Open Access

Effects of Egr1 on pancreatic acinar intracellular trypsinogen activation and the associated ceRNA network

  • Authors:
    • Bo Gao
    • Xueming Zhang
    • Dongbo Xue
    • Weihui Zhang
  • View Affiliations

  • Published online on: July 9, 2020     https://doi.org/10.3892/mmr.2020.11316
  • Pages: 2496-2506
  • Copyright: © Gao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Acute pancreatitis (AP) is a common digestive disorder with high morbidity and mortality. The present study aimed to investigate the expression of early growth response protein 1 (Egr1), and the effect of competing endogenous (ce)RNA network on trypsinogen activation. Pancreatic acinar intracellular trypsinogen activation (PAITA) is an important event in the early stage of AP; however, the underlying mechanisms remain unclear. The present study used taurolithocholic acid 3‑sulfate (TLC‑S)‑treated AR42J cells (pancreatic cell line) to establish a PAITA model. A gene microarray and bioinformatics analysis was performed to identify the potential key targets in PAITA. The results demonstrated that Egr1, an important transcription factor, was significantly overexpressed in PAITA. In Egr1 small interfering (si)RNA‑transfected cells, Egr1 expression was decreased and trypsinogen activation was significantly decreased compared with negative control siRNA‑transfected cells, indicating that in TLC‑S‑induced PAITA, overexpression of Egr1 enhanced trypsinogen activation. A ceRNA network [mRNA‑microRNA (miRNA/miR)‑long non‑coding (lnc)RNA] generated using the PAITA model revealed that the effects of Egr1 on PAITA may be regulated by multiple ceRNA pairs, and the lncRNAs (including NONRATT022624 and NONRATT031002) and miRNAs [including Rattus norvegicus (rno)‑miR‑214‑3p and rno‑miR‑764‑5p] included in the ceRNA pairs may serve roles in PAITA by regulating the expression of Egr1. The results of the present study may provide novel targets for researching the underlying mechanisms of, and developing treatments for AP.

Introduction

Acute pancreatitis (AP) is a common digestive disorder with high morbidity and mortality. It has been reported that in the USA, AP is a leading cause of inpatient care among gastrointestinal conditions: >275,000 patients are hospitalized for AP annually (1). AP is the second leading cause of hospitalization, the largest contributor to total hospitalization cost and the fifth leading cause of inpatient mortality (2,3) In China, 20–30% of patients with AP are clinically dangerous, and the overall case fatality rate is 5–10% (4). At present, the pathogenic mechanisms of AP remain unclear. Pancreatic acinar intracellular trypsinogen activation (PAITA) is considered to be an important cause of AP and is an important event in the early stages of AP (5). Studies have demonstrated that normal activation of trypsinogen is a key factor for the pancreas to maintain normal function and that the abnormal activation of trypsinogen in pancreatic acinar cells is an initiating factor for the occurrence of AP (6,7). Thus, elucidation of the mechanisms underlying trypsinogen activation and identification of targets that serve key roles in this process are important for determining the pathogenesis of AP and providing clinical treatment.

As an important transcription factor, early growth response 1 (Egr1) is closely associated with the occurrence and development of various diseases, including AP (810). Ji et al (11) demonstrated that Egr1 was expressed in pancreatic acinar cells and served a role in the development of caerulein-induced AP in mice. Gong et al (12) reported that as a proinflammatory transcription factor, Egr1 may serve an important role in the development of early AP by regulating the expression of tissue factor. These studies suggest that PAITA is likely to be the most important early event of AP, and that Egr1 may serve a role in early AP. However, it remains unclear whether Egr1 serves a role in PAITA.

In recent years, non-coding RNAs have been demonstrated to serve important roles in the occurrence and development of a variety of diseases including cancer and leukemia (13,14). As an important non-coding RNA, long non-coding RNA (lncRNAs) have been used as a type of competing endogenous RNA (ceRNA) to affect multiple target genes and participate in the regulation of various biological processes that are closely associated with the occurrence, development and prevention of human diseases (15,16). lncRNAs can affect mRNA expression by competing for a common microRNA (miRNA/miR) binding site; the mRNA-miRNA-lncRNA network is termed a ceRNA network (17). Studies have confirmed that the lncRNA has a close relationship with AP. For example, Zhao et al (18) demonstrated that the lncRNA Fendrr promoted the apoptosis of pancreatic acinar cells in caerulein-induced AP by interacting with annexin A2. Wang et al (19) reported that overexpression of lncRNA B3GALT5-AS1 may alleviate caerulein-induced cell injury in AR42J cells through the regulation of miR-203/NFIL3 axis and by inhibiting the activation of the NF-κB signals. These studies suggested that lncRNAs may be used as an important target for research and treatment of AP. However, it remains unclear whether lncRNAs serve a role in PAITA and whether there is an interaction between Egr1 and PAITA.

