs network was constructed and analyzed using Cytoscape software. Then, the downstream analyses of hub genes were performed. Results: The primary candidate genes, HNF4A, FLI1, MITF, E2F4, SPI1, FOXA2, VDR, and SMAD3, which are overlapped nodes with the highest degree, betweenness, and closeness centrality parameters in the top 30 nodes were selected. Also, the hsa-miR-26b-5p, hsa-miR-16-5p, hsa-miR-124-3p, and hsa-miR-92a-3p were introduced as key non-coding RNAs with the highest degree and betweenness centralities. For the determination of the clinical significance of hub genes, the mutation, differential expression, and survival analysis were assayed. Also, the drugs related to candidate hub genes were determined. The functional analysis revealed pathways significantly related to cancer progression. Conclusion: Employing systems biology approaches with holistic insight can identify essential genes and their regulation as possibilities for further experimental testing. Keywords: Gene regulatory network, meta-analysis, microarray, protein interaction network INTRODUCTION Colorectal cancer (CRC) is the third most commonly diagnosed cancer worldwide and the second and fourth leading cause of cancer death for men and women, respectively.[[30]1] Consequently, it signifies a serious health issue, and early detection is imperative to facilitate prompt medical intervention. Approximately 25% of CRCs are diagnosed at the metastatic stage when treatment is less likely to be effective, and survival is low.[[31]2] Pathologically, molecular biomarkers, along with the Tumor, Nodes, Metastases (TNM) staging system and histological features, are critical in diagnosing and treating CRC.[[32]3] In particular, KRAS, BRAF, and microsatellite instability (MSI) molecular markers are currently considered CRC prognostic factors by the American Joint Committee on Cancer (AJCC).[[33]4] A significant increase in overall survival in patients with cancer has been documented over the past 20 years. This is attributed to identifying novel molecular pathways and using biological agent targeting for diagnosis and treatment.[[34]5] New biomarkers can be uncovered using traditional molecular biology techniques. Combining distinct data sources from different approaches provides less biased and more systematic discovery of biomarkers in clinical contexts. Bioinformatics and computational research advances have paved the way to data processing and understanding biomarker discovery. Applied bioinformatics research leads attempts to disseminate biomarker study results and develops advanced infrastructures empowering researchers to go beyond “single-marker” analysis.[[35]6] High-throughput data analysis approaches provide a robust platform for analyzing biological pathogenesis.[[36]7] The survey of protein interaction maps provides the best picture of how each protein contributed to the molecular function. As a result of the rapid development of microarray technology, there is an abundance of microarray data in the cancer genetics field. There is no doubt that meta-analysis and other tools based on systems biology can be relied on for a more precise assessment of tumorigenic processes and an accurate choice of treatment targets. Our study aims to identify the key genes obtained from the meta-analysis; then, integrated network survey with microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and transcription factors (TFs) were enriched. Moreover, gene set enrichment analysis identifies the main signaling pathways involved in CRC. Then, the rate of mutation, the expression range for hub genes across tissues in normal and tumor, and also survival analysis of hub genes were identified. Drugs related to hub genes are also determined for the identification of proper treatment options. The results of this study may provide clues to suggest proper candidate biomarkers for CRC. MATERIALS AND METHODS Microarray analysis and meta-analysis Datasets of expression profiling by array were searched in the Gene Expression Omnibus (GEO) database.