Skip to main content
medRxiv
  • Home
  • About
  • Submit
  • ALERTS / RSS
Advanced Search

Identification and Interaction Analysis of Molecular Markers in Pancreatic Ductal Adenocarcinoma by Integrated Bioinformatics Analysis and Molecular Docking Experiments

View ORCID ProfileBasavaraj Vastrad, View ORCID ProfileChanabasayya Vastrad, View ORCID ProfileAnandkumar Tengli
doi: https://doi.org/10.1101/2020.12.20.20248601
Basavaraj Vastrad
1Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Basavaraj Vastrad
Chanabasayya Vastrad
2Biostatistics and Bioinformatics,Chanabasava Nilaya, Bharthinagar, Dharwad, Karanataka 580001, India
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Chanabasayya Vastrad
  • For correspondence: channu.vastrad{at}gmail.com
Anandkumar Tengli
3Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru-570015, Karnataka, India
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Anandkumar Tengli
  • Abstract
  • Full Text
  • Info/History
  • Metrics
  • Data/Code
  • Preview PDF
Loading

Abstract

The current investigation aimed to mine therapeutic molecular targets that play an key part in the advancement of pancreatic ductal adenocarcinoma (PDAC). The expression profiling by high throughput sequencing dataset profile GSE133684 dataset was downloaded from the Gene Expression Omnibus (GEO) database. Limma package of R was used to identify differentially expressed genes (DEGs). Functional enrichment analysis of DEGs were performed. Protein-protein interaction (PPI) networks of the DEGs were constructed. An integrated gene regulatory network was built including DEGs, microRNAs (miRNAs), and transcription factors. Furthermore, consistent hub genes were further validated. Molecular docking experiment was conducted. A total of 463 DEGs (232 up regulated and 231 down regulated genes) were identified between very early PDAC and normal control samples. The results of Functional enrichment analysis revealed that the DEGs were significantly enriched in vesicle organization, secretory vesicle, protein dimerization activity, lymphocyte activation, cell surface, transferase activity, transferring phosphorus-containing groups, hemostasis and adaptive immune system. The PPI network and gene regulatory network of up regulated genes and down regulated genes were established, and hub genes were identified. The expression of hub genes (CCNB1, FHL2, HLA-DPA1 and TUBB1) were also validated to be differentially expressed among PDAC and normal control samples. Molecular docking experiment predicted the novel inhibitory molecules for CCNB1 and FHL2. The identification of hub genes in PDAC enhances our understanding of the molecular mechanisms underlying the progression of this disease. These genes may be potential diagnostic biomarkers and/or therapeutic molecular targets in patients with PDAC.

Introduction

Pancreatic ductal adenocarcinoma (PDAC) is one of the most prevalent cancers in the world and primary tumor of the pancreas [1]. PDAC is a global burden ranking 15th in terms of incidence and fourth in terms of mortality [2]. Despite new developments in multimodal therapy its overall 5-year survival rate remains less than 8% [3]. PDAC treatment commonly includes surgery, radiation, chemotherapy and immunotherapy [4]. However, PDAC remains common and malignant due to recurrence and metastasis, and ultimately key cause of PDAC associated death [5]. Therefore, there is a vital need to advance new diagnostic strategies and therapeutic agents to upgrade the prognosis of patients with PDAC.

The molecular mechanisms of PDAC tumorigenesis and development remain imprecise. It is therefore key to identify novel genes and pathways that are linked with PDAC tumorigenesis and patient prognosis, which may not only help to illuminate the underlying molecular mechanisms involved, but also to disclose novel diagnostic markers and therapeutic targets. Oji et al [6] demonstrated that the over expression of WT1 is linked with prognosis in patients with PDAC. A previous investigation reported that phosphoinositide 3-kinase signaling pathway is linked with development of PDAC [7]. Expression profiling by high throughput sequencing can promptly uncover gene expression on a global basis and are specially useful in identifying for differentially expressed genes (DEGs) [8]. A huge amount of data has been generated through the use of microarrays and the majority of such data has been deposited and saved in public databases. Previous investigations concerning PDAC gene expression profiling have diagnosed hundreds of DEGs [9].

The aim of this investigation was to identify hub genes and pathways in PDAC using bioinformatics methods. Our investigation contributes predictable biomarkers for early detection and prognosis, as well as effective drug targets for treating PDAC.

Materials and methods

Sequencing data

PDAC expression profiling by high throughput sequencing dataset in this investigation was downloaded from the GEO database (https://www.ncbi.nlm.nih.gov/gds/) [10]. The DEGs were considered by 1 independent PDAC dataset, GSE133684 [11] with 284 PDAC and 117 normal samples. The GSE133684 expression profiling by high throughput sequencing data was based on the GPL20795 HiSeq X Ten (Homo sapiens) platform..

Identification of DEGs

The Limma package of R language were used to normalize and convert the raw data to expression profiles [12]. The limma package of R language was used for DEGs between PDAC and normal control samples [12]. The P-value was adjusted by the Benjamini-Hochberg method [13]. An adjusted P-value <0.05 and |log2 fold change (FC) | >1 were considered as threshold values for DEGs identification. The ggplot2 package and gplots package of R language was used to generate volcano plot and heat map. The identified DEGs were preserved for further bioinformatics analysis.

GO analysis and pathway enrichment analysis of DEGs

The GO repository (http://geneontology.org/) [14] consists of a massive set of annotation terms and is generally used for annotating genes and identifying the distinctive biological aspects for expression profiling by high throughput sequencing data. The REACTOME database (https://reactome.org/) [15] contains data on known genes and their biochemical functions and is used for identifying functional and metabolic pathways. By performing the GO and REACTOME enrichment analysis at the functional level, we can boost a better understanding of the roles of these DEGs in the induction and in the advancement of PDAC. The ToppGene (ToppFun) (https://toppgene.cchmc.org/enrichment.jsp) [16] is an online resource that add tools for functional annotation and bioinformatics analysis. Both GO categories and REACTOME pathway enrichment analysis were implemented using ToppGene to inform the functions of these DEGs. P<0.05 was considered to indicate a statistically significant difference.

Protein-protein interaction (PPI) network construction and module analysis

The online database IID interactome (http://iid.ophid.utoronto.ca//)[17] was used to construct a PPI network of the proteins encoded by DEGs. Then, Cytoscape software (Version 3.8.1) [18] was applied to perform protein interaction association network analysis and analyze the interaction correlation of the candidate proteins encoded by the DEGs in PDAC. Next, the Network Analyzer Cytoscape plug-in was applied to calculate node degree [19], betweenness centrality [20], stress centrality [21], closeness centrality [22]. Finally, the PEWCC1 (http://apps.cytoscape.org/apps/PEWCC1) [23] module for Cytoscape was used to collect the significant modules in the PPI network complex.

Construction of miRNA-DEG regulatory network

The miRNet database (https://www.mirnet.ca/) [24] is a database, containing miRNAs involved in various diseases. The miRNAs related to PDAC were searched from miRNet database. Through getting the intersection of the miRNAs and the DEGs, the miRNA-DEG regulatory relationships were selected. Finally, miRNA-DEG regulatory network was built using Cytoscape software.

Construction of TF-DEG regulatory network

The NetworkAnalyst database (https://www.networkanalyst.ca/) [25] is a database, containing TFs involved in various diseases. The TFs related to PDAC were searched from TF database. Through getting the intersection of the TFs s and the DEGs, the TF-DEG regulatory relationships were selected. Finally, TFs-DEG regulatory network was built using Cytoscape software.

Hub genes validation

After hub genes identified from expression profiling by high throughput sequencing dataset, UALCAN (http://ualcan.path.uab.edu/analysis.html) [26] was used to validate the selected up regulated and down regulated hub genes. UALCAN is an online tool for gene expression analysis between PDAC and normal data from The Cancer Genome Atlas (TCGA). It adds data such as gene expression, tumor staging, and survival period for PDAC. cBioPortal is an online platform (http://www.cbioportal.org) [27] for gene alteration of hub genes analysis from TCGA. Human protein atlas is an online database (HPA, www.proteinatlas.org) [28] for protein expression analysis between PDAC and normal data from TCGA. TIMER is an online platform (https://cistrome.shinyapps.io/timer/) [29] for immune infiltration analysis from TCGA. To explore diagnostic biomarkers of PDAC, we used the above hub genes as candidates to find their diagnostic value based on generalized linear model (GLM) [30]. The pROC in R was used for Receiver operating characteristic (ROC) curve analysis [30]. In brief, half of the samples (PDAC = 142, controls = 59) were aimlessly distributed as the training set and remaining data were used as the test set, which were used to set up a model. An ROC curve analysis was tested to calculate the specificity and sensitivity of the GLM prediction model. The area under the curve (AUC) was figure out to determine the diagnostic efficiency of the classifier.

RT-PCR analysis

TRI Reagent® (Sigma, USA) was used to extract total RNA from the culture cells of PDAC (CRL-2549™) and normal (CRL-2989™) according to the manufacturer’s protocol. Reverse transcription cDNA kit (Thermo Fisher Scientific, Waltham, MA, USA) and random primers were used to synthesize cDNA. Quantitative real-time PCR (qRT-PCR) was conducted on the 7 Flex real-time PCR system (Thermo Fisher Scientific, Waltham, MA, USA). The reaction guideline included a denaturation program (5 min at 95 °C), followed by an amplification and quantification program over 40 cycles (15 s at 95°C and 45 s at 65°C). Each sample was tested in triplicates. Table 1 depicts the primer sequences of hub genes. The expression level was resolved as a ratio between the hub genes and the internal control β-actin in the same mRNA sample, and determined by the comparative CT method [31]. Levels of CCNB1, FHL2, HLA-DPA1 and TUBB1 expression were determined by the 2−ΔΔCt method.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 1

The sequences of primers for quantitative RT-PCR

Molecular docking experiment

The module SYBYL-X 2.0 perpetual software were used for Surflex-Docking of the designed molecules. The molecules were sketched by using ChemDraw Software and imported and saved in sdf. format using openbabel free software. The protein structures of CyclinB1 (CCNB1) its co-crystallisedprotein of PDB code 4Y72, 5H0V and Four and half LIM domains 2 (FHL2) its NMR structure of proteins 2D8Z and 2EHE was retrieved from Protein Data Bank [32-34]. Together with the TRIPOS force field, GasteigerHuckel (GH) charges were added to all designed derivatives for the structure optimization process.In addition, energy minimization was carried outusing MMFF94s and MMFF94 algorithm process. Protein processing was carried out after the incorporation of protein.The co-crystallized ligand and all water molecules were removed from the crystal structure; more hydrogens were added and the side chain was set. TRIPOS force field was used for the minimization of structure. The compounds’ interaction efficiency with the receptor was represented by the Surflex-Dock score in kcal / mol units. The interaction between the protein and the ligand, the best pose was incorporated into the molecular area. The visualisation of ligand interaction with receptor is done by using discovery studio visualizer.

Results

Identification of DEGs

We analyzed the DEGs of GSE133684 by using the limma package. We used p□<□0.05 and |logFC|□≥□1 as the cutoff criteria. We screened 463 DEGs, including 232 up regulated genes and 231 down regulated genes in PDAC samples compared with normal control samples and are listed in Table 2. We identified all the DEGs which were shown in the above volcano map according to the value of |logFC| is shown in Fig. 1 and then displayed the DEGs on a heatmap is shown in Fig. 2.

View this table:
  • View inline
  • View popup
Table 2

The statistical metrics for key differentially expressed genes (DEGs)

Fig. 1.
  • Download figure
  • Open in new tab
Fig. 1.

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.

Fig. 2.
  • Download figure
  • Open in new tab
Fig. 2.