The present study used taurolithocholic acid 3-sulfate (TLC-S) to induce AR42J cells to establish a PAITA model. A gene microarray was used to detect the differential expression of lncRNAs, miRNAs and mRNAs in PAITA. Bioinformatics analyses were performed to identify a protein-protein interaction (PPI) network in PAITA in order to investigate the potential role of Egr1 in PAITA. Confocal laser microscopy and flow cytometry were then used to analyze the effects of Egr1 silencing on PAITA. Finally, a ceRNA regulatory network was established to predict the potential mechanisms underlying the influence of Egr1 on PAITA. The results of the present study may provide novel insight for studies into the pathogenesis and clinical treatment of AP.

Materials and methods

Cell culture and treatment

Cell culture and treatment were performed in accordance with a previous study (20). The rat pancreatic acinar AR42J cells were obtained from the China Center for Type Culture Collection (Wuhan, China) and cultured in F12K medium (Sigma-Aldrich; Merck KGaA) supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and 100 U/ml penicillin/streptomycin (Beyotime Institute of Biotechnology) in a 5% CO2 environment at 37°C. A total of 200 µM TLC-S (Sigma-Aldrich; Merck KGaA) was used to treat AR42J cells for 40 min at 37°C to establish the PAITA cell model as previously described (21,22).

Measurement of trypsinogen activation

Quantification of the activity of trypsin serine protease in intact living acinar cells was performed as previously described (22). Briefly, after an equilibration period of 30 min, 200 µM TLC-S was added for 40 min at 37°C. Acinar cells were washed and resuspended in NaHEPES without TLC-S, and then supplemented with 10 µM of the cell-permeant synthetic trypsin substrate bis-(CBZ-Ile-Pro-Arg)-rhodamine 110 (BZiPAR; Molecular Probes; Thermo Fisher Scientific, Inc.) and allowed to react in the dark at 37°C for 20 min. BZiPAR is a specific substrate for trypsin that emits fluorescence after cleavage of the two oligopeptide side chains. Activation may be observed by fluorescence of rhodamine 110 by using an excitation wavelength of 485 nm. Then 0.5 µg/ml DAPI (Beyotime Institute of Biotechnology) was used for 5 min at 37°C to locate the nuclei, which fluoresced green under the laser confocal microscope). To ensure that the observed tryptic activity was solely from intracellular enzymes and not from trypsin released into the extracellular fluid, cells for these experiments were prepared in NaHEPES containing 5 mM soybean trypsin inhibitor (SBTI; Sigma-Aldrich; Merck KGaA); the solutions used all contained SBTI (SBTI and BZiPAR are added to the cell dish together). At this concentration, SBTI can inhibit 1,000 U/ml trypsin. Trypsin activity was investigated by flow cytometry [FACSCalibur II, BD Biosciences; CellQuest software (version 6.0; BD Biosciences) and Kaluza Analysis software program (version 2.1; Beckman Coulter, Inc.)] and confocal microscopy (A1R, Nikon Corporation). A total of 5 different fields in each group were randomly chosen to capture images at 200× magnification.

Detection of mRNAs, lncRNAs and miRNAs in PAITA

An Agilent-062716 Rat lncRNA Microarray (8×60K; Agilent Technologies, Inc.) was used to detect the expression of mRNA and lncRNA in the present study, and then data analyses of the TLC-S group (trypsinogen activation model; 3 samples) and control group (untreated AR42J cells; 3 samples) were performed. Total RNA was quantified using a NanoDrop™ ND-2000 (NanoDrop Technologies; Thermo Fisher Scientific, Inc.) and the RNA integrity was assessed using an Agilent Bioanalyzer 2100 (Agilent Technologies, Inc.). Sample labeling, microarray hybridization and washing were performed based on the manufacturer's standard protocols. Briefly, total RNA was reverse transcribed to double-stranded cDNA, then synthesized into cRNA and labeled with Cyanine-3-CTP. The labeled cRNAs were hybridized onto the microarray. After washing, the arrays were scanned by an Agilent G2505C Scanner (Agilent Technologies, Inc.). Feature Extraction software (version 10.7.1.1, Agilent Technologies, Inc.) was used to analyze array images.

The miRNA expression analysis was performed using Affymetrix® GeneChip® miRNA Arrays (Affymetrix; Thermo Fisher Scientific, Inc.). Poly (A) Tailing, FlashTag ligation, hybridization, washing, staining, and detection were performed in accordance with the protocols of the Affymetrix GeneChip miRNA Arrays using the Library file (http://www.affymetrix.com/support/downloads/manuals/agcc_command_console_user_guide.pdf). Data were normalized using the median normalization. After normalization, differentially expressed (DE) miRNAs were identified through fold change filtering.