[[37]8] The keywords “colorectal cancer” and “colon cancer” were used in a systematic search in homo sapiens samples. Datasets that criteria included; less than three samples in each group, cell line studies, datasets with no healthy control samples, treated, and unrelated to the study hypothesis were ignored. We focused on datasets with Affymetrix and Illumina platforms. Due to the importance of evaluating the quality control of the datasets in the first step,[[38]9] the quality control was identified by principal component analysis (PCA) with the ggplot2 package[[39]10] of the prcomp function of R software.[[40]11] The eight microarray datasets were selected for further analysis, including [41]GSE110224, [42]GSE23878, [43]GSE37364, [44]GSE110223, [45]GSE77953, [46]GSE54986, [47]GSE74602, and [48]GSE25070. The NetworkAnalyst V3.0 database was used for meta-analysis.[[49]12] After normalization by log2 transformation and batch effect, the meta-analysis was performed by the random effect model (REM) approach. A significance level of adjusted P value less than 0.05 was considered for determining significant DE genes. Ethics approval and consent to participate: IR.MUI.MED.REC.1399.558. The miRNAs, lncRNAs, and transcription factors (TFs) prediction Enrichr database[[50]13] was utilized for investigation at other levels. The mRNA-miRNA interaction was determined using the miRTarBase library.[[51]14] At the transcriptional level, using the ChEA library identified regulatory TFs.[[52]15] LncHUB library provided predictions of the lncRNAs correlated with DE genes. The adj. P value < 0.05 was considered for significant results. Network construction and analysis After miRNAs, lncRNAs, and TFs interactions with DE genes were determined, the gene regulatory networks (GRNs) were created using Cytoscape software V7.2.0.[[53]16] The protein-protein interaction (PPI) network was made via the STRING database[[54]17] in the CluePedia application.[[55]18] The interactions were retrieved based on activation, inhibition, binding, and post-translational modification with a confidence cut-off 0.8. The Merge tool of Cytoscape software was used to create an integrated network from four networks. Using the Network Analyzer tool, we measured the centrality parameters based on treating the network as directed to ranked hub nodes in the merged network. Clustering and module mining were performed by the MCODE plug-in (version 1.6.1),[[56]19] a Cytoscape plug-in, to identify sub-networks that have a significant function with degree cut-off = 2, node score cut-off = 0.2, k-core = 2, and max. depth = 100. Survival analysis of candidate hub genes For the clinical significance assay, the survival evaluation of selected candidate hub genes in the Kaplan-Meier (KM) plotter online database (kmplot.com) was performed to estimate the effect of genes on survival. This correlation analysis utilizes the data from 30,000 samples of 21 tumor types obtained from gene arrays, RNA sequence, or next-generation sequencing.[[57]20] In this study, relapse-free survival (RFS) data were achieved from colon cancer. Mutation and expression analysis of candidate hub genes The information on somatic mutation and nucleotide changes of selected candidate hub genes were determined. Data are generated by the Catalogue of Somatic Mutations in Cancer (COSMIC),[[58]21] The International Cancer Genome Consortium (ICGC),[[59]22] and The Cancer Genome Atlas (TCGA) ([60]www.cancer.gov/tcga). Then, the comparison of gene expression in normal, tumor, and metastatic tissues based on the integrated database using available transcriptome-level datasets, such as NCBI’s Gene Expression Omnibus (GEO), TCGA, Therapeutically Applicable Research to Generate Effective Treatments (TARGET), and The Genotype-Tissue Expression (GTEx), was obtained.[[61]23] Significant differences by a Mann-Whitney U-test are marked with red color. Pathway and functional analysis of candidate hub genes, genes clustered in modules, miRNAs, and lncRNAs Functional analysis is conducted to perceive the main molecular mechanisms in the pathogenesis of cancer. The ClueGo plug-in (version 2.5.4)[[62]24] was employed based on the Kyoto Encyclopedia of Genes and Genomes (KEGG)[[63]25] and Reactome pathway database.