Heat map of differentially expressed genes. Legend on the top left indicate log fold change of genes. (A1 – A114 = normal control samples; B1 – B284 = PDAC samples)

GO analysis and pathway enrichment analysis of DEGs

To symbolize the function of the DEGs and to identify important candidate pathways, GO functional enrichment analysis and REACTOME pathway enrichment analysis were performed. The results of GO categories analysis including biological processes (BP), cellular components (CC) and molecular functions (MF) are listed in Table 3. Firstly, the up regulated genes were annotated with the BP category, including vesicle organization and secretion, whereas the down regulated genes were annotated with the GO terms, including lymphocyte activation and regulation of cell death. Secondly, the up regulated genes were annotated with the GO terms of the CC category, namely secretory vesicle and whole membrane, whereas the down regulated genes were annotated with the GO terms, including cell surface and intrinsic component of plasma membrane.

View this table:
  • View inline
  • View popup
Table 3

The enriched GO terms of the up and down regulated differentially expressed genes

Thirdly, the up regulated genes were annotated with the GO terms of the MF category, such as protein dimerization activity and signaling receptor binding, whereas the down regulated genes were annotated with the GO terms, including transferase activity, transferring phosphorus-containing groups and drug binding. As shown in Table 4, the significantly enriched REACTOME pathways of the up regulated genes with P<0.05 were hemostasis and cell cycle, whereas down regulated genes with P<0.05 were adaptive immune system and transmembrane transport of small molecules.

View this table:
  • View inline
  • View popup
Table 4

The enriched pathway terms of the up and down regulated differentially expressed genes

Protein-protein interaction (PPI) network construction and module analysis

After all the DEGs were uploaded to the online IID interactome database, the PPI network with 6188 nodes and 13153 edges was constructed using the Cytoscape software (Fig. 3A). Hub DEGs with the node degree, betweenness centrality, stress centrality and closeness centrality were obtained and are listed in Table 5. Among them, CCNB1 and FHL2 were the major up regulated genes, while HLA-DPA1 and TUBB1 were the major down regulated genes. Then, two significant module that fulfilled the cut-off criteria, namely, PEWCC1 scores >3 and number of nodes >5, was screened (Fig. 3B and Fig. 3C). The FGB, FGA, FGG, EEF1A1, RPL13A, ITGA4, RPL27A, RPL23A and RPL10 genes were identified in these modules. GO analysis of these genes showed that they were annotated with vesicle organization, regulation of cell death and lymphocyte activation. In addition, the REACTOME enrichment analysis suggested that these genes were mainly involved in hemostasis, innate immune system, disease and adaptive immune system.

View this table:
  • View inline
  • View popup
Table 5

Topology table for up and down regulated genes.

Fig. 3.
  • Download figure
  • Open in new tab
Fig. 3.

PPI network and the most significant modules of DEGs. (A) The PPI network of DEGs was constructed using Cytoscape (B) The most significant module was obtained from PPI network with 16 nodes and 44 edges for up regulated genes (C) The most significant module was obtained from PPI network with 6 nodes and 20 edges for up regulated genes. Up regulated genes are marked in green; down regulated genes are marked in red

Construction of miRNA-DEG regulatory network

The regulatory network of miRNA-DEG and predicted targets is presented in Fig. 4A. Notably, MAP1B targeted 202 miRNAs, including hsa-mir-4461; CCNB1 targeted 94 miRNAs, including hsa-mir-3928-3p; AHNAK targeted 256 miRNAs, including hsa-mir-2682-5p; KMT2D targeted 209 miRNAs, including hsa-mir-1202 and top 20 are listed in Table 6. As a group, a total of 257 of the 463 DEGs were contained in the miRNA-DEG regulatory network.

View this table:
  • View inline
  • View popup
Table 6

miRNA - target gene and TF - target gene interaction

Fig. 4.
  • Download figure
  • Open in new tab
Fig. 4.

(A) Target gene - miRNA regulatory network between target genes and miRNAs (B) Target gene - TF regulatory network between target genes and TFs. Up regulated genes are marked in green; down regulated genes are marked in red; The blue color diamond nodes represent the key miRNAs; the gray color triangle nodes represent the key TFs

Construction of TF-DEG regulatory network

The regulatory network of TF-DEG and predicted targets is presented in Fig. 4B. Notably, EZH2 targeted 45 TFs, including SOX2; TPM1 targeted 40 TFs, including MYC; AHNAK targeted 58 TFs, including KLF4, TXNIP targeted 51 TFs, including TP63 and top 20 are listed in Table 6. As a group, a total of 259 of the 463 DEGs were contained in the TF-DEG regulatory network.

Hub genes validation

All of the hub genes were validated in TCGA data. Hub genes contributed to the survival period in patients with PDAC, we analyzed the overall survival (OS) for each hub gene by UALCAN (Fig. 5). The results showed that the high expression of CCNB1 and FHL2 mRNA level were associated with the worse OS in patients with PDAC, while low expression of HLA-DPA1 and TUBB1 mRNA level were associated with the worse OS in patients with PDAC. As shown in Fig. 6, the expression of the up regulated hub genes CCNB1 and FHL2 in PDAC were significantly elevated compared with normal, while expression of the down regulated hub genes HLA-DPA1 and TUBB1 in PDAC were significantly decreased compared with normal. The expression of each hub gene in PDAC patients was analyzed according to the individual cancer stage. As shown in Fig. 7, the expression of CCNB1 and FHL2 were higher in patients with all individual cancer stages than that in normal, which revealed that these up regulated hub genes might be associated with tumor progression positively, where as the expression of HLA-DPA1 and TUBB1 were lower in patients with all individual cancer stages than that in normal, which revealed that these down regulated hub genes might be associated with tumor progression positively. We used cBioportal tool to explore the specific mutation of hub genes in PDAC dataset with 184 samples. From the OncoPrint, percentages of alterations in CCNB1, FHL2, HLA-DPA1 and TUBB1 genes among lung cancer ranged from 0% to 2.3% in individual genes (CCNB1, 0%; FHL2, 0.6%; HLA-DPA1, 2.3%; TUBB1, 2.3%) and is shown in Fig. 8. In addition, we used the ‘HPA’ to examine the protein expression levels of CCNB1 and FHL2, and observed that the protein expression levels of the these hub genes were noticeably up regulated in PDAC compared with normal tissues, whereas protein expression levels of HLA-DPA1 and TUBB1, and observed that the protein expression levels of the these hub genes were noticeably down regulated in PDAC compared with normal tissues (Fig. 9). The association of CCNB1, FHL2, HLA-DPA1 and TUBB1 expression level with immune infiltration abundance in PDAC was evaluated using TIMER database. CCNB1 and FHL2 expression were negatively correlated with infiltration degree of B cells, CD8+ T cells, macrophage, neutrophil, and dendritic cells, where as HLA-DPA1 and TUBB1 were positively correlated with infiltration degree of B cells, CD8+ T cells, macrophage, neutrophil, and dendritic cells and is shown in Fig. 10. As these 4 genes are prominently expressed in PDAC, we performed a ROC curve analysis to evaluate their sensitivity and specificity for the diagnosis of PDAC. As shown in Fig. 11, CCNB1, FHL2, HLA-DPA1 and TUBB1 achieved an AUC value of >0.70, demonstrating that these genes have high sensitivity and specificity for PDAC diagnosis. The results suggested that CCNB1, FHL2, HLA-DPA1 and TUBB1 can be used as biomarkers for the diagnosis of PDAC.

Fig. 5.
  • Download figure
  • Open in new tab
Fig. 5.

Overall survival analysis of hub genes. Overall survival analyses were performed using the UALCAN online platform. Red line denotes - high expression; Blur line denotes – low expression. A) CCNB1 B) FHL2 C) HLA-DPA1 D) TUBB1

Fig. 6.
  • Download figure
  • Open in new tab
Fig. 6.

Box plots (expression analysis) hub genes were produced using the UALCAN platform. A) CCNB1 B) FHL2 C) HLA-DPA1 D) TUBB1

Fig. 7.
  • Download figure
  • Open in new tab
Fig. 7.

Box plots (clinical stage analysis) hub genes were produced using the UALCAN platform. A) CCNB1 B) FHL2 C) HLA-DPA1 D) TUBB1

Fig. 8.
  • Download figure
  • Open in new tab
Fig. 8.

Mutation analyses of hub genes were produced using the CbioPortal online platform. A) CCNB1 B) FHL2 C) HLA-DPA1 D) TUBB1

Fig. 9.
  • Download figure
  • Open in new tab
Fig. 9.

Immunohisto chemical (IHC) analyses of hub genes were produced using the human protein atlas (HPA) online platform. A) CCNB1 B) FHL2 C) HLA-DPA1 D) TUBB1

Fig. 10.
  • Download figure
  • Open in new tab
Fig. 10.

Scatter plot for immune infiltration for hub genes. A) CCNB1 B) FHL2 C) HLA-DPA1 D) TUBB1

Fig. 11.
  • Download figure
  • Open in new tab
Fig. 11.

ROC curve validated the sensitivity, specificity of hub genes as a predictive biomarker for PDAC prognosis. A) CCNB1 B) FHL2 C) HLA-DPA1 D) TUBB1

RT-PCR analysis

Next, in order to verify the results of previous bioinformatics analysis, the gene expression levels of CCNB1, FHL2, HLA-DPA1 and TUBB1 were detected by RT-PCR. As shown in Fig 12, CCNB1 and FHL2 mRNA expression levels were significantly up regulated in the PDAC compared to normal, and HLA-DPA1 and TUBB1 mRNA level were down regulated compared to normal, which was consistent with the results of bioinformatics analysis.

Fig. 12.
  • Download figure
  • Open in new tab
Fig. 12.

Validation of hub genes by RT-PCR. A) CCNB1 B) FHL2 C) HLA-DPA1 D) TUBB1

Molecular docking experiment

The docking simulation was performed in the present study to recognize the active site conformation andsignificant interactions, which are responsible for complex stability with ligand receptor. Novel molecules containing alkylating group and purine heterocyclic ring were designed and performed docking studies using Sybyl X 2.1 drug design software. Molecules containing alkylating group is designed due to non-specific alkylation of physiologically important groupings and purine heterocyclic ring is incorporated due to structural similarity of purine derivatives and to compete for the synthesis of proteins. The proteins which are over expressed in pancreatic duct adenocarcinoma are selected for docking studies. The two proteins of each over expressedcyclin B1 (CCNB1)its co-crystallised protein of PDB code 4Y72, 5H0V and Four and half LIM domains 2 (FHL2) of NMR structure of proteins 2D8Z and 2EHE were selected for docking. The investigation of designed molecules was performed to identify the potential molecule. The most of the designed molecules obtained C-score greater than 5 and are active having the c-score greater than 5 are said to be an active, among total of 48 designed molecules few molecules have excellent good binding energy (C-score) greater than 8 respectively. Few of the designed molecules IM 11 & PU 42, shown good binding score of 7.83& 8.57 and the molecules IM 13, TZ 23, TZ 27, TZ 37, PU 41, PU 43 & PU 49 have good binding score 8.013, 8.523, 8.235, 8.800, 10.338, 10.891& 9.411 with CCNB1 PDB code 1H0V and 4Y72 respectively and are shown in Fig. 13. Molecules of IM 09, IM 10, IM & 18 shown good binding score of 7.14, 7.75 & 7.80 and the molecules IM 8 and TZ 24 with binding score 6.24 and 6.32 with FHL2 of PDB code 2D8Z and 2EQQ respectively, the values are depicted in Table 7. The molecule IM 8has highest binding score its interaction with protein 1H0V and hydrogen bonding and other bonding interactions with amino acids are depicted by 3D and 2D shown in Fig. 14 and Fig. 15.

View this table:
  • View inline
  • View popup
Table 7.

Docking results of designed molecules on CCNB1 and FHL2

Fig. 13.
  • Download figure
  • Open in new tab
Fig. 13.

Structures of Designed Molecules

Fig. 14.
  • Download figure
  • Open in new tab
Fig. 14.