Identification of the DE genes in PAITA

The limma package (http://bioconductor.org/packages/Limma/) (23) in R software (version 3.2.11, http://www.bioconductor.org/packages/release/bioc/html/clusterProfiler.html) (24) was used to perform data preprocessing, including background correction, quantile normalization and probe summarization, to obtain the probe expression matrix (25). UCSC RefSeq database (http://genome.ucsc.edu/), miRBase database 21 (http://mirbase.org/) and NONCODE V4 database (http://www.noncode.org/) were used to perform the blast comparison (identity >95%, coverage >95) and obtain the gene ID (26). Combining the probe expression matrix, a gene expression matrix was obtained.

Limma was used to identify the DE mRNAs, miRNAs and lncRNAs. For each significant DE mRNA and lncRNA, the significance level of differential expression was set to log2 fold change ≥2 and P<0.05. For each significant DE miRNA, the significance level of differential expression was set to log2 fold change ≥2. In addition, MultiExperiment Viewer software 4.6.0 (http://mev.tm4.org/) was used to conduct the cluster analysis and draw the heatmaps.

PPI network of the DE mRNAs in PAITA

A PPI network of the DE mRNAs in trypsinogen activation was obtained from the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database (version 8.0; www.string-db.org) and analyzed using Limma (22,27). Cytoscape software (version 3.6.1; www.cytoscape.org) was used to visualize the PPI network. The node color of the genes was set to different gradients according to the fold change and the node size was set according to the P-value.

Analysis of gene function and signaling pathway enrichment

The function of DE genes and the enriched signaling pathway were investigated via Gene Ontology (GO) (28) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) (29) analysis through the Database for Annotation, Visualization and Integrated Discovery (DAVID; http://david.ncifcrf.gov). Fisher's exact test was applied and P<0.05 was considered to indicate a statistically significant difference.

Small interfering (si)RNA transfection

siRNAs were transfected into cells using Lipofectamine® RNAiMAX transfection reagent (cat. no. 13778-150, Gibco; Thermo Fisher Scientific, Inc.). AR42J cells (1×105 cells) were seeded into six-well plates in complete DMEM and cultured overnight at 37°C. The siRNA lipoRNAiMAX mixture was prepared and added to a culture wells containing cells suspended in 800 µl F12K medium, with a cell density of about 30–40% (2×105 cells). The siRNAs were used at a final concentrations of 50 nM. After 4–6 h of culture at 37°C, the culture medium was replaced with fresh medium (containing fetal bovine serum and penicillin/streptomycin), followed by continuous culturing for 48 h at 37°C before subsequent experimentation.

The siRNAs were purchased from Shanghai GenePharma Co., Ltd., and their sequences were as follows: Egr1-siRNA, 5′-CCAGGACUUAAAGGCUCUUTTAAGAGCCUUUAAGUCCUGGTT-3′ and negative control (NC)-siRNA, 5′-UUCUCCGAACGUGUCACGUTT-3′.

AR42J cells were divided into four experimental groups: i) Control group, normally cultured AR42J cells; ii) NC-siRNA group, AR42J cells transfected with negative control siRNA; iii) TLC-S + NC-siRNA group, AR42J cells transfected with NC-siRNA and treated with 200 µM TLC-S for 40 min; and iv) TLC-S + Egr1-siRNA group, AR42J cells transfected with Egr1-siRNA and treated with 200 µM TLC-S for 40 min.

Reverse transcription-quantitative (RT-q)PCR

Total RNA from AR42J cell lines was isolated using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and the isolated RNA was reverse transcribed (cat. no. FSQ-101; Toyobo Life Science): 37°C for 15 min, 50°C for 5 min and 98°C enzyme inactivation reaction for 5 min. RT-qPCR was conducted using FastStart Universal SYBR-Green Master (ROX; Roche Diagnostics) and an ABI 7300 system (Applied Biosystems; Thermo Fisher Scientific, Inc.) under the following conditions: 95°C for 10 min, followed by 40 cycles of 95°C for 2 sec and 60°C for 30 sec, with a final extension at 72°C for 10 min. The primers for Egr1 and GAPDH were synthesized by Guangzhou RiboBio Co., Ltd. The relative expression levels of Egr1 were normalized to GAPDH and quantified using the 2−∆∆Cq method (30). The Egr1 and GAPDH primers were as follows: Egr1 forward, 5′-ACTGGAGGAGATGATGCTGCTGAG-3′ and reverse, 5′-CCGCTGCTGCTGCTGCTG-3′; and GAPDH forward, 5′-ACAACTTTGGTATCGTGGAAGG-3′ and reverse, 5′-GCCATCAGCCACAGTTC-3′.