[[64]26] P value < 0.05 corrected with Bonferroni’s step-down is considered for significant results. Using DIANA-miRPath v3.0[[65]27] an online software, the miRNA function was analyzed. The enrichment analysis of lncRNAs was performed by LncSEA, a platform for lncRNA.[[66]28] For significant results, adj. P value < 0.05 was considered a cut-off. The framework of the steps mentioned is depicted in [67]Figure 1. Figure 1. Figure 1 [68]Open in a new tab Framework designed and flowed in this study is represented RESULTS Analysis of the significant differentially expressed genes The [69]GSE110224, [70]GSE23878, [71]GSE37364, [72]GSE110223, [73]GSE77953, [74]GSE54986, [75]GSE74602, and [76]GSE25070 datasets selected for meta-analysis are a total number of 308 samples, which included 151 control and 157 cancer samples [[77]Table 1]. The quality check result shows the acceptable samples in each group [[78]Supplementary Figure 1^ (1.2MB, tif) ]. The 5245 significant differentially expressed (DE) genes were recognized using meta-analysis based on the REM method. A total of 2727 DE genes with combined effect size (ES) >|1|, which included 1444 up-regulated and 1283 down-regulated genes, were selected for further analysis [[79]Supplementary Table 1^ (320KB, pdf) shows DE genes with P value-corrected and combined ES]. Table 1. Studies retrieved from the GEO database for meta-analysis are described Serial Sample (tumor/ctrl) DEGs Platform [80]GSE110224 n=26 (13/13) 8395 Affymetrix Human Genome U133 Plus 2.0 Array [81]GSE23878 n=55 (31/24) 6510 Affymetrix Human Genome U133 Plus 2.0 Array [82]GSE37364 n=63 (25/38) 11116 Affymetrix Human Genome U133 Plus 2.0 Array [83]GSE110223 n=25 (13/12) 5591 Affymetrix Human Genome U133A Array [84]GSE77953 n=24 (17/7) 7867 Affymetrix Human Genome U133A Array [85]GSE54986 n=11 (5/6) 2187 Illumina HumanHT-12 V4.0 expression beadchip [86]GSE74602 n=55 (29/26) 10593 Illumina HumanRef-8 v2.0 expression beadchip [87]GSE25070 n=49 (24/25) 5330 Illumina HumanRef-8 v3.0 expression beadchip [88]Open in a new tab Prediction of miRNAs, lncRNAs, and transcription factors that regulate the target genes We identified miRNAs, lncRNAs, and transcription factors -targeted DEGs to reveal the regulatory of DEGs at transcriptional and post-transcriptional levels and construction of GRN. The analysis revealed 175 miRNAs, 187 lncRNAs, and 41 TFs significantly enriched with DE genes [Supplementary Tables [89]2^ (246.7KB, pdf) -[90]4^ (178.8KB, pdf) show the non-coding RNAs and TFs with related genes and adjusted P value]. The hsa-miR-26b-5p (adj. P value < 0.0001), hsa-miR-16-5p (adj. P value < 0.0001), hsa-miR-124-3p (adj. P value < 0.0001), hsa-miR-92a-3p (adj. P value < 0.0001), hsa-let-7b-5p (adj. P value < 0.0001), H2AZ1-DT (adj. P value < 0.0001), PRC1-AS1 (adj. P value < 0.0001), TMPO-AS1 (adj. P value < 0.0001), DEPDC1-AS1 (adj. P value < 0.0001), and RRM1-AS1 (adj. P value < 0.0001) are non-coding RNAs having highest connection with genes. HNF4A (adj. P value < 0.0001), FLI1 (adj. P value < 0.01), MITF (adj. P value = 0.0336), FOXP1 (adj. P value = 0.0325), SPI1 (adj. P value = 0.0006), E2F4 (adj. P value < 0.0001), and FOXA2 (adj. P value < 0.0001) are the TFs enriched with high connectivity. Integrated network construction and analysis The characteristics of nodes in the network were investigated to identify their significance. In the first step, GRNs were created from the miRNA-mRNA, lncRNA-mRNA, and TF-mRNA interaction network with 2311, 1512, and 2435 nodes, respectively. The protein interaction network with 1177 interconnected nodes was created. Then, a merged network with 3063 nodes and 41328 edges was constructed. The topological network analysis revealed hub nodes that 30 (1%) of them and their interaction are represented in [91]Figure 2. HNF4A, FLI1, MITF, E2F4, SPI1, FOXA2, VDR, and SMAD3 are the central nodes with overlapped centrality parameters. [92]Table 2 also shows the 10 nodes in each class with the highest value. Sub-network and module analysis showed 35 clusters, of which six key clusters were selected based on functional analysis. Modules c, d, e, and f have interaction with non-coding RNAs. Clusters are the dense interaction sub-networks