3D Binding of Molecule IM 8 with 1H0V

Fig. 15.
  • Download figure
  • Open in new tab
Fig. 15.

2D Binding of Molecule IM 8 with 1H0V

Discussion

Due to the high heterogeneity of PDAC, PDAC was still a disease with high rates of pervasiveness and fatality. With surgery as the main, the other treatments including radiotherapy, chemotherapy, targeted therapy, and gene therapy as a additive to the finite treatment measures of PDAC, the 5-year survival rate was still less than 8% [35]. Therefore, the early diagnosis and effective treatment of PDAC is crucially required, which may be achieved via the identification of the DEGs between PDAC and normal control, and by considerate the underlying molecular mechanism. Microarray and high throughput sequencing analysis can screen a massive number of genes in the human genome for farther functional analysis, and can be extensively used to screen biomarkers for early diagnosis and unique therapeutic targets. Therefore, they may help the diagnosis of PDAC in the early stages and the advancement of targeted treatment, thus developing prognosis.

The current investigation systematically applied integrated bioinformatics methods to identify novel biomarkers that serve roles in the advancement PDAC. The data extracted from the GEO dataset contained 31 pairs of lung cancer and normal samples. A total of 232 up regulated and 231 down regulated genes in PDAC, when compared with normal control samples, were identified using bioinformatics analysis, indicating that the incidence and advancement of PDAC. The results of the DEGs may provide potential biomarkers for the diagnosis of PDAC. DAP (death associated protein) [36], KRT8 [37], IGFBP2 [38], KRT19 [39], CD44 [40], AHNAK (AHNAK nucleoprotein) [41] and BTG1 [42] are a noticeable factors in the PDAC progression. Wang et al [43] reported that KIF2C induces proliferation, migration, and invasion in gastric cancer patients through the MAPK signaling pathway, but this gene might be associated with development of PDAC. DBN1 expression was significantly increased in breast cancer [44], but this gene might be liable for development of PDAC. MAP1B was reported to lung cancer progression [45], but this gene may be key for advancement of PDAC. BNIP3L down regulation was required to develop breast and ovarian cancer [46], but this down regulation of gene might be involved in progression of PDAC. Yen et al [47] reported that ITGA4 was expressed in oral cancer, but this gene might be novel biomarker for PDAC. Tomsic et al. [48] showed that mutation in SRRM2 was associated with progression of thyroid carcinoma, but alteration in this gene might be key factor for advancement of PDAC. Recent studies have shown that down regulation of IL7R is associated with progression of esophageal squamous cell carcinoma [49], but this gene might be involved in pathogenesis of PDAC. Lee et al [50] found that reduced expression of the HLA-DRA is key factor for development of colorectal cancer, but this gene might be linked with advancement of PDAC. Liu et al. [51] reported that the absence of SESN3 linked with development of hepatocellular carcinoma, but this gene might be associated with progression of PDAC.

Then, GO and REACTOME pathway analyses were used to investigate the interactions of these DEGs. Increasing evidence shows that LAPTM4B [52], CEACAM6 [53], SERPINE2 [54] and VNN1 [55], SPHK1 [56], HRG (histidine rich glycoprotein) [57], VEGFC (vascular endothelial growth factor C) [58], ANXA3 [59], APOA2 [60], LCN2 [61], TIMP1 [62], CD63 [63], CD151 [64], MAL2 [65], ARNTL2 [66], PKD2 [67], E2F1 [68], MMP1 [69], CCR7 [70], NOTCH2 [71], BTLA (B and T lymphocyte associated) [72], TFRC (transferrin receptor) [73], CD4 [74], ATM (ATM serine/threonine kinase) [75], LEF1 [76], CSF1R [77], CTSB (cathepsin B) [78], DUSP2 [79] and NR4A1 [80] are closely associated with progression of PDAC. PTGER3 [81] and MAGI2 [82] are linked with angiogenesis, chemoresistance, cell proliferation and migration in ovary cancer, but these genes might be liable for growth PDAC. Hoagland et al [83] demonstrated that HP (haptoglobin) expression was responsible for progression of lung cancer, but this gene might be involved in PDAC progression. FGA (fibrinogen alpha chain) was demonstrated to be a lung cancer susceptibility gene through activation of integrin–AKT signaling pathway [84], but this gene might be liable for progression of PDAC. Repetto et al [85] investigated the importance of FGB (fibrinogen beta chain) in the pathogenesis of gastric carcinoma, but this gene might be responsible for progression of PDAC. PLA2G4A [86], FGG (fibrinogen gamma chain) [87] and TYMS (thymidylatesynthetase) [88] have been demonstrated to be up regulated in cancer, but these genes might be liable for progression of PDAC. RAB32 [89], SEPTIN4 [90], TPM2 [91], ACOT7 [92], PRTFDC1 [93], CABLES1 [94], HLA-DMB [95], PTPRC (protein tyrosine phosphatase receptor type C) [96], CD5 [97], CD6 [97], MS4A1 [98], CD22 [99], CD27 [100], MRC2 [101], CLEC2D [102], EEF1A1 [103] and APOB (apolipoprotein B) [104] played a predominant role in the cancer progression, but these genes might be associated with development of PDAC. Jung et al [105] found that SMPD1 stimulates the drug resistance in colorectal cancer, but this gene might be linked with development of PDAC. Liu et al. [106], Yang et al [107], Song et al [108], Seachrist et al [109], Zhu et al [110], Wu et al [111], Wang et al [112], Yi et al [113], Lan et al [114] and Appert-Collin et al [115] revealed that PADI4, MAOB (monoamine oxidase B), TRPC6, BCL11A, CXCR5, TCF7, POU2F2, SLC4A1, STK17B and LRP1 were associated with cancer cell invasion, but these genes might be liable for progression of PDAC. Kairouz et al [116], Diez-Bello et al [117], Xue et al [118], Abo-Elfadl et al [119], Li et al [120] and Zhao et al [121] reported that GRB14, TRPC6, ZFPM2, TNFRSF13B, ADAM19 and PIK3IP1enhance the cancer cell proliferation, but this gene might be involved in advancement of PDAC. Leite et al [122], Feng et al [123], Wang et al [124], Zhong et al [125], Yokoyama-Mashima et al [126], Guo et al [127], Lawson et al [128] and Wang et al [129] demonstrated that low levels of HLA-DPA1, FGL2, CBLB (Cbl proto-oncogene B), NCKAP1L, DYRK2, OGT (O-linked N-acetylglucosamine (GlcNAc) transferase), CAMK1D and RNF213 are linked with progression of cancer, but these genes might be essential for progression of PDAC. Polymorphic genes such as RORA (RAR related orphan receptor A) [130], IGF2R [131] and ZBTB20 [132] are contribute to progression of cancer, but these genes might be crucial for advancement of PDAC. Hope et al. [133] identified that the VCAN (versican) was central role in immune cell infiltration in cancer, but this gene may be associated with immune cell infiltration in PDAC.

Construction of PPI network and modules of DEGs may be helpful for understanding the relationship of developmental PDAC. Bai et al [134] reported that CCNB1 plays a positive role in proliferation of cancer cells, but this gene might be involved in development of PDAC. FHL2 [135] and RPL10 [136] are associated with progression of PDAC. Further investigation is required in order to clarify the underlying biological mechanisms of novel biomarkers HLA-DPA1, TUBB1, RPL13A, RPL27A and RPL23A on PDAC.

The miRNA-DEG regulatory network and TF-DEG regulatory network were constructed to explore the molecular mechanism of PDAC. The EZH2 [137], KMT2D [138], TXNIP (thioredoxin interacting protein) [139], TP63 [140], SOX2 [141], MYC [142] and KLF4 [143] genes are associated with PDAC. TPM1 [144] and hsa-mir-1202 [145] have been associated with cancer risk, but these genes and miRNAs might be responsible for progression of PDAC. Hsa-mir-4461, hsa-mir-3928-3p and hsa-mir-2682-5p might be considered as novel biomarkers for progression of PDAC.

In conclusion, we aim to identify DEGs by bioinformatics analysis to find the potential biomarkers which may be involved in the advancement of PDAC. The investigation contributes a set of useful DEGs for future studies into molecular mechanisms and biomarkers of PDAC. And the application of data mining and integration is accessible for prediction of PDAC advancement. Nevertheless, further molecular biological analyses are recommended to certify the function of the DEGs in PDAC.

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical approval

This article does not contain any studies with human participants or animals performed by any of the authors.

Informed consent

No informed consent because this study does not contain human or animals participants.

Availability of data and materials

The datasets supporting the conclusions of this article are available in the GEO (Gene Expression Omnibus) (https://www.ncbi.nlm.nih.gov/geo/) repository. [(GSE133684) (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE133684]

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Data Availability

The datasets supporting the conclusions of this article are available in the GEO (Gene Expression Omnibus) (https://www.ncbi.nlm.nih.gov/geo/) repository. [(GSE133684) (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE133684]

Author Contributions

Basavaraj Vastrad-Writing original draft, and review and editing

Anandkumar Tengli - Writing original draft and investigation

Chanabasayya Vastrad - Software and investigation

Acknowledgement

I thank Shenglin Huang, Fudan University, Shanghai, China, very much, the author who deposited their microarray dataset, GSE133684, into the public GEO database.