Western blotting

For protein analysis, cells were lysed directly in RIPA buffer (cat. no. P0013K; Beyotime Institute of Biotechnology), and then centrifuged for 10 min at 12,000 × g at 4°C, and the protein concentration was determined using the BCA method. The protein (40 µg) was separated by 8% SDS-PAGE and transferred onto 0.45 µm PVDF membranes. The membranes were blocked with 5% BSA (A8020; Beyotime Institute of Biotechnology) in PBST (0.1% Tween-20, cat. no. 9005-64-5; Beyotime Institute of Biotechnology) for 1 h at room temperature. The membranes were incubated with Egr1 (1:500; cat. no. 4153; Cell Signaling Technology, Inc.) and GAPDH (1:2,000; cat. no. TA-08; ZSGB-BIO) at 4°C overnight. Membranes were then incubated with horseradish peroxidase secondary antibody (1:2,000; cat. no. ZB-2301; OriGene Technologies, Inc.) for 1 h at room temperature. A MiniChemi imager (Beijing Sage Creation Science Co., Ltd.) was used to detect the immunoreactivity. The intensity of the band was relatively quantified using ImageJ software (version 1.8.0; National Institutes of Health). All proteins were normalized to the internal control GAPDH.

Establishing the lncRNA-miRNA-mRNA ceRNA network

According to the results of the differential expression analysis, significant DE lncRNAs, miRNAs and mRNAs were filtered to construct an mRNA-miRNA-lncRNA regulatory network. DE lncRNA and DE miRNA pairs were generated based on the lnCeDB database (31). DE miRNA and DE mRNA pairs were generated based on the miRBase database. When screening the DE lncRNA-miRNA pairs and DE miRNA-mRNA pairs, a constraint that the regulatory directions of the miRNA and mRNA must be different was applied. Based on the results, Cytoscape(version 3.6.1; www.cytoscape.org) was used to construct the mRNA-miRNA-lncRNA ceRNA network.

Statistical analysis

All data are presented as the mean ± SD. RT-qPCR data were analyzed with unpaired Student's t-test. The group differences in flow cytometry analysis were determined with one-way ANOVA followed by Tukey's post hoc test. Statistical analysis was performed using GraphPad Prism 5.0 software (GraphPad Software, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

DE mRNAs, lncRNAs and miRNAs in PAITA

The gene microarray was used to identify the DE mRNAs, lncRNAs and miRNAs in PAITA. Compared with the control group, 206 DE mRNAs, 19 DE lncRNAs and 23 DE miRNAs were observed in PAITA (Table I). The scatter plot and heatmap of DE genes are presented in Fig. 1A and B. In the scatter plot, the blue node represents the gene decrease, the yellow node represents the gene increase, and the node size is positively correlated with the P-value. In the heat map red sections represent an increase in expression and green sections represent a decrease in expression. The values −1 and +1 represent Z-score.

Table I.

Number of DE mRNAs, miRNAs and lncRNAs in a cell model of PAITA.

Table I.

Number of DE mRNAs, miRNAs and lncRNAs in a cell model of PAITA.

ExpressionDE mRNADE miRNADE lncRNA
Upregulated172916
Downregulated34143
Sum2062319

[i] DE, differentially expressed; lncRNA, long non-coding RNA; miRNA, microRNA; PAITA, pancreatic acinar intracellular trypsinogen activation.

PPI network in PAITA

In order to further investigate the potential mechanisms involved in PAITA, the STRING database and Cytoscape software were used to establish a PPI network in PAITA. The results demonstrated 262 interaction pairs among the 206 DE mRNAs (Fig. 2). Important genes in the PAITA network are indicated by darker colors, larger sizes and higher degrees of interaction, such as nuclear receptor subfamily 4 group a member 1 (NR4A1), ARIIGAP5, FOS, COX11, BTG2, Egr1, dual specificity phosphatase 5 (Dusp5) and adrenomedullin (ADM; Fig. 2). A literature review of differentially expressed proteins was conducted to determine the association between proteins from regulatory networks and AP. The results suggested that although Egr1 did not display the most pronounced fold change, it is indeed most closely associated with AP.

Functional and pathway analysis of the PPI network in PAITA

The DAVID database was used to analyze the enriched functions and pathways of genes in the PPI network. GO analyses demonstrated that the network genes were mainly enriched in ‘regulation of cell cycle’, ‘positive regulation of translation’, ‘cellular response to DNA damage stimulus’, ‘meiotic chromosome condensation’ and ‘mitotic nuclear division’ (P<0.05; Fig. 3A).

KEGG analyses demonstrated that the network genes were mainly enriched in ‘MAPK signaling pathway’, ‘RNA transportation’, ‘cell cycle’, ‘ubiquitin mediated proteolysis’ and ‘mRNA surveillance pathway’ (P<0.05; Fig. 3B).