that could have crucial functions in the network. SMNDC1, PSMD5, ANAPC1, INO80B, and GNAO1 are seed genes in clusters a-e, respectively. Cluster f is the lack of seed genes [[93]Figure 3]. Figure 2. Figure 2 [94]Open in a new tab Network with hub nodes. Nodes with the highest degree, betweenness, or closeness centrality are considered the hub genes Table 2. Hub nodes. The 10 top nodes with the highest index in each group in the gene regulatory network are shown. Nodes with the highest degree that is the number of the total number of edges connected to a node, betweenness centrality is used to determine the nodes that are in the shortest paths, nodes that bridge from one part of a graph to another, and closeness centrality is used to identify the nodes that rapidly interact with other nodes in the network. These nodes are considered the hub genes Term Degree Betweenness centrality Closeness centrality Targeted genes FOXM1 227 FOXM1 0.01 PLK1 0.47 UBA52 169 NR1H3 0.01 AURKB 0.47 NR1H3 152 GNG12 0.01 HSP90AA1 0.47 PLK1 129 − − TNRC6B 0.47 NCBP2 125 − − NDE1 0.46 POLR2D 123 − − CCND1 0.46 RNPS1 121 − − PTBP1 0.46 ESR1 119 − − GSK3B 0.46 HSPA8 117 − − RANGAP1 0.46 POLR2F 113 − − WASL 0.46 TFs HNF4A 979 HNF4A 0.12 FLI1 0.53 FLI1 871 FLI1 0.11 HNF4A 0.51 MITF 820 MITF 0.08 SMAD3 0.49 E2F4 587 E2F4 0.04 MITF 0.48 FOXP1 560 FOXP1 0.04 VDR 0.48 SPI1 509 SPI1 0.04 E2F4 0.47 FOXA2 492 FOXA2 0.03 SPI1 0.46 GABP 427 SOX2 0.03 FOXA2 0.46 SOX2 425 GABP 0.02 FOXP1 0.45 VDR 403 VDR 0.02 TTF2 0.45 miRNAs hsa-miR-26b-5p 404 hsa-miR-26b-5p 0.03 hsa-miR-124-3p 0.44 hsa-miR-16-5p 337 hsa-miR-124-3p 0.02 hsa-miR-155-5p 0.43 hsa-miR-124-3p 296 hsa-miR-16-5p 0.01 hsa-miR-26b-5p 0.42 hsa-miR-92a-3p 281 hsa-miR-92a-3p 0.01 hsa-miR-16-5p 0.42 hsa-let-7b-5p 229 hsa-let-7b-5p 0.01 hsa-miR-92a-3p 0.4 hsa-miR-484 219 hsa-miR-484 0.01 hsa-miR-193b-3p 0.4 hsa-miR-193b-3p 215 hsa-miR-193b-3p 0.01 hsa-miR-17-5p 0.4 hsa-miR-93-5p 209 hsa-miR-192-5p 0.01 hsa-let-7b-5p 0.39 hsa-miR-17-5p 205 hsa-miR-155-5p 0.01 hsa-miR-484 0.39 hsa-miR-192-5p 195 hsa-miR-1-3p 0.01 hsa-miR-192-5p 0.39 LncRNAs H2AZ1-DT 65 − − PRC1-AS1 0.36 PRC1-AS1 58 − − TMPO-AS1 0.36 TMPO-AS1 54 − − DDX11-AS1 0.36 DEPDC1-AS1 53 − − PRMT5-AS1 0.36 RRM1-AS1 53 − − LINC01775 0.36 CSRP3-AS1 52 − − DIAPH 3-AS1 0.36 HMMR-AS1 52 − − LINC01224 0.36 DDX11-AS1 51 − − GTF3C2-AS1 0.36 PRMT5-AS1 50 − − LINC01842 0.36 LINC01357 48 − − ODF2-AS1 0.36 [95]Open in a new tab Figure 3. Figure 3 [96]Open in a new tab Prominent modules with cluster mining. The (a, b, c, d, e, and f) are the selected modules, (clusters 1, 3, 8, 10, 11, and 15 from MCODE analysis results). To classify the sub-networks, densely connected region in the network with significant functions, the default setting was considered. The degree cut-off = 2, node score cut-off = 0.2, k-core = 2, and max. depth = 100 Survival analysis of candidate hub genes The correlation of selected hub genes with survival times was identified. The correlation analysis between the target gene expression level and colon cancer survival rate was conducted. For HNF4A, upper quartile survival is 19 and 60 months for low and high expression in cohort samples, respectively. Also, low and high expression of FLI1 and MITF correlated with approximately 45 and 18 months and 56 and 19 months, respectively. The results of survival analysis of E2F4, SPI1, FOXA2, VDR, and SMAD3 are shown in [97]Figure 4. Also, from hub miRNAs, the low and high expression hsa-miR-484 is correlated with changes in survival times in 21 tumor types. Figure 4. Figure 4 [98]Open in a new tab Results of survival analysis are shown in the Kaplan-Meier survival plots. The parameters hazard ratio (HR) and 95% confidence are represented as the reliable range. The P value less than 0.05 indicates a significant results Mutation and expression analysis of candidate hub genes The analysis of mutation distribution of HNF4A, FLI1, MITF, E2F4, SPI1, FOXA2, VDR, and SMAD3 based on COSMIC, ICGC, and TCGA databases was conducted. Cancer mutation distribution of HNF4A is 5342 mutations, 179 high-impact mutations, 269 mutations, and 3,760 copy number variation (CNV) based on COSMIC, ICGC, and TCGA databases, respectively. The