References

  1. 1.↵
    Thiruvengadam, S.S.; Chuang, J.; Huang, R.; Girotra, M.; Park, W.G. Chronic pancreatitis changes in high-risk individuals for pancreatic ductal adenocarcinoma. Gastrointest. Endosc 2019,89,842-851.e1. doi:10.1016/j.gie.2018.08.029
    OpenUrlCrossRef
  2. 2.↵
    Al-Hawary, M.M.; Kaza, R.K.; Azar, S.F.; Ruma, J.A.; Francis, I.R. Mimics of pancreatic ductal adenocarcinoma. Cancer. Imaging 2013,13,342–349. doi:10.1102/1470-7330.2013.9012
    OpenUrlCrossRef
  3. 3.↵
    Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2018. CA. Cancer. J. Clin 2018,68,7–30. doi:10.3322/caac.21442
    OpenUrlCrossRefPubMed
  4. 4.↵
    Adamska, A.; Domenichini, A.; Falasca, M. Pancreatic Ductal Adenocarcinoma: Current and Evolving Therapies. Int. J. Mol. Sci 2017,18,1338. doi:10.3390/ijms18071338
    OpenUrlCrossRef
  5. 5.↵
    Tsuchiya, N.; Matsuyama, R.; Murakami, T.; Yabushita, Y.; Sawada, Y.U; Kumamoto, T.; Endo, I. Risk Factors Associated With Early Recurrence of Borderline Resectable Pancreatic Ductal Adenocarcinoma After Neoadjuvant Chemoradiation Therapy and Curative Resection. Anticancer. Res 2019,39,4431–4440. doi:10.21873/anticanres.13615
    OpenUrlAbstract/FREE Full Text
  6. 6.↵
    Oji, Y.; Nakamori, S.; Fujikawa, M.; Nakatsuka, S.; Yokota, A.; Tatsumi, N.; Abeno, S.; Ikeba, A.; Takashima, S.; Tsujie, M.; et al. Overexpression of the Wilms’ tumor gene WT1 in pancreatic ductal adenocarcinoma. Cancer. Sci. 2004,95,583–587. doi:10.1111/j.1349-7006.2004.tb02490.x
    OpenUrlCrossRefPubMed
  7. 7.↵
    Murthy, D.; Attri, K.S.; Singh, P.K. Phosphoinositide 3-Kinase Signaling Pathway in Pancreatic Ductal Adenocarcinoma Progression, Pathogenesis, and Therapeutics. Front. Physiol 2018,9,335. doi:10.3389/fphys.2018.00335
    OpenUrlCrossRef
  8. 8.↵
    Reddy, R.R.S; Ramanujam, M.V. High Throughput Sequencing-Based Approaches for Gene Expression Analysis. Methods. Mol. Biol 2018,1783,299–323. doi:10.1007/978-1-4939-7834-2_15
    OpenUrlCrossRef
  9. 9.↵
    Gu, Y.; Feng, Q.; Liu, H.; Zhou, Q.; Hu, A.; Yamaguchi, T.; Xia, S.; Kobayashi, H. Bioinformatic evidences and analysis of putative biomarkers in pancreatic ductal adenocarcinoma. Heliyon 2019,5,e02378. doi:10.1016/j.heliyon.2019.e02378
    OpenUrlCrossRef
  10. 10.↵
    Clough, E.; Barrett, T. The Gene Expression Omnibus Database. Methods. Mol. Biol 2016,1418,93□110. doi:10.1007/978-1-4939-3578-9_5
    OpenUrlCrossRefPubMed
  11. 11.↵
    Yu, S.; Li, Y.; Liao, Z.; Wang, Z.; Wang, Z.; Li, Y.; Qian, L.; Zhao, J.; Zong, H.; Kang, B.; et al. Plasma extracellular vesicle long RNA profiling identifies a diagnostic signature for the detection of pancreatic ductal adenocarcinoma. Gut 2020,69,540–550. doi:10.1136/gutjnl-2019-318860
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    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. doi:10.1093/nar/gkv007
    OpenUrlCrossRefPubMed
  13. 13.↵
    Ferreira, J.A. The Benjamini-Hochberg method in the case of discrete test statistics. Int. J. Biostat 2007,3. doi:10.2202/1557-4679.1065
    OpenUrlCrossRef
  14. 14.↵
    Thomas, P.D. The Gene Ontology and the Meaning of Biological Function. Methods. Mol. Biol 2017,1446,15□24. doi:10.1007/978-1-4939-3743-1_2
    OpenUrlCrossRef
  15. 15.↵
    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. doi:10.1093/nar/gkx1132
    OpenUrlCrossRefPubMed
  16. 16.↵
    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. doi:10.1093/nar/gkp427
    OpenUrlCrossRefPubMedWeb of Science
  17. 17.
    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. doi:10.1093/nar/gky1037
    OpenUrlCrossRefPubMed
  18. 18.↵
    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. doi:10.1101/gr.1239303
    OpenUrlAbstract/FREE Full Text
  19. 19.↵
    Przulj, N.; Wigle, D.A.; Jurisica, I. Functional topology in a network of protein interactions. Bioinformatics 2004,20,340–348. doi:10.1093/bioinformatics/btg415
    OpenUrlCrossRefPubMedWeb of Science
  20. 20.↵
    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. doi:10.1186/1752-0509-5-179
    OpenUrlCrossRefPubMed
  21. 21.↵
    Shi, Z.; Zhang, B. Fast network centrality analysis using GPUs. BMC. Bioinformatics 2011,12,149. doi:10.1186/1471-2105-12-149
    OpenUrlCrossRefPubMed
  22. 22.↵
    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. doi:10.1186/1752-0509-8-6
    OpenUrlCrossRef
  23. 23.↵
    Zaki, N.; Efimov, D.; Berengueres, J. Protein complex detection using interaction reliability assessment and weighted clustering coefficient. BMC. Bioinformatics 2013,14,163. doi:10.1186/1471-2105-14-163
    OpenUrlCrossRefPubMed
  24. 24.↵
    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. doi:10.1007/978-1-4939-8618-7_10
    OpenUrlCrossRef
  25. 25.↵
    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. doi:10.1093/nar/gkz240
    OpenUrlCrossRef
  26. 26.↵
    Chandrashekar, D.S.; Bashel, B.; Balasubramanya, S.A.H.; Creighton, C.J: Ponce-Rodriguez, I.; Chakravarthi, B.V.S.K.; Varambally, S. UALCAN: A Portal for Facilitating Tumor Subgroup Gene Expression and Survival Analyses. Neoplasia 2017,19,649–658. doi:10.1016/j.neo.2017.05.002
    OpenUrlCrossRefPubMed
  27. 27.↵
    Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha R.; Larsson E.; et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013,6,pl1. doi:10.1126/scisignal.2004088
    OpenUrlAbstract/FREE Full Text
  28. 28.↵
    Uhlen, M.; Oksvold, P.; Fagerberg, L.; Lundberg, E.; Jonasson, K.; Forsberg, M.; Zwahlen, M.; Kampf, C.; Wester, K.; Hober, S.; et al. Towards a knowledge-based Human Protein Atlas. Nat. Biotechnol 2010,28,1248–1250. doi:10.1038/nbt1210-1248
    OpenUrlCrossRefPubMed
  29. 29.↵
    Li, T.; Fan, J.; Wang, B.; Traugh, N.; Chen Q.; Liu J.S.; Li B.; Liu X.S. TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells. Cancer. Res 2017,77,e108–e110. doi:10.1158/0008-5472.CAN-17-0307
    OpenUrlAbstract/FREE Full Text
  30. 30.↵
    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. Bioinformatics 2011,12,77. doi:10.1186/1471-2105-12-77
    OpenUrlCrossRefPubMed
  31. 31.↵
    Livak, K.J.; Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 2001,25,402–408. doi:10.1006/meth.2001.1262
    OpenUrlCrossRefPubMedWeb of Science
  32. 32.↵
    El-Saadi M.W.; Williams-Hart T.; Salvatore B.A.; Mahdavian E. Use of insilico assays to characterize the ADMET profile and identify potential herapeutic targets of fusarochromanone, a novel anticancer agent. In. Silico. Pharmacol 2015,3,6. doi:10.1186/s40203-015-0010-5
    OpenUrlCrossRef
  33. 33.
    Li, J.; Ma, X.; Liu, C.; Li, H.; Zhuang, J.; Gao, C.; Zhou, C.; Liu, L.; Wang, K.; Sun C. Exploring the Mechanism of Danshen against Myelofibrosis by Network Pharmacology and Molecular Docking. Evid. Based. Complement. Alternat. Med. 2018,2018,8363295. doi:10.1155/2018/8363295
    OpenUrlCrossRef
  34. 34.↵
    Ruddarraju, R.R.; Murugulla, A.C.; Kotla, R.; Tirumalasetty, M.C.B.; Wudayagiri, R.; Donthabakthuni, S.; Maroju, R. Design, synthesis, anticancer activity and docking studies of theophylline containing 1,2,3-triazoles with variant amide derivatives. Medchemcomm 2016,8,176–183. doi:10.1039/c6md00479b
    OpenUrlCrossRef
  35. 35.↵
    Moffitt, R.A.; Marayati, R.; Flate, E.L.; Volmar, K.E.; Loeza, S.G.; Hoadley, K.A.; Rashid, N.U.; Williams, L.A.; Eaton, S.C.; Chung, A.H.; et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet 2015,47,1168–1178. doi:10.1038/ng.3398
    OpenUrlCrossRefPubMed
  36. 36.↵
    Dansranjavin, T.; Möbius, C.; Tannapfel, A.; Bartels, M.; Wittekind, C.; Hauss, J.; Witzigmann, H. E-cadherin and DAP kinase in pancreatic adenocarcinoma and corresponding lymph node metastases. Oncol. Rep 2006,15,1125–1131.
    OpenUrlPubMed
  37. 37.↵
    Treiber, M.; Schulz, H.U.; Landt, O.; Drenth, J.P.; Castellani, C.; Real, F.X.; Akar, N.; Ammann, R.W.; Bargetzi, M.; Bhatia, E.; et al. Keratin 8 sequence variants in patients with pancreatitis and pancreatic cancer. J. Mol. Med (Berl) 2006,84,1015–1022. doi:10.1007/s00109-006-0096-7
    OpenUrlCrossRefPubMed
  38. 38.↵
    Liu, H.; Li, L.; Chen, H.; Kong, R.; Pan, S.; Hu, J.; Wang, Y.; Li, Y.; Sun B. Silencing IGFBP-2 decreases pancreatic cancer metastasis and enhances chemotherapeutic sensitivity. Oncotarget 2017,8,61674–61686. doi:10.18632/oncotarget.18669
    OpenUrlCrossRef
  39. 39.↵
    Yao, H.; Yang, Z.; Liu, Z.; Miao, X.; Yang, L.; Li, D.; Zou, Q.; Yuan, Y. Glypican-3 and KRT19 are markers associating with metastasis and poor prognosis of pancreatic ductal adenocarcinoma. Cancer. Biomark 2016,17,397–404. doi:10.3233/CBM-160655
    OpenUrlCrossRef
  40. 40.↵
    Huynh, D.L.; Koh, H.; Chandimali, N.; Zhang, J.J.; Kim, N.; Kang, T.Y.; Ghosh, M.; Gera, M.; Park, Y.H.; Kwon, T.; et al. BRM270 Inhibits the Proliferation of CD44 Positive Pancreatic Ductal Adenocarcinoma Cells via Downregulation of Sonic Hedgehog Signaling. Evid. Based. Complement Alternat. Med. 2019,2019,8620469. doi:10.1155/2019/8620469
    OpenUrlCrossRef
  41. 41.↵
    Zhang, Z.; Liu, X.; Huang, R.; Liu, X.; Liang, Z.; Liu, T. Upregulation of nucleoprotein AHNAK is associated with poor outcome of pancreatic ductal adenocarcinoma prognosis via mediating epithelial-mesenchymal transition. J. Cancer 2019,10,3860–3870. doi:10.7150/jca.31291
    OpenUrlCrossRef
  42. 42.↵
    Huang, Y.; Zheng, J.; Tan, T.; Song, L.; Huang, S.; Zhang, Y.; Lin, L.; Liu, J.; Zheng P.; Chen X.; et al. BTG1 low expression in pancreatic ductal adenocarcinoma is associated with a poorer prognosis. Int. J. Biol. Markers. 2018,33,189–194. doi:10.5301/ijbm.5000310
    OpenUrlCrossRef
  43. 43.↵
    Wang, P.B.; Chen, Y.; Ding, G.R.; Du, H.W.; Fan, H.Y. Keratin 18 induces proliferation, migration, and invasion in gastric cancer via the MAPK signalling pathway. Clin. Exp. Pharmacol. Physiol. 2020, 10.1111/1440-1681.13401. doi:10.1111/1440-1681.13401
    OpenUrlCrossRef
  44. 44.↵
    Alfarsi, L.H.; El Ansari, R.; Masisi, B.K.; Parks, R.; Mohammed, O.J.; Ellis, I.O.; Rakha, E.A.; Green, A.R. Integrated Analysis of Key Differentially Expressed Genes Identifies DBN1 as a Predictive Marker of Response to Endocrine Therapy in Luminal Breast Cancer. Cancers. (Basel). 2020,12,1549. doi:10.3390/cancers12061549
    OpenUrlCrossRef
  45. 45.↵
    Tessema, M.; Yingling, C.M.; Picchi, M.A.; Wu, G.; Liu, Y.; Weissfeld, J.L.; Siegfried, J.M.; Tesfaigzi, Y.; Belinsky, S.A. Epigenetic Repression of CCDC37 and MAP1B Links Chronic Obstructive Pulmonary Disease to Lung Cancer. J. Thorac. Oncol 2015,10,1181–1188. doi:10.1097/JTO.0000000000000592