Egr1 knockdown

Among the important genes of the PPI network, Egr1 exhibited the closest relationship with AP according to a review of the literature (812). The microarray analysis demonstrated that Egr1 was upregulated in PAITA (Fig. 4A), but its effects remain unclear. Thus, siRNA was used to knockdown the expression of Egr1. RT-qPCR and western blotting were performed to detect the efficiency of knockdown. RT-qPCR and western blotting demonstrated that the expression levels of Egr1 were significantly decreased in the Egr1-siRNA group compared with in the of the NC-siRNA group; the knockdown rate was ~75% (Fig. 4B and C).

Egr1 regulates the activation of trypsinogen

To understand the effects of Egr1 on PAITA, laser scanning confocal microscopy was performed to detect the levels of trypsinogen activation in the TLC-S + Egr1-siRNA, TLC-S + NC-siRNA and NC-siRNA groups. The results demonstrated that the intracellular fluorescent area in the TLC-S + NC-siRNA group was notably larger compared with that of the NC-siRNA group. In addition, compared with the intracellular fluorescent area of the TLC-S + NC-siRNA group, the TLC-S + Egr1-siRNA group demonstrated a decreased intracellular fluorescent area (Fig. 5).

To quantify the fluorescence levels, flow cytometry was performed to detect the levels of trypsinogen activation in the three groups. The result demonstrated that the percentage of PAITA-positive cells was 9.12±1.40, 8.95±2.43, 25.61±2.16 and 0.07±0.03% in the control, NC-siRNA, TLC-S + NC-siRNA and TLC-S + Egr1-siRNA groups, respectively (Fig. 6B). No significant difference was observed between the control group and the NC-siRNA group (P>0.05), but the percentage of positive cells in the TLC-S + NC-siRNA group was significantly higher compared with that of the NC-siRNA and TLC-S + Egr1-siRNA groups (P<0.05) and the percentage of positive cells in the NC-siRNA group was significantly higher compared with that of the TLC-S + Egr1-siRNA group (P<0.01; Fig. 6A and B).

The results also demonstrated that the mean fluorescence intensity was 11.53±1.05, 11.61±1.71, 20.71±1.65 and 6.25±0.52 in the control, NC-siRNA, TLC-S + NC-siRNA and TLC-S + Egr1-siRNA groups, respectively. No significant difference was observed between the control group and the NC-siRNA group (P>0.05), but the mean fluorescence intensity in the TLC-S + NC-siRNA group was significantly higher than that of the NC-siRNA group and the TLC-S + Egr1-siRNA group (P<0.01), and the mean fluorescence intensity in the NC-siRNA group was significantly higher compared with the TLC-S + Egr1-siRNA group (P<0.01; Fig. 6C).

ceRNA network of PAITA

The microarray and bioinformatics analyses were used to establish an lncRNA-miRNA-mRNA network of PAITA, which contained 16 lncRNAs, 18 miRNAs and 25 mRNAs, indicating that PAITA has a complex ceRNA regulatory network (Fig. 7). In order to further clarify the mechanism of Egr1 in PAITA, it was demonstrated that the Egr1-associated ceRNA sub-network included four miRNAs [Rattus norvegicus (rno)-miR-214-3p, rno-miR-764-5p, rno-miR-338-5p and rno-miR-466b-5p] and 10 lncRNAs (NONRATT022624, NONRATT031002, NONRATT021194, NONRATT029766, NONRATT023334, NONRATT031103, NONRATT006386, NONRATT012585, NONRATT017440 and NONRATT015824) in total (Fig. 7).

The microarray and bioinformatics results also demonstrated that Egr1 displayed the top three degrees in the network, which suggested that Egr1 maybe serve an important role in PAITA. Additionally, two miRNAs and four lncRNAs in the Egr1-associated ceRNA sub-network displayed the top five degrees in the network, including rno-miR-338-5p, rno-miR-466b-5p, NONRATT029766, NONRATT023334, NONRATT012585 and NONRATT015824. These results suggested that the effects of Egr1 on PAITA may be regulated by multiple ceRNA pairs, and the lncRNAs and miRNAs included in the ceRNA pairs may serve important roles in PAITA through increasing the expression of Egr1.