results of other hub genes are shown in [99]Table 3. Based on the ICGC database, only the VDR gene has targeting compounds. The dn-101 (ZINC000100015048) and Dovonex (ZINC000003921872) are the FDA-approved components. The level of gene expression of each gene in several types of cancer was obtained. The results are shown in [100]Figure 5. Table 3. Results of mutation distribution of selected hub genes are based on data collected from the COSMIC, ICGC, and TCGA databases Genes COSMIC ICGC TCGA HNF4A 5342 mutations 179 high-impact mutations 252 cases affected by 269 mutations across 35 projects 3,886 cases affected by 3,760 CNV events across 33 projects FLI1 4563 mutations 17 high-impact mutations 196 cases affected by 194 mutations across 31 projects 3,401 cases affected by 3,300 CNV events across 33 projects MITF 10611 mutations 37 high-impact mutations 172 cases affected by 176 mutations across 28 projects 3,585 cases affected by 3,493 CNV events across 33 projects E2F4 197 mutations 72 high-impact mutations 68 cases affected by 69 mutations across 25 projects 3,671 cases affected by 3,630 CNV events across 33 projects SPI1 1047 mutations 8 high-impact mutations 80 cases affected by 74 mutations across 26 projects 2,609 cases affected by 2,561 CNV events across 33 projects FOXA2 637 mutations 93 high-impact mutations 150 cases affected by 145 mutations across 31 projects 3,752 cases affected by 3,684 CNV events across 33 projects VDR 2067 mutations 112 high-impact mutations 107 cases affected by 102 mutations across 25 projects 2,683 cases affected by 2,623 CNV events across 33 projects SMAD3 2990 mutations 135 high-impact mutations 160 cases affected by 155 mutations across 25 projects 2,769 cases affected by 2,681 CNV events across 33projects [101]Open in a new tab Figure 5. Figure 5 [102]Open in a new tab DE of selected hub genes in the most common tumor types is presented Downstream functional analysis The downstream functional analysis determines the relation of selected hub genes with significant biological states. Pathway enrichment analysis results of the top hub genes shown are related to cell cycle, oncogene-induced senescence, signaling by transforming growth factor-beta (TGF-β) receptor complex, signaling by FGFR2, signaling by PTK6, and transcriptional regulation by RUNX1-3. The enrichment analysis of genes in modules was conducted. Terms were summarized based on the repeated terms; the results are shown in [103]Figure 6. The miRNA functional analysis revealed the hub miRNAs strongly related to cancer and especially enriched with proteoglycans in cancer, pathways in cancer, focal adhesion, signaling pathways regulating pluripotency of stem cells, gap junction, fatty acid metabolism, and regulation of actin cytoskeleton. The hub lncRNA enrichment analysis discovered was correlated with cancer phenotypes, such as breast carcinoma, lung adenocarcinoma, esophageal carcinoma, stomach carcinoma, and colon carcinoma. Figure 6. Figure 6 [104]Open in a new tab Pathway enrichment analysis from each of the selected modules based on KEGG and Reactome pathway database, respectively (1: a; 2: b; 3: c; 4: d; 5: e; 6: f). The results are depicted based on significant results. Signaling pathways with an adjusted P value < 0.05 were selected. For P value correction, Bonferroni’s step-down was considered. The number of involved genes and the percentage of association genes related in each term are shown DISCUSSION In this study, we identified 2727 DE genes obtained with a meta-analysis of CRC from the microarray datasets with 308 samples. Genes were enriched with miRNAs, lncRNAs, and TFs at transcriptional and post-transcription levels. The integrated network was created by merging the protein interaction network and GRNs, including miRNA-mRNA, lncRNA-mRNA, and TF-mRNA interaction. According to the merged network analysis with 3063 nodes, the hub nodes, including all of the parameters measured, are HNF4A, FLI1, MITF, E2F4, SPI1, FOXA2, VDR, and SMAD3. A high degree indicates they have many connections and are vital for the surveillance of the network. Betweenness centrality measures the number of shortest paths going through a node. Closeness centrality is the physically nearest node to all nodes.