    OpenUrlCrossRef
  46. 46.↵
    Lai, J.; Flanagan, J.; Phillips, W.A.; Chenevix-Trench, G.; Arnold, J. Analysis of the candidate 8p21 tumour suppressor, BNIP3L, in breast and ovarian cancer. Br. J. Cancer 2003,88,270–276. doi:10.1038/sj.bjc.6600674
    OpenUrlCrossRefPubMedWeb of Science
  47. 47.↵
    Yen, C.Y.; Huang, C.Y.; Hou, M.F.; Yang, Y.H.; Chang, C.H.; Huang, H.W.; Chen, C.H.; Chang, H.W. Evaluating the performance of fibronectin 1 (FN1), integrin α4β1 (ITGA4), syndecan-2 (SDC2), and glycoprotein CD44 as the potential biomarkers of oral squamous cell carcinoma (OSCC). Biomarkers 2013,18,63–72. doi:10.3109/1354750X.2012.737025
    OpenUrlCrossRef
  48. 48.↵
    Tomsic, J.; He, H.; Akagi, K.; Liyanarachchi, S.; Pan, Q.; Bertani, B.; Nagy, R.; Symer, D.E.; Blencowe, B.J.; de la Chapelle, A. A germline mutation in SRRM2, a splicing factor gene, is implicated in papillary thyroid carcinoma predisposition. Sci. Rep 2015,5,10566. doi:10.1038/srep10566
    OpenUrlCrossRef
  49. 49.↵
    Kim, M.J.; Choi, S.K.; Hong, S.H.; Eun, J.W.; Nam, S.W.; Han, J.W.; You, J.S. Oncogenic IL7R is downregulated by histone deacetylase inhibitor in esophageal squamous cell carcinoma via modulation of acetylated FOXO1. Int. J. Oncol 2018,53,395–403. doi:10.3892/ijo.2018.4392
    OpenUrlCrossRef
  50. 50.↵
    Lee, J.; Li, L.; Gretz, N.; Gebert, J.; Dihlmann, S. Absent in Melanoma 2 (AIM2) is an important mediator of interferon-dependent and -independent HLA-DRA and HLA-DRB gene expression in colorectal cancers. Oncogene 2012,31,1242–1253. doi:10.1038/onc.2011.320
    OpenUrlCrossRefPubMed
  51. 51.↵
    Liu, Y.; Kim, H.G.; Dong, E.; Dong, C.; Huang, M.; Liu, Y.; Liangpunsakul, S; Dong, X.C. Sesn3 deficiency promotes carcinogen-induced hepatocellular carcinoma via regulation of the hedgehog pathway. Biochim. Biophys. Acta. Mol. Basis. Dis 2019,1865,2685–2693. doi:10.1016/j.bbadis.2019.07.011
    OpenUrlCrossRef
  52. 52.↵
    Yang, Z.; Senninger, N.; Flammang, I.; Ye, Q.; Dhayat, S.A. Clinical impact of circulating LAPTM4B-35 in pancreatic ductal adenocarcinoma. J. Cancer. Res. Clin. Oncol 2019,145,1165–1178. doi:10.1007/s00432-019-02863-w
    OpenUrlCrossRef
  53. 53.↵
    Pandey, R.; Zhou, M.; Islam, S.; Chen, B.; Barker, N.K.; Langlais, P.; Srivastava, A.; Luo, M.; Cooke, L.S.; Weterings, E.; et al. Carcinoembryonic antigen cell adhesion molecule 6 (CEACAM6) in Pancreatic Ductal Adenocarcinoma (PDA): An integrative analysis of a novel therapeutic target. Sci. Rep 2019,9,18347. doi:10.1038/s41598-019-54545-9
    OpenUrlCrossRef
  54. 54.↵
    Neesse, A.; Wagner, M.; Ellenrieder, V.; Bachem, M.; Gress, T.M.; Buchholz, M. Pancreatic stellate cells potentiate proinvasive effects of SERPINE2 expression in pancreatic cancer xenograft tumors. Pancreatology 2007,7,380–385. doi:10.1159/000107400
    OpenUrlCrossRefPubMed
  55. 55.↵
    Kang, M.; Qin, W.; Buya, M.; Dong, X.; Zheng, W.; Lu, W.; Chen, J.; Guo, Q.; Wu, Y. VNN1, a potential biomarker for pancreatic cancer-associated new-onset diabetes, aggravates paraneoplastic islet dysfunction by increasing oxidative stress. Cancer. Lett 2016,373,241–250. doi:10.1016/j.canlet.2015.12.031
    OpenUrlCrossRef
  56. 56.↵
    Li, J.; Wu, H.; Li, W.; Yin, L.; Guo, S.; Xu, X.; Ouyang, Y.; Zhao, Z.; Liu, S.; Tian, Y.; et al. Downregulated miR-506 expression facilitates pancreatic cancer progression and chemoresistance via SPHK1/Akt/NF-κB signaling. Oncogene 2016,35,5501–5514. doi:10.1038/onc.2016.90
    OpenUrlCrossRefPubMed
  57. 57.↵
    Chen, X.L.; Xie, K.X.; Yang, Z.L.; Yuan, L.W. Expression of FXR and HRG and their clinicopathological significance in benign and malignant pancreatic lesions. Int. J. Clin. Exp. Pathol 2019,12,2111–2120.
    OpenUrl
  58. 58.↵
    Guo, J.; Lou, W.; Ji, Y.; Zhang, S. Effect of CCR7, CXCR4 and VEGF-C on the lymph node metastasis of human pancreatic ductal adenocarcinoma. Oncol. Lett 2013,5,1572–1578. doi:10.3892/ol.2013.1261
    OpenUrlCrossRef
  59. 59.↵
    Wan, Y.C.E.; Liu, J.; Zhu, L.; Kang, T.Z.E.; Zhu, X.; Lis, J.; Ishibashi, T.; Danko, C.G.; Wang, X.; Chan, K.M. The H2BG53D oncohistone directly upregulates ANXA3 transcription and enhances cell migration in pancreatic ductal adenocarcinoma. Signal. Transduct. Target. Ther 2020,5,106. doi:10.1038/s41392-020-00219-2
    OpenUrlCrossRef
  60. 60.↵
    Sato, Y.; Kobayashi, T.; Nishiumi, S.; Okada, A.; Fujita, T.; Sanuki, T.; Kobayashi, M.; Asahara, M.; Adachi, M.; Sakai, A.; et al. Prospective Study Using Plasma Apolipoprotein A2-Isoforms to Screen for High-Risk Status of Pancreatic Cancer. Cancers (Basel) 2020,12,e2625. doi:10.3390/cancers12092625
    OpenUrlCrossRef
  61. 61.↵
    Gumpper, K.; Dangel, A.W.; Pita-Grisanti, V.; Krishna, S.G.; Lara, L.F.; Mace, T.; Papachristou, G.I.; Conwell, D.L.; Hart, P.A.; Cruz-Monserrate, Z. Lipocalin-2 expression and function in pancreatic diseases. Pancreatology 2020,20,419–424. doi:10.1016/j.pan.2020.01.00262.D’
    OpenUrlCrossRef
  62. 62.↵
    Costa, Z.; Jones, K.; Azad, A.; van Stiphout, R.; Lim, S.Y.; Gomes, A.L.; Kinchesh, P.; Smart, S.C.; Gillies McKenna, W.; Buffa, F.M.; et al. Gemcitabine-Induced TIMP1 Attenuates Therapy Response and Promotes Tumor Growth and Liver Metastasis in Pancreatic Cancer. Cancer. Res. 2017,77,5952–5962. doi:10.1158/0008-5472.CAN-16-2833
    OpenUrlAbstract/FREE Full Text
  63. 63.↵
    Buscail, E.; Chauvet, A.; Quincy, P.; Degrandi, O.; Buscail, C.; Lamrissi, I.; Moranvillier, I.; Caumont, C.; Verdon, S.; Brisson, A.; et al. CD63-GPC1-Positive Exosomes Coupled with CA19-9 Offer Good Diagnostic Potential for Resectable Pancreatic Ductal Adenocarcinoma. Transl. Oncol 2019,12,1395–1403. doi:10.1016/j.tranon.2019.07.009
    OpenUrlCrossRef
  64. 64.↵
    Zhu, G.H.; Huang, C.; Qiu, Z.J.; Liu, J.; Zhang, Z.H.; Zhao, N.; Feng, Z.Z.; Lv, X.H. Expression and prognostic significance of CD151, c-Met, and integrin alpha3/alpha6 in pancreatic ductal adenocarcinoma. Dig. Dis. Sci 2011,56,1090–1098. doi:10.1007/s10620-010-1416-x
    OpenUrlCrossRefPubMed
  65. 65.↵
    Eguchi, D.; Ohuchida, K.; Kozono, S.; Ikenaga, N.; Shindo, K.; Cui, L.; Fujiwara, K.; Akagawa, S.; Ohtsuka, T.; Takahata, S.; et al. MAL2 expression predicts distant metastasis and short survival in pancreatic cancer. Surgery 2013,154,573–582. doi:10.1016/j.surg.2013.03.010
    OpenUrlCrossRefPubMedWeb of Science
  66. 66.↵
    Wang, Z.; Liu, T.; Xue, W.; Fang, Y.; Chen, X.; Xu, L.; Zhang, L.; Guan, K.; Pan, J.; Zheng, L.; et al. ARNTL2 promotes pancreatic ductal adenocarcinoma progression through TGF/BETA pathway and is regulated by miR-26a-5p. Cell. Death. Dis 2020,11,692. doi:10.1038/s41419-020-02839-6
    OpenUrlCrossRef
  67. 67.↵
    Yuan, J.; Rozengurt, E. PKD, PKD2, and p38 MAPK mediate Hsp27 serine-82 phosphorylation induced by neurotensin in pancreatic cancer PANC-1 cells. J. Cell. Biochem 2008,103,648–662. doi:10.1002/jcb.21439
    OpenUrlCrossRefPubMedWeb of Science
  68. 68.↵
    Schild, C.; Wirth, M.; Reichert, M.; Schmid, R.M.; Saur, D.; Schneider, G.; PI3K signaling maintains c-myc expression to regulate transcription of E2F1 in pancreatic cancer cells. Mol. Carcinog 2009,48,1149–1158. doi:10.1002/mc.2056
    OpenUrlCrossRefPubMedWeb of Science
  69. 69.↵
    Chen, Y.; Peng, S.; Cen, H.; Lin, Y.; Huang, C.; Chen, Y.; Shan, H.; Su, Y.; Zeng, L. MicroRNA hsa-miR-623 directly suppresses MMP1 and attenuates IL-8-induced metastasis in pancreatic cancer. Int. J. Oncol 2019,55,142–156. doi:10.3892/ijo.2019.4803
    OpenUrlCrossRef
  70. 70.↵
    Wang, L.; Zhao, X.Y.; Zhu, J.S.; Chen, N.W.; Fan, H.N.; Yang, W.; Guo, J.H. CCR7 regulates ANO6 to promote migration of pancreatic ductal adenocarcinoma cells via the ERK signaling pathway. Oncol. Lett 2018,16,2599–2605. doi:10.3892/ol.2018.8962
    OpenUrlCrossRef
  71. 71.↵
    Mazur, P.K.; Einwächter, H.; Lee, M.; Sipos, B.; Nakhai, H.; Rad, R.; Zimber-Strobl, U.; Strobl, L.J.; Radtke, F.; Klöppel, G.; et al. Notch2 is required for progression of pancreatic intraepithelial neoplasia and development of pancreatic ductal adenocarcinoma. Proc. Natl. Acad. Sci. U. S. A. 2010,107,13438–13443. doi:10.1073/pnas.1002423107
    OpenUrlAbstract/FREE Full Text
  72. 72.↵
    Bian, B.; Fanale, D.; Dusetti, N.; Roque, J.; Pastor, S.; Chretien, A.S.; Incorvaia, L.; Russo, A.; Olive, D.; Iovanna, J.; et al. Prognostic significance of circulating PD-1, PD-L1, pan-BTN3As, BTN3A1 and BTLA in patients with pancreatic adenocarcinoma. Oncoimmunology 2019,8,e1561120. doi:10.1080/2162402X.2018.1561120
    OpenUrlCrossRefPubMed
  73. 73.↵
    Ryschich, E.; Huszty, G.; Knaebel, H.P.; Hartel, M.; Büchler, M.W.; Schmidt, J. Transferrin receptor is a marker of malignant phenotype in human pancreatic cancer and in neuroendocrine carcinoma of the pancreas. Eur. J. Cancer 2004,40,1418–1422. doi:10.1016/j.ejca.2004.01.036
    OpenUrlCrossRefPubMedWeb of Science
  74. 74.↵
    Sonntag, K.; Hashimoto, H.; Eyrich, M.; Menzel, M.; Schubach, M.; Döcker, D.; Battke, F.; Courage, C.; Lambertz, H.; Handgretinger, R.; et al. Immune monitoring and TCR sequencing of CD4 T cells in a long term responsive patient with metastasized pancreatic ductal carcinoma treated with individualized, neoepitope-derived multipeptide vaccines: a case report. J. Transl. Med 2018,16,23. doi:10.1186/s12967-018-1382-1
    OpenUrlCrossRef
  75. 75.↵
    Hutchings, D.; Jiang, Z.; Skaro, M.; Weiss, M.J.; Wolfgang, C.L.; Makary, M.A.; He, J.; Cameron, J.L.; Zheng, L.; Klimstra, D.S.; et al. Histomorphology of pancreatic cancer in patients with inherited ATM serine/threonine kinase pathogenic variants. Mod. Pathol 2019,32,1806–1813. doi:10.1038/s41379-019-0317-6
    OpenUrlCrossRefPubMed
  76. 76.↵
    Singhi, A.D.; Lilo, M.; Hruban, R.H.; Cressman, K.L.; Fuhrer, K.; Seethala, R.R. Overexpression of lymphoid enhancer-binding factor 1 (LEF1) in solid-pseudopapillary neoplasms of the pancreas. Mod. Pathol 2014,27,1355–1363. doi:10.1038/modpathol.2014.40
    OpenUrlCrossRef
  77. 77.↵
    Zhu, Y.; Knolhoff, B.L.; Meyer, M.A.; Nywening, T.M.; West, B.L.; Luo, J.; Wang-Gillam, A.; Goedegebuure, S.P.; Linehan, D.C.; DeNardo, D.G. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer. Res 2014,74,5057–5069. doi:10.1158/0008-5472.CAN-13-3723
    OpenUrlAbstract/FREE Full Text
  78. 78.↵