Discussion

Studies have demonstrated that maintaining the normal activated form of trypsinogen is key to maintaining the normal function of the pancreas and that inappropriate early activation is the initiating factor for the occurrence of AP (3234). However, the pathogenic mechanisms of AP and PAITA have not been studied thoroughly. Previous studies have observed that complex genetic network regulation, including coding genes and non-coding genes, is involved in the pathogenesis and development of diseases (35,36). Thus, in the present study, gene chips were used to discover the gene networks and corresponding important network nodes involved in a cell model of PAITA. The microarray results demonstrated that 206 mRNAs, 19 lncRNAs and 23 miRNAs were differentially expressed, and that these differential genes were mainly highly expressed. Based on this result, the present study established a genetic interaction network in PAITA. Through functional analysis, the present study demonstrated that these genes mainly served roles in regulating the cell cycle and positive translation, and that the network genes are mainly enriched in signaling pathways such as the mitogen-activated protein kinase (MAPK) pathway, RNA transportation and the cell cycle. de Dios et al (37) observed that cell cycle changes are important features of AP and occur throughout the entire process of AP; in early AP, pancreatic cells exhibit S phase arrest, and with the aggravation of inflammation, G2/M phase arrest occurs. The results of the present study suggested that PAITA, as an early AP event, is characterized by cell cycle changes. The MAPK signaling pathway is one of the most classical signaling pathways in the occurrence and development of AP (38). For example, Cao et al (39) demonstrated that inhibition of MAPK signaling in mice effectively inhibited the development of pancreatitis. Additionally, another study demonstrated that inhibition of MAPK signaling accelerated the apoptosis of pancreatic acinar cells and decreased the AP-associated inflammatory response (40). In the present study, the DE network genes in PAITA were mainly enriched in the MAPK signaling pathway, suggesting that activation of the MAPK signaling pathway affects the process of PAITA and may generate a subsequent cascade reaction, ultimately promoting further development of AP.

In addition, the gene network in PAITA revealed some potentially important genes, including Fos, Egr1, Dusp5, ADM and NR4A1. These genes exhibited higher degree scores in the network, larger fold changes and smaller P-values. Among these genes, the transcription factor Egr1 has a very close relationship with AP. For example, Sandoval et al (41) observed that Egr1 was expressed at high levels in the early inflammatory reaction of AP, and Ji et al (11) demonstrated that after initiating caerulein-induced AP in mice, the severity of AP in Egr1 gene-deficient mice was significantly lower compared with that in normal mice. Their study suggested that Egr1 may be a key regulator of early development of AP. However, it has not been reported whether Egr1 is involved in PAITA (the initial stage of AP). The microarray and gene network results of the present study suggested that Egr1 may serve a role in PAITA. To further verify this effect, siRNA was used to silence Egr1, and then confocal laser microscopy and flow cytometry were used to detect the effect of Egr1 on PAITA. The results demonstrated that silencing of Egr1 significantly inhibited PAITA.

Previous studies have observed that Egr1 is located on the q31.1 ‘cytokine aggregation’ region of human chromosome 5, and as a transcription factor, it is closely associated with cell proliferation, differentiation, apoptosis and inflammatory responses (4245). A number of transcription factors, including activator protein 1, NF-κB and Egr1, can regulate the expression of tumor necrosis factor-α (TNF-α), transforming growth factor β1 and other cytokines by various stimulating factors in different types of cells, such as macrophages and epidermal keratinocytes (46,47). In addition, a number of studies have observed that the transcription factor Egr1 serves an important role in regulating the expression of these stimulating factors and affects the occurrence and development of diseases (4852). These results suggested that Egr1 may cause PAITA via the regulation of PAITA-associated pathogenic factors. However, the effect of Egr1 on the regulation of PAITA through the regulation of downstream genes require validation in further molecular experiments.

With the generation of large quantities of basic experimental data, bioinformatics has been widely used as an effective means for big data analysis in various diseases. To further investigate the potential mechanisms underlying the effects of Egr1 on PAITA, gene mapping and bioinformatics methods were used to establish an mRNA-miRNA-lncRNA network in the cell model of PAITA. The expression of lncRNAs was increased, and these can reduce miRNA inhibition of mRNA by competing to bind miRNA binding sites, thereby causing increased mRNA expression. The results of the present study demonstrated that there was an Egr1-associated ceRNA sub-network in PAITA, including four miRNAs and 10 lncRNAs. miRNA was the first type of non-coding RNA to be discovered, and its function has been extensively studied (53). Of the four miRNAs in the Egr1-related ceRNA network, two have been clearly associated with inflammation. Liu et al (54) reported that miR-338 inhibits TNF-α and thereby inhibits the occurrence of sebaceous inflammation. Zhao et al (55) demonstrated that miR-214 is closely associated with inflammatory responses, and could promote the release of the inflammatory cytokines TNF-α and interleukin-6. These four miRNAs (miR-214-3p, rno-miR-764-5p, rno-miR-338-5p and rno-miR-466b-5p) displayed low expression in the cell model of PAITA, suggesting that they may serve a role in processes involved in PAITA. Notably, these miRNAs all have complementary binding base sites to Egr1, suggesting that Egr1 may be regulated by these upstream miRNAs. A previous study suggested that the regulation of target RNAs by miRNAs is negative and unidirectional (54), but later studies have reported that lncRNAs can compete with their encoded genes for miRNAs via conserved miRNA response elements, resulting in decreased miRNA content and thereby reduced inhibition by miRNAs of target RNAs (56,57). The Egr1-related ceRNA sub-network contains 10 lncRNAs; currently, the role of these lncRNAs in PAITA remains unclear. Combined with the results of microarray and bioinformatics analysis, these 10 lncRNAs were determined to be highly expressed in PAITA and exhibited a competitive relationship with Egr1, suggesting that these lncRNAs may become therapeutic targets for PAITA.