[[105]29] As mentioned, the hub genes are frequently mutated in several types of cancers and correlated with DE genes in different types of cancers, especially in CRC. Also, these genes are related to RFS time in patients with colon cancer. There was a significant relationship between these hub genes with cell cycle, oncogene-induced senescence, and TGF-β receptor complex signaling based on functional analysis. TFs particularly had higher centrality scores. Also, the top main miRNAs named miR-26b-5p, miR-16-5p, miR-124-3p, miR-92a-3p, let-7b-5p, miR-484, miR-193b-3p, and miR-192-5p. The miR-26b-5p as a tumor-suppressive miRNA was known in various cancer tissues[[106]30] and was connected with 404 genes in this study. The miR-16-5p is known as a tumor suppressor by inhibiting proliferation and invasion[[107]31] and controlling 337 genes in the network. The miR-124-3p acts as an inhibitor of tumor growth and is down-regulated in multiple human neoplasms[[108]32] and here linked to 296 genes. miR-92a-3p correlated with 281 nodes is involved in several tumorigenic processes, including cell proliferation, survival, and invasion.[[109]33] Other non-coding RNAs, lncRNA, were screened, including PRC1-AS1, TMPO-AS1, DDX11-AS1, and PRMT5-AS1. In the GeneCards database reference, PRC1-AS1 is related to hepatocellular carcinoma. TMPO-AS1 contributes significantly to CRC progression.[[110]34] DDX11-AS1 has potential roles in proliferation and invasion in CRC cells linked to 51 genes.[[111]35] FOXM1, NR1H3, and PLK1 could be pointed out as key genes. FOXM1 was shown to be required for malignancy of CRC[[112]36] correlated with 227 nodes. NR1H3 is a regulatory lipid metabolism and has an important role in cancer progression[[113]37] and is connected with 152 nodes. PLK1, linked with 129 nodes, acts as a progression factor of invasion in CRC and many human cancers.[[114]38] Results of previous experimental studies demonstrate the accuracy of explored approaches and emphasize the significance of their roles in CRC progression. Modules are high-density regions in the network that detect functional genes. The critical module assessment was shown to be closely linked to cancer development. The first cluster-related signaling is Rho GTPases, metabolism of RNA, signaling by FGFR2, interferon signaling, and cell cycle. Rho GTPases play a vital role in forming dynamic actin-rich membrane protrusions and cell-cell turnover and cell-extracellular matrix (ECM) adhesions required for efficient cancer cell invasion.[[115]39] It has been reported that Rho GTPases could regulate the expression of Wnt target genes in colon cancer cells. Moreover, the knockdown of Rac1, as a member of Rho GTPases, could suppress the expression of Wnt-induced genes NKD1, BAMBI, and MMP-7, which are involved in invasion and metastasis.[[116]40] The second module is correlated to the regulation of apoptosis, stabilization of p53, signaling by NOTCH4, signaling by Wnt, MAPK signaling cascades, signaling by Hedgehog, cellular response to hypoxia, and regulation of PTEN stability and activity. The Wnt/β-catenin signaling pathway is a developmentally preserved and one-of-a-kind signaling pathway that regulates gene expression and cell invasion, migration, proliferation, and differentiation within the start and progression of CRC. Wnt signaling cascades and at least one protein of the Wnt/β-catenin signaling pathway are mutated in more than 94% of CRC cases. The development of these mutations is thought to be an early occasion and the essential driving force in early-stage CRC.[[117]41] ECM-receptor interaction, cell cycle, and MET-promoted cell motility were enriched with hubs of the third cluster. The ECM is a noncellular component of tissue that biochemically and structurally supports cells. The ECM comprises different glycoproteins, such as collagens, laminins, and fibronectins. Interactions of the ECM and cellular receptors constitute one of the crucial pathways in CRC progression and metastasis.