    Dumartin, L.; Whiteman, H.J.; Weeks, M.E.; Hariharan, D.; Dmitrovic, B.; Iacobuzio-Donahue, C.A.; Brentnall, T.A.; Bronner, M.P.; Feakins, R.M.; Timms, J.F.; et al. AGR2 is a novel surface antigen that promotes the dissemination of pancreatic cancer cells through regulation of cathepsins B and D. Cancer. Res 2011,71,7091–7102. doi:10.1158/0008-5472.CAN-11-1367
    OpenUrlAbstract/FREE Full Text
  79. 79.↵
    Wang, C.A.; Chang, I.H.; Hou, P.C.; Tai, Y.J.; Li, W.N.; Hsu, P.L.; Wu, S.R.; Chiu, W.T.; Li, C.F.; Shan, Y.S.; et al. DUSP2 regulates extracellular vesicle-VEGF-C secretion and pancreatic cancer early dissemination. J. Extracell. Vesicles 2020,9,1746529. doi:10.1080/20013078.2020.1746529
    OpenUrlCrossRef
  80. 80.↵
    Yoon, K.; Lee, S.O.; Cho, S.D.; Kim, K.; Khan, S.; Safe, S. Activation of nuclear TR3 (NR4A1) by a diindolylmethane analog induces apoptosis and proapoptotic genes in pancreatic cancer cells and tumors. Carcinogenesis 2011,32,836–842. doi:10.1093/carcin/bgr040
    OpenUrlCrossRefPubMedWeb of Science
  81. 81.↵
    Rodriguez-Aguayo, C.; Bayraktar, E.; Ivan, C.; Aslan, B.; Mai, J.; He, G.; Mangala, L.S.; Jiang, D.; Nagaraja, A.S.; Ozpolat, B.; et al. PTGER3 induces ovary tumorigenesis and confers resistance to cisplatin therapy through up-regulation Ras-MAPK/Erk-ETS1-ELK1/CFTR1 axis. EBioMedicine 2019,40,290–304. doi:10.1016/j.ebiom.2018.11.045
    OpenUrlCrossRef
  82. 82.↵
    Chang, H.; Zhang, X.; Li, B.; Meng, X. MAGI2-AS3 suppresses MYC signaling to inhibit cell proliferation and migration in ovarian cancer through targeting miR-525-5p/MXD1 axis. Cancer. Med 2020,9,6377–6386. doi:10.1002/cam4.3126
    OpenUrlCrossRef
  83. 83.↵
    Hoagland, L.F.; Campa, M.J.; Gottlin, E.B.; Herndon, J.E.; Patz, E.F. Haptoglobin and posttranslational glycan-modified derivatives as serum biomarkers for the diagnosis of nonsmall cell lung cancer. Cancer 2007,110,2260–2268. doi:10.1002/cncr.23049
    OpenUrlCrossRefPubMed
  84. 84.↵
    Wang, M.; Zhang, G.; Zhang, Y.; Cui, X.; Wang, S.; Gao, S.; Wang, Y.; Liu, Y.; Bae, J.H.; Yang, W.H.; et al. Fibrinogen Alpha Chain Knockout Promotes Tumor Growth and Metastasis through Integrin-AKT Signaling Pathway in Lung Cancer. Mol. Cancer. Res 2020,18,943–954. doi:10.1158/1541-7786.MCR-19-1033
    OpenUrlAbstract/FREE Full Text
  85. 85.↵
    Repetto, O.; Maiero, S.; Magris, R.; Miolo, G.; Cozzi, M.R.; Steffan, A.; Canzonieri, V.; Cannizzaro, R.; De Re, V. Quantitative Proteomic Approach Targeted to Fibrinogen β Chain in Tissue Gastric Carcinoma. Int. J. Mol. Sci 2018,19,759. doi:10.3390/ijms19030759
    OpenUrlCrossRef
  86. 86.↵
    Bazhan, D.; Khaniani, M.S. Supplementation with omega fatty acids increases the mRNA expression level of PLA2G4A in patients with gastric cancer. J. Gastrointest. Oncol 2018,9,1176–1183. doi:10.21037/jgo.2018.08.12
    OpenUrlCrossRef
  87. 87.↵
    Duan, S.; Gong, B.; Wang, P.; Huang, H.; Luo, L.; Liu, F. Novel prognostic biomarkers of gastric cancer based on gene expression microarray: COL12A1, GSTA3, FGA and FGG. Mol. Med. Rep 2018,18,3727–3736. doi:10.3892/mmr.2018.9368
    OpenUrlCrossRef
  88. 88.↵
    Lee, S.W.; Chen, T.J.; Lin, L.C.; Li, C.F.; Chen, L.T.; Hsing, C.H.; Hsu, H.P.; Tsai, C.J.; Huang, H.Y.; Shiue, Y.L. Overexpression of thymidylate synthetase confers an independent prognostic indicator in nasopharyngeal carcinoma. Exp. Mol. Pathol 2013,95,83–90. doi:10.1016/j.yexmp.2013.05.006
    OpenUrlCrossRef
  89. 89.↵
    Shibata, D.; Mori, Y.; Cai, K.; Zhang, L.; Yin, J.; Elahi, A.; Hamelin, R.; Wong, Y.F.; Lo, W.K.; Chung T.K.; et al. RAB32 hypermethylation and microsatellite instability in gastric and endometrial adenocarcinomas. Int. J. Cancer 2006,119,801–806. doi:10.1002/ijc.21912
    OpenUrlCrossRefPubMed
  90. 90.↵
    Zhao, X.; Feng, H.; Wang, Y.; Wu, Y.; Guo, Q.; Feng, Y.; Ma, M.; Guo, W.; Song, X.; Zhang, Y.; et al. Septin4 promotes cell death in human colon cancer cells by interacting with BAX. Int. J. Biol. Sci 2020,16,1917–1928. doi:10.7150/ijbs.44429
    OpenUrlCrossRef
  91. 91.↵
    Zhang, J.; Zhang, J.; Xu, S.; Zhang, X.; Wang, P.; Wu, H.; Xia, B.; Zhang, G.; Lei, B.; Wan, L.; et al. Hypoxia-Induced TPM2 Methylation is Associated with Chemoresistance and Poor Prognosis in Breast Cancer. Cell. Physiol. Biochem 2018,45,692–705. doi:10.1159/000487162
    OpenUrlCrossRef
  92. 92.↵
    Feng, H.; Liu, X. Interaction between ACOT7 and LncRNA NMRAL2P via Methylation Regulates Gastric Cancer Progression. Yonsei. Med. J 2020,61,471–481. doi:10.3349/ymj.2020.61.6.471
    OpenUrlCrossRef
  93. 93.↵
    Suzuki, E.; Imoto, I.; Pimkhaokham, A.; Nakagawa, T.; Kamata, N.; Kozaki, K.I.; Amagasa, T.; Inazawa, J. PRTFDC1, a possible tumor-suppressor gene, is frequently silenced in oral squamous-cell carcinomas by aberrant promoter hypermethylation. Oncogene 2007,26,7921–7932. doi:10.1038/sj.onc.1210589
    OpenUrlCrossRefPubMedWeb of Science
  94. 94.↵
    Sakamoto, H.; Friel, A.M.; Wood, A.W.; Guo, L.; Ilic, A.; Seiden, M.V.; Chung, D.C.; Lynch, M.P.; Serikawa, T.; Munro, E.; et al. Mechanisms of Cables 1 gene inactivation in human ovarian cancer development. Cancer. Biol. Ther 2008,7,180–188. doi:10.4161/cbt.7.2.5253
    OpenUrlCrossRefPubMedWeb of Science
  95. 95.↵
    Callahan, M.J.; Nagymanyoki, Z.; Bonome, T.; Johnson, M.E.; Litkouhi, B.; Sullivan, E.H.; Hirsch, M.S.; Matulonis, U.A.; Liu, J.; Birrer, M.J.; et al. Increased HLA-DMB expression in the tumor epithelium is associated with increased CTL infiltration and improved prognosis in advanced-stage serous ovarian cancer. Clin. Cancer. Res 2008,14,7667–7673. doi:10.1158/1078-0432.CCR-08-0479
    OpenUrlAbstract/FREE Full Text
  96. 96.↵
    Wu, Y.; Han, J.; Vladimirovna, K.E.; Zhang, S.; Lv, W.; Zhang, Y.; Jamaspishvili, E.; Sun, J.; Fang, Q.; Meng, J.; et al. Upregulation Of Protein Tyrosine Phosphatase Receptor Type C Associates To The Combination Of Hashimoto’s Thyroiditis And Papillary Thyroid Carcinoma And Is Predictive Of A Poor Prognosis. Onco. Targets. Ther 2019,12,8479–8489. doi:10.2147/OTT.S226426
    OpenUrlCrossRef
  97. 97.↵
    Moreno-Manuel, A.; Jantus-Lewintre, E.; Simões, I.; Aranda, F.; Calabuig-Fariñas, S.; Carreras, E.; Zúñiga, S.; Saenger, Y.; Rosell, R.; Camps, C.; et al. CD5 and CD6 as immunoregulatory biomarkers in non-small cell lung cancer. Transl. Lung. Cancer. Res 2020,9,1074–1083. doi:10.21037/tlcr-19-445
    OpenUrlCrossRef
  98. 98.↵
    Wright, C.M.; Savarimuthu Francis, S.M., Tan, M.E.; Martins, M.U.; Winterford, C.; Davidson, M.R.; Duhig, E.E.; Clarke, B.E.; Hayward, N.K.; et al. MS4A1 dysregulation in asbestos-related lung squamous cell carcinoma is due to CD20 stromal lymphocyte expression. PLoS. One 2012,7,e34943. doi:10.1371/journal.pone.0034943
    OpenUrlCrossRefPubMed
  99. 99.↵
    Tuscano, J.M.; Kato, J.; Pearson, D.; Xiong, C.; Newell, L.; Ma, Y.; Gandara, D.R.; O’Donnell, R.T. CD22 antigen is broadly expressed on lung cancer cells and is a target for antibody-based therapy. Cancer. Res 2012,72,5556–5565. doi:10.1158/0008-5472.CAN-12-0173
    OpenUrlAbstract/FREE Full Text
  100. 100.↵
    Nielsen, J.S.; Sahota, R.A.; Milne, K.; Kost, S.E.; Nesslinger, N.J.; Watson, P.H.; Nelson, B.H. CD20+ tumor-infiltrating lymphocytes have an atypical CD27-memory phenotype and together with CD8+ T cells promote favorable prognosis in ovarian cancer. Clin. Cancer. Res 2012,18,3281–3292. doi:10.1158/1078-0432.CCR-12-0234
    OpenUrlAbstract/FREE Full Text
  101. 101.↵
    Gai, X.; Tu, K.; Lu, Z.; Zheng, X. MRC2 expression correlates with TGFβ1 and survival in hepatocellular carcinoma. Int. J. Mol. Sci 2014,15,15011–15025. doi:10.3390/ijms150915011
    OpenUrlCrossRef
  102. 102.↵
    Mathew, S.O.; Chaudhary, P.; Powers, S.B.; Vishwanatha, J.K.; Mathew, P.A. Overexpression of LLT1 (OCIL, CLEC2D) on prostate cancer cells inhibits NK cell-mediated killing through LLT1-NKRP1A (CD161) interaction. Oncotarget 2016,7,68650–68661. doi:10.18632/oncotarget.11896
    OpenUrlCrossRef
  103. 103.↵
    Joung, E.K.; Kim, J.; Yoon, N.; Maeng, L.S.; Kim, J.H.; Park, S.; Kang, K.; Kim, J.S.; Ahn, Y.H.; Ko, Y.H.; et al. Expression of EEF1A1 Is Associated with Prognosis of Patients with Colon Adenocarcinoma. J. Clin. Med 2019,8,1903. doi:10.3390/jcm8111903
    OpenUrlCrossRef
  104. 104.↵
    Yang, D.D.; Chen, Z.H.; Wang, D.S.; Yu, H.E.; Lu, J.H.; Xu, R.H.; Zeng, Z.L. Prognostic value of the serum apolipoprotein B to apolipoprotein A-I ratio in metastatic colorectal cancer patients. J. Cancer 2020,11,1063–1074. doi:10.7150/jca.3565
    OpenUrlCrossRef
  105. 105.↵
    Jung, J.H.; Taniguchi, K.; Lee, H.M.; Lee, M.Y.; Bandu, R.; Komura, K.; Lee, K.Y.; Akao, Y.; Kim, K.P. Comparative lipidomics of 5-Fluorouracil-sensitive and -resistant colorectal cancer cells reveals altered sphingomyelin and ceramide controlled by acid sphingomyelinase (SMPD1). Sci. Rep 2020,10,6124. doi:10.1038/s41598-020-62823-0
    OpenUrlCrossRef
  106. 106.↵
    Liu, M.; Qu, Y.; Teng, X.; Xing, Y.; Li, D.; Li, C.; Cai, L.; PADI4□mediated epithelial□mesenchymal transition in lung cancer cells. Mol. Med. Rep 2019,19,3087–3094. doi:10.3892/mmr.2019.9968
    OpenUrlCrossRef
  107. 107.↵
    Yang, Y.C.; Chien, M.H.; Lai, T.C.; Su, C.Y.; Jan, Y.H.; Hsiao, M.; Chen, C.L. Monoamine Oxidase B Expression Correlates with a Poor Prognosis in Colorectal Cancer Patients and Is Significantly Associated with Epithelial-to-Mesenchymal Transition-Related Gene Signatures. Int. J. Mol. Sci 2020,21,2813. doi:10.3390/ijms21082813
    OpenUrlCrossRef
  108. 108.↵
    Song, Y.; Liu, G.; Liu, S.; Chen, R.; Wang, N.; Liu, Z.;; Zhang, X.; Xiao, Z.; Liu, L. Helicobacter pylori upregulates TRPC6 via Wnt/β-catenin signaling to promote gastric cancer migration and invasion. Onco. Targets. Ther 2019,12,5269–5279. doi:10.2147/OTT.S201025
    OpenUrlCrossRef
  109. 109.↵
    Seachrist, D.D.; Hannigan, M.M.; Ingles, N.N.; Webb, B.M.; Weber- Bonk, K.L., Yu, P.; Bebek, G.; Singh, S.; Sizemore, S.T.; Varadan, V.; et al. The transcriptional repressor BCL11A promotes breast cancer metastasis. J. Biol. Chem 2020,295,11707–11719. doi:10.1074/jbc.RA120.014018
    OpenUrlAbstract/FREE Full Text
  110. 110.↵
    Zhu, Z.; Zhang, X.; Guo, H.; Fu, L.; Pan, G.; Sun, Y. CXCL13-CXCR5 axis promotes the growth and invasion of colon cancer cells via PI3K/AKT pathway. Mol. Cell. Biochem 2015,400,287–295. doi:10.1007/s11010-014-2285-y
    OpenUrlCrossRefPubMed
  111. 111.↵
    Wu, B.; Chen, M.; Gao, M.; Cong, Y.; Jiang, L.; Wei, J.; Huang, J. Down-regulation of lncTCF7 inhibits cell migration and invasion in colorectal cancer via inhibiting TCF7 expression. Hum. Cell 2019,32,31–40. doi:10.1007/s13577-018-0217-y