In summary, the results of the present study suggested that Egr1 was highly expressed in PAITA and may affect the development of PAITA. Further analysis revealed that a lncRNA-miRNA-Egr1 regulatory network may be the basis for the effects of Egr1 on PAITA. The results of the present study may provide novel insight for studies into the pathogenesis of AP and PAITA, as well as the development of strategies for the clinical treatment of AP.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81570581), the Youth Program of National Natural Science Foundation of China (grant no. 81800573) and by the Peking University People's Hospital Research And Development Funds (grant no. RDY2018-06).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

DX and WZ designed the study. BG and XZ provided study material, performed the experiments and assembled the data. BG performed experiments and wrote the manuscript. All authors approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Krishna SG, Kruger AJ, Patel N, Hinton A, Yadav D and Conwell DL: Cholecystectomy during index admission for acute biliary pancreatitis lowers 30-day readmission rates. Pancreas. 47:996–1002. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Chaitoff A, Cifu AS and Niforatos JD: Initial management of acute pancreatitis. JAMA. May 7–2020.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

3 

Lankisch PG, Apte M and Banks PA: Acute pancreatitis. Lancet. 386:85–96. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Pancreas Study Group, Chinese Society of Gastroenterology, Chinese Medical Association, Editorial Board of Chinese Journal of Pancreatology & Editorial Board of Chinese Journal of Digestion, . Chinese guidelines for the management of acute pancreatitis (Shanghai, 2013). J Clin Hepatol. 29:656–660. 2013.

5 

Hofbauer B, Saluja AK, Lerch MM, Bhagat L, Bhatia M, Lee HS, Frossard JL, Adler G and Steer ML: Intra-acinar cell activation of trypsinogen during caerulein-induced pancreatitis in rats. Am J Physiol. 275:G352–G362. 1998.PubMed/NCBI

6 

Dawra R, Sah RP, Dudeja V, Rishi L, Talukdar R, Garg P and Saluja AK: Intra-acinar trypsinogen activation mediates early stages of pancreatic injury but not inflammation in mice with acute pancreatitis. Gastroenterology. 141:2210–2217.e2. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Lerch MM and Gorelick FS: Early trypsinogen activation in acute pancreatitis. Med Clin North Am. 84549–563. (viii)2000. View Article : Google Scholar : PubMed/NCBI

8 

Hu F, Xue M, Li Y, Jia YJ, Zheng ZJ, Yang YL, Guan MP, Sun L and Xue YM: Early growth response 1 (Egr1) is a transcriptional activator of NOX4 in oxidative stress of diabetic kidney disease. J Diabetes Res. 2018:34056952018. View Article : Google Scholar : PubMed/NCBI

9 

Kaufmann A, Rössler OG and Thiel G: Expression of the transcription factor Egr-1 in pancreatic acinar cells following stimulation of cholecystokinin or Gαq-coupled designer receptors. Cell Physiol Biochem. 33:1411–1425. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Shajahan-Haq AN, Boca SM, Jin L, Bhuvaneshwar K, Gusev Y, Cheema AK, Demas DD, Raghavan KS, Michalek R, Madhavan S and Clarke R: EGR1 regulates cellular metabolism and survival in endocrine resistant breast cancer. Oncotarget. 8:96865–96884. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Ji B, Chen XQ, Misek DE, Kuick R, Hanash S, Ernst S, Najarian R and Logsdon CD: Pancreatic gene expression during the initiation of acute pancreatitis: Identification of EGR-1 as a key regulator. Physiol Genomics. 14:59–72. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Gong LB, He L, Liu Y, Chen XQ and Jiang B: Expression of early growth response factor-1 in rats with cerulein-induced acute pancreatitis and its significance. World J Gastroenterol. 11:5022–5024. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Wang W, Han Y, Jo HA, Lee J and Song YS: Non-coding RNAs shuttled via exosomes reshape the hypoxic tumor microenvironment. J Hematol Oncol. 13:672020. View Article : Google Scholar : PubMed/NCBI