[[118]42] Smooth muscle contraction, PPAR signaling pathway, leukocyte transendothelial migration, NF-κB signaling pathway, and signaling by erythropoietin are the main terms linked with genes of the fourth cluster. NF-κB activation supports tumorigenesis by enhancing cell proliferation and angiogenesis, inhibiting apoptosis, and promoting cell attack and metastasis. It plays a central part in regulating cell death in the development and progression of CRC. The antiapoptotic activity of NF-κB is mediated through Bcl2, Bcl-xL, cFLIP, and cIAP2, among other genes. Antiapoptotic protein BAG-1 (Bcl-2-associated athanogene-1) is included in crucial processes, such as proliferation, cell signaling, transcription, and apoptosis. Overexpression of BAG-1 has been reported in colorectal adenomas and carcinomas.[[119]43] Interaction between L1 and ankyrins, Eph-ephrin signaling, circadian clock, and Ca2+ pathway are the critical pathways related to the fifth module. The physiologic function of the colon is to treat and assimilate undigested nourishment, electrolytes, and water—the colon exchanges Na+, K+, and Ca2+ to preserve electrolyte adjustment. Appropriately, ion channels are commonly communicated on the colonic epithelial cell film. Within the case of colorectal colitis inflammation and electrolyte unsettling influence, the event of colon cancer may be encouraged. Colonic inflammation could be a risk factor for the development of CRC. Chronic intestinal inflammatory diseases regularly lead to CRC through a handle called colitis-associated carcinogenesis. As a result, maintaining adjust of electrolytes within the environment of colon epithelial cells is vital to avoid CRC. Among those electrolytes, Ca2+ is the foremost high-profile ion in CRC progression, and its impact on cancer development is controversial.[[120]44] Finally, the sixth sub-network is associated with mTOR signaling, collagen formation, TP53-regulated metabolic genes, and macroautophagy. mTOR signaling pathway plays a significant part in cancer development, counting proliferation, metastasis, survival, and angiogenesis. The CRC pathogenesis is exceptionally complex and expects the presence of numerous genetic mutations that will be included in cancer progression. Besides, CRC progression is affected by inflammatory cells and their inflammatory mediators, such as cytokines, that can enact signaling pathways, leading to tumoral development.[[121]45] Each node in each module, particularly seed genes, has the potential to select as a candidate for experimental validation. CONCLUSION Here, using computational tools we generate a systematic view of the CRC disorder to highlight the central genes, miRNAs, lncRNAs, and TFs as effective target therapies. With the determination of key pathways, we explored the comprehensive molecular mechanisms involved in the progression of this disorder. Conflicts of interest There are no conflicts of interest. Supplementary table-1 Differentially expressed genes obtained with meta- analysis. [122]ABR-14-99_Suppl1.pdf^ (320KB, pdf) Supplementary table-2 micro-RNAs Enriched with DEGs. [123]ABR-14-99_Suppl2.pdf^ (246.7KB, pdf) Supplementary table-3 Long- non coding RNAs Enriched with DEGs. [124]ABR-14-99_Suppl3.pdf^ (155.1KB, pdf) Supplementary table-4 Transcription factors enriched with DEGs. [125]ABR-14-99_Suppl4.pdf^ (178.8KB, pdf) Supplementary Figure 1 The quality control assessment were performed. Some of samples were omitted due to inappropriate quality, including: ([126]GSM2982932, GSM29829324, [127]GSM2982935, [128]GSM2982936, [129]GSM2982937, [130]GSM2982939, [131]GSM2982941, and [132]GSM2982956) in [133]GSE110224, ([134]GSM588834, [135]GSM588857, [136]GSM588858, and [137]GSM588860) in [138]GSE23878, ([139]GSM916734 and [140]GSM916721) in [141]GSE37364, ([142]GSM292928) in [143]GSE110223, ([144]GSM2062358, [145]GSM2062359, [146]GSM2062360, [147]GSM2062361, [148]GSM2062362, and [149]GSM2062363) in [150]GSE77953, ([151]GSM1327497) in [152]GSE54986, (GSM192368, GSM192369, [153]GSM192370, GSM192371, and GSM192377) in [154]GSE74602, and ([155]GSM615880, [156]GSM615882, and [157]GSM615895) in [158]GSE25070. Last PCA plot were obtained from NetworkAnalyst database based on merged data [159]ABR-14-99_Suppl1.tif^ (1.2MB, tif) Acknowledgment