    OpenUrlCrossRef
  112. 112.↵
    Wang, S.M.; Tie, J.; Wang, W.L.; Hu, S.J.; Y+in, J.P.; Yi, X.F.; Tian, Z.H.; Zhang, X.Y.; Li, M.B.; Li, Z.S.; et al. POU2F2-oriented network promotes human gastric cancer metastasis. Gut 2016,65,1427–1438. doi:10.1136/gutjnl-2014-308932
    OpenUrlAbstract/FREE Full Text
  113. 113.↵
    Yi, T.; Zhou, X.; Sang, K.; Huang, X.; Zhou, J.; Ge, L. Activation of lncRNA lnc-SLC4A1-1 induced by H3K27 acetylation promotes the development of breast cancer via activating CXCL8 and NF-kB pathway. Artif. Cells. Nanomed. Biotechnol 2019,47,3765–3773. doi:10.1080/21691401.2019.1664559
    OpenUrlCrossRef
  114. 114.↵
    Lan, Y.; Han, J.; Wang, Y.; Wang, J.; Yang, G.; Li, K.; Song, R.; Zheng, T.; Liang, Y.; Pan, S.; et al. STK17B promotes carcinogenesis and metastasis via AKT/GSK-3β/Snail signaling in hepatocellular carcinoma. Cell. Death. Dis 2018,9,236. doi:10.1038/s41419-018-0262-1
    OpenUrlCrossRef
  115. 115.↵
    Appert-Collin, A.; Bennasroune, A.; Jeannesson, P.; Terryn, C.; Fuhrmann, G.; Morjani, H.; Dedieu, S. Role of LRP-1 in cancer cell migration in 3-dimensional collagen matrix. Cell. Adh. Migr 2017,11,316–326. doi:10.1080/19336918.2016.1215788
    OpenUrlCrossRef
  116. 116.↵
    Kairouz, R.; Parmar, J.; Lyons, R.J.; Swarbrick, A.; Musgrove, E.A.; Daly, R.J. Hormonal regulation of the Grb14 signal modulator and its role in cell cycle progression of MCF-7 human breast cancer cells. J. Cell. Physiol 2005,203,85–93. doi:10.1002/jcp.20199
    OpenUrlCrossRefPubMed
  117. 117.↵
    Diez-Bello, R.; Jardin, I.; Lopez, J.J.; El Haouari, M.; Ortega-Vidal, J.; Altarejos, J.; Salido, G.M.; Salido, S.; Rosado, J.A. (-)□Oleocanthal inhibits proliferation and migration by modulating Ca2+ entry through TRPC6 in breast cancer cells. Biochim. Biophys. Acta. Mol. Cell. Res. 2019,1866,474–485. doi:10.1016/j.bbamcr.2018.10.010
    OpenUrlCrossRef
  118. 118.↵
    Xue, M.; Tao, W.; Yu, S.; Yan, Z.; Peng, Q.; Jiang, F.; Gao, X. lncRNA ZFPM2-AS1 promotes proliferation via miR-18b-5p/VMA21 axis in lung adenocarcinoma. J. Cell. Biochem 2020,121,313–321. doi:10.1002/jcb.29176
    OpenUrlCrossRef
  119. 119.↵
    Abo-Elfadl, M.T.; Gamal-Eldeen, A.M.; Ismail, M.F.; Shahin, N.N. Silencing of the cytokine receptor TNFRSF13B: A new therapeutic target for triple-negative breast cancer. Cytokine 2020,125,154790. doi:10.1016/j.cyto.2019.154790
    OpenUrlCrossRef
  120. 120.↵
    Li, J.; Xu, X.; Wei, C.; Liu, L.; Wang, T. Long noncoding RNA NORAD regulates lung cancer cell proliferation, apoptosis, migration, and invasion by the miR-30a-5p/ADAM19 axis. Int. J. Clin. Exp. Pathol 2020,13,1–13.
    OpenUrl
  121. 121.↵
    Zhao, J.; Cheng, L. Long non-coding RNA CCAT1/miR-148a axis promotes osteosarcoma proliferation and migration through regulating PIK3IP1. Acta. Biochim. Biophys. Sin (Shanghai) 2017,49,503–512. doi:10.1093/abbs/gmx041
    OpenUrlCrossRef
  122. 122.↵
    Leite, F.A.; Lira, R.C.; Fedatto, P.F.; Antonini, S.R.; Martinelli, C.E.; de Castro, M.; Neder, L.; Ramalho, L.N.; Tucci, S.; Mastelaro, M.J.; et al. Low expression of HLA-DRA, HLA-DPA1, and HLA-DPB1 is associated with poor prognosis in pediatric adrenocortical tumors (ACT). Pediatr. Blood. Cancer 2014,61,1940–1948. doi:10.1002/pbc.25118
    OpenUrlCrossRef
  123. 123.↵
    Feng, Y.; Guo, C.; Wang, H.; Zhao, L.; Wang, W.; Wang, T.; Feng, Y.; Yuan, K.; Huang, G. Fibrinogen-Like Protein 2 (FGL2) is a Novel Biomarker for Clinical Prediction of Human Breast Cancer. Med. Sci. Monit 2020,26,e923531. doi:10.12659/MSM.923531
    OpenUrlCrossRef
  124. 124.↵
    Wang, S.; Xu, L.; Che, X.; Li, C.; Xu, L.; Hou, K.; Fan, Y.; Wen, T.; Qu, X.; Liu, Y. E3 ubiquitin ligases Cbl-b and c-Cbl downregulate PD-L1 in EGFR wild-type non-small cell lung cancer. FEBS. Lett 2018,592,621–630. doi:10.1002/1873-3468.12985
    OpenUrlCrossRef
  125. 125.↵
    Zhong, X.P.; Kan, A.; Ling, Y.H.; Lu, L.H.; Mei, J.; Wei, W.; Li, S.H.; Guo, R.P. NCKAP1 improves patient outcome and inhibits cell growth by enhancing Rb1/p53 activation in hepatocellular carcinoma. Cell. Death. Dis 2019,10,369.. doi:10.1038/s41419-019-1603-4
    OpenUrlCrossRef
  126. 126.↵
    Yokoyama-Mashima, S.; Yogosawa, S.; Kanegae, Y.; Hirooka, S.; Yoshida, S.; Horiuchi, T.; Ohashi, T.; Yanaga, K.; Saruta, M.; Oikawa, T.; et al. Forced expression of DYRK2 exerts anti-tumor effects via apoptotic induction in liver cancer. Cancer. Lett 2019,451,100–109. doi:10.1016/j.canlet.2019.02.046
    OpenUrlCrossRef
  127. 127.↵
    Guo, H.; Zhang, B.; Nairn, A.V.; Nagy, T.; Moremen, K.W.; Buckhaults, P.; Pierce, M. O-Linked N-Acetylglucosamine (O-GlcNAc) Expression Levels Epigenetically Regulate Colon Cancer Tumorigenesis by Affecting the Cancer Stem Cell Compartment via Modulating Expression of Transcriptional Factor MYBL1. J. Biol. Chem 2017,292,4123–4137. doi:10.1074/jbc.M116.763201
    OpenUrlAbstract/FREE Full Text
  128. 128.↵
    Lawson, J.; Dickman, C.; MacLellan, S.; Towle, R.; Jabalee, J.; Lam, S.; Garnis, C. Selective secretion of microRNAs from lung cancer cells via extracellular vesicles promotes CAMK1D-mediated tube formation in endothelial cells. Oncotarget 2017,8,83913–83924. doi:10.18632/oncotarget.19996
    OpenUrlCrossRefPubMed
  129. 129.↵
    Wang, X.; Ye, M.; Wu., M., Fang, H.; Xiao, B.; Xie, L.; Zhu, X. RNF213 suppresses carcinogenesis in glioblastoma by affecting MAPK/JNK signaling pathway. Clin. Transl. Oncol 2020,22,1506–1516. doi:10.1007/s12094-020-02286-x
    OpenUrlCrossRef
  130. 130.↵
    Taheri, M.; Omrani, M.D.; Noroozi, R.; Ghafouri-Fard, S.; Sayad, A. Retinoic acid-related orphan receptor alpha (RORA) variants and risk of breast cancer. Breast. Dis 2017,37,21–25. doi:10.3233/BD-160248
    OpenUrlCrossRef
  131. 131.↵
    Hoyo, C.; Murphy, S.K.; Schildkraut, J.M.; Vidal, A.C.; Skaar, D.;; Millikan, R.C.; Galanko, J.; Sandler R.S.; Jirtle R.; Keku T. IGF2R genetic variants, circulating IGF2 concentrations and colon cancer risk in African Americans and Whites. Dis. Markers 2012,32,133–141. doi:10.3233/DMA-2011-0865
    OpenUrlCrossRefPubMedWeb of Science
  132. 132.↵
    Bai, F.; Xiao, K. Prediction of gastric cancer risk: association between ZBTB20 genetic variance and gastric cancer risk in Chinese Han population. Biosci. Rep 2020,40,BSR20202102. doi:10.1042/BSR20202102
    OpenUrlCrossRef
  133. 133.↵
    Hope, C.; Emmerich, P.B; Papadas, A.; Pagenkopf, A.; Matkowskyj, K.A.; Van De Hey, D.R.; Payne, S.N.; Clipson, L.; Callander, N.S.; Hematti, P.; et al. Versican-Derived Matrikines Regulate Batf3-Dendritic Cell Differentiation and Promote T Cell Infiltration in Colorectal Cancer. J. Immunol 2017,199,1933–1941. doi:10.4049/jimmunol.1700529
    OpenUrlAbstract/FREE Full Text
  134. 134.↵
    Bai, X.; Wang, W.; Zhao, P.; Wen, J.; Guo, X.; Shen, T.; Shen, J.; Yang, X. LncRNA CRNDE acts as an oncogene in cervical cancer through sponging miR-183 to regulate CCNB1 expression. Carcinogenesis 2020,41,111–121. doi:10.1093/carcin/bgz166
    OpenUrlCrossRef
  135. 135.↵
    Zienert, E.; Eke, I.; Aust, D.; Cordes, N. LIM-only protein FHL2 critically determines survival and radioresistance of pancreatic cancer cells. Cancer. Lett 2015,364,17–24. doi:10.1016/j.canlet.2015.04.019
    OpenUrlCrossRef
  136. 136.↵
    Yang, J.; Chen, Z.; Liu, N.; Chen, Y. Ribosomal protein L10 in mitochondria serves as a regulator for ROS level in pancreatic cancer cells. Redox Biol 2018,19,158–165. doi:10.1016/j.redox.2018.08.016
    OpenUrlCrossRef
  137. 137.↵
    Xie, F.; Huang, Q.; Wang, C.; Chen, S.; Liu, C.; Lin, X.; Lv, X.; Wang, C. Downregulation of long noncoding RNA SNHG14 suppresses cell proliferation and invasion by regulating EZH2 in pancreatic ductal adenocarcinoma (PDAC). Cancer. Biomark 2020,27,357–364. doi:10.3233/CBM-190908
    OpenUrlCrossRef
  138. 138.↵
    Li, S.S.; Jiang, W.L.; Xiao, W.Q.; Li, K.; Zhang, Y.F.; Guo, X.Y.; Dai, Y.Q.; Zhao, Q.Y.; Jiang, M.J.; Lu, Z.J.; et al. KMT2D deficiency enhances the anti-cancer activity of L48H37 in pancreatic ductal adenocarcinoma. World. J. Gastrointest. Oncol 2019,11,599–621. doi:10.4251/wjgo.v11.i8.599
    OpenUrlCrossRef
  139. 139.↵
    Zhu, G.; Zhou, L.; Liu, H.; Shan, Y.; Zhang, X. MicroRNA-224 Promotes Pancreatic Cancer Cell Proliferation and Migration by Targeting the TXNIP-Mediated HIF1α Pathway. Cell. Physiol. Biochem 2018,48,1735–1746. doi:10.1159/000492309
    OpenUrlCrossRef
  140. 140.↵
    Zhang, K.D.; Hu, B.; Cen, G.; Yang, Y.H.; Chen, W.W.; Guo, Z.Y.; Wang, X.F.; Zhao, Q.; Qiu, Z.J. MiR-301a transcriptionally activated by HIF-2α promotes hypoxia-induced epithelial-mesenchymal transition by targeting TP63 in pancreatic cancer. World. J. Gastroenterol 2020,26,2349–2373. doi:10.3748/wjg.v26.i19.2349
    OpenUrlCrossRef
  141. 141.↵
    Wuebben, E.L.; Wilder, P.J.; Cox, J.L.; Grunkemeyer, J.A.; Caffrey, T.; Hollingsworth, M.A.; Rizzino, A. SOX2 functions as a molecular rheostat to control the growth, tumorigenicity and drug responses of pancreatic ductal adenocarcinoma cells. Oncotarget 2016,7,34890–34906. doi:10.18632/oncotarget.8994
    OpenUrlCrossRef
  142. 142.↵
    Muthalagu, N.; Monteverde, T.; Raffo-Iraolagoitia, X.; Wiesheu, R.; Whyte, D.; Hedley, A.; Laing, S.; Kruspig, B.; Upstill-Goddard R.; Shaw R.; et al. Repression of the Type I Interferon Pathway Underlies MYC- and KRAS-Dependent Evasion of NK and B Cells in Pancreatic Ductal Adenocarcinoma. Cancer. Discov 2020,10,872–887. doi:10.1158/2159-8290.CD-19-0620
    OpenUrlAbstract/FREE Full Text
  143. 143.↵
    Wang, Z.; Chen, Y.; Lin, Y.; Wang, X.; Cui, X.; Zhang, Z.; Xian, G.; Qin, C. Novel crosstalk between KLF4 and ZEB1 regulates gemcitabine resistance in pancreatic ductal adenocarcinoma. Int. J. Oncol 2017,51,1239–1248. doi:10.3892/ijo.2017.4099
    OpenUrlCrossRef
  144. 144.↵
    Hu, L.; Fang, L.; Zhang, Z.P.; Yan, Z.L. TPM1 is a Novel Predictive Biomarker for Gastric Cancer Diagnosis and Prognosis. Clin Lab. 2020,66, 10.7754/Clin.Lab.2019.190235. doi:10.7754/Clin.Lab.2019.190235
    OpenUrlCrossRef
  145. 145.↵
    Chen, H.; Fan, Y.; Xu, W.; Chen, J.; Meng, Y.; Fang, D.; Wang, J. Exploration of miR-1202 and miR-196a in human endometrial cancer based on high throughout gene screening analysis. Oncol. Rep 2017,37,3493–3501. doi:10.3892/or.2017.5596
    OpenUrlCrossRef
Back to top
PreviousNext
Posted December 23, 2020.
Download PDF
Data/Code
Email

Thank you for your interest in spreading the word about medRxiv.

NOTE: Your email address is requested solely to identify you as the sender of this article.

Enter multiple addresses on separate lines or separate them with commas.
Identification and Interaction Analysis of Molecular Markers in Pancreatic Ductal Adenocarcinoma by Integrated Bioinformatics Analysis and Molecular Docking Experiments
(Your Name) has forwarded a page to you from medRxiv
(Your Name) thought you would like to see this page from the medRxiv website.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Identification and Interaction Analysis of Molecular Markers in Pancreatic Ductal Adenocarcinoma by Integrated Bioinformatics Analysis and Molecular Docking Experiments
Basavaraj Vastrad, Chanabasayya Vastrad, Anandkumar Tengli
medRxiv 2020.12.20.20248601; doi: https://doi.org/10.1101/2020.12.20.20248601
Twitter logo Facebook logo LinkedIn logo Mendeley logo
Citation Tools
Identification and Interaction Analysis of Molecular Markers in Pancreatic Ductal Adenocarcinoma by Integrated Bioinformatics Analysis and Molecular Docking Experiments
Basavaraj Vastrad, Chanabasayya Vastrad, Anandkumar Tengli
medRxiv 2020.12.20.20248601; doi: https://doi.org/10.1101/2020.12.20.20248601

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Subject Area

  • Genetic and Genomic Medicine
Subject Areas
All Articles
  • Addiction Medicine (349)
  • Allergy and Immunology (668)
  • Allergy and Immunology (668)
  • Anesthesia (181)
  • Cardiovascular Medicine (2648)
  • Dentistry and Oral Medicine (316)
  • Dermatology (223)
  • Emergency Medicine (399)
  • Endocrinology (including Diabetes Mellitus and Metabolic Disease) (942)
  • Epidemiology (12228)
  • Forensic Medicine (10)
  • Gastroenterology (759)
  • Genetic and Genomic Medicine (4103)
  • Geriatric Medicine (387)
  • Health Economics (680)
  • Health Informatics (2657)
  • Health Policy (1005)
  • Health Systems and Quality Improvement (985)
  • Hematology (363)
  • HIV/AIDS (851)
  • Infectious Diseases (except HIV/AIDS) (13695)
  • Intensive Care and Critical Care Medicine (797)
  • Medical Education (399)
  • Medical Ethics (109)
  • Nephrology (436)
  • Neurology (3882)
  • Nursing (209)
  • Nutrition (577)
  • Obstetrics and Gynecology (739)
  • Occupational and Environmental Health (695)
  • Oncology (2030)
  • Ophthalmology (585)
  • Orthopedics (240)
  • Otolaryngology (306)
  • Pain Medicine (250)
  • Palliative Medicine (75)
  • Pathology (473)
  • Pediatrics (1115)
  • Pharmacology and Therapeutics (466)
  • Primary Care Research (452)
  • Psychiatry and Clinical Psychology (3432)
  • Public and Global Health (6527)
  • Radiology and Imaging (1403)
  • Rehabilitation Medicine and Physical Therapy (814)
  • Respiratory Medicine (871)
  • Rheumatology (409)
  • Sexual and Reproductive Health (410)
  • Sports Medicine (342)
  • Surgery (448)
  • Toxicology (53)
  • Transplantation (185)
  • Urology (165)