14 

Ren N, Jiang T, Wang C, Xie S, Xing Y, Piao D, Zhang T and Zhu Y: lncRNA ADAMTS9-AS2 inhibits gastric cancer (GC) development and sensitizes chemoresistant GC cells to cisplatin by regulating miR-223-3p/NLRP3 axis. Aging (Albany NY). 12:110252020. View Article : Google Scholar : PubMed/NCBI

15 

Renganathan A and Felley-Bosco E: Long noncoding RNAs in cancer and therapeutic potential. Adv Exp Med Biol. 1008:199–222. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Li B, Mao R, Liu C, Zhang W, Tang Y and Guo Z: lncRNA FAL1 promotes cell proliferation and migration by acting as a CeRNA of miR-1236 in hepatocellular carcinoma cells. Life Sci. 197:122–129. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Salmena L, Poliseno L, Tay Y, Kats L and Pandolfi PP: A ceRNA hypothesis: The rosetta stone of a hidden RNA language? Cell. 146:353–358. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Zhao D, Ge H, Ma B, Xue D, Zhang W, Li Z and Sun H: The interaction between ANXA2 and lncRNA Fendrr promotes cell apoptosis in caerulein-induced acute pancreatitis. J Cell Biochem. Nov 26–2018.(Epub ahead of print).

19 

Wang L, Zhao X and Wang Y: The pivotal role and mechanism of long non-coding RNA B3GALT5-AS1 in the diagnosis of acute pancreatitis. Artif Cells Nanomed Biotechnol. 47:2307–2315. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Li Z, Lu M, Chu J, Qiao X, Meng X, Sun B, Zhang W and Xue D: Early proteome analysis of rat pancreatic acinar AR42J cells treated with taurolithocholic acid 3-sulfate. Pancreatology. 12:248–256. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Yang Z, Yang W, Lu M, Li Z, Qiao X, Sun B, Zhang W and Xue D: Role of the c-Jun N-terminal kinase signaling pathway in the activation of trypsinogen in rat pancreatic acinar cells. Int J Mol Med. 41:1119–1126. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Song Z, Huang Y, Liu C, Lu M, Li Z, Sun B, Zhang W and Xue D: miR-352 participates in the regulation of trypsinogen activation in pancreatic acinar cells by influencing the function of autophagic lysosomes. Oncotarget. 9:10868–10879. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Fan T, Qu R, Yu Q, Sun B, Jiang X, Yang Y, Huang X, Zhou Z, Ouyang J, Zhong S and Dai J: Bioinformatics analysis of the biological changes involved in the osteogenic differentiation of human mesenchymal stem cells. J Cell Mol Med. May 28–2020.(Epub ahead of print). View Article : Google Scholar

24 

Zhang J, Cong R, Zhang Q, Zeng T, Song R and Meng X: Integrative analysis of ceRNA network and DNA methylation associated with gene expression in malignant pheochromocytomas: A study based on the cancer genome atlas. Transl Androl Urol. 9:344–354. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Mente S and Kuhn M: The use of the R language for medicinal chemistry applications. Curr Top Med Chem. 12:1957–1964. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Xie C, Yuan J, Li H, Li M, Zhao G, Bu D, Zhu W, Wu W, Chen R and Zhao Y: NONCODEv4: Exploring the world of long non-coding RNA genes. Nucleic Acids Res 42 (Database Issue). D98–D103. 2014. View Article : Google Scholar

27 

Szklarczyk D, Morris JH, Cook H, Kuhn M, Wyder S, Simonovic M, Santos A, Doncheva NT, Roth A, Bork P, et al: The STRING database in 2017: Quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 45(D1): D362–D368. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Kanehisa M and Goto S: KEGG: Kyoto encyclopaedia of genes and genomes. Nucleic Acids Res. 28:27–30. 2000. View Article : Google Scholar : PubMed/NCBI

29 

Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, et al: Gene ontology: Tool for the unification of biology. The gene ontology consortium. Nat Genet. 25:25–29. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Das S, Ghosal S, Sen R and Chakrabarti J: lnCeDB: Database of human long noncoding RNA acting as competing endogenous RNA. PLoS One. 9:e989652014. View Article : Google Scholar : PubMed/NCBI

32 

Yao W, Zhu D, Lu H, Liu C, Sun B, Zhang W and Xue D: The regulatory effect of the kinase inhibitor PD98059 on autophagic flux during trypsinogen activation in pancreatic acinar cells. Pancreas. 49:290–299. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Gukovskaya AS, Gorelick FS, Groblewski GE, Mareninova OA, Lugea A, Antonucci L, Waldron RT, Habtezion A, Karin M, Pandol SJ and Gukovsky I: Recent insights into the pathogenic mechanism of pancreatitis: Role of acinar cell organelle disorders. Pancreas. 48:459–470. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Bhatia M, Wong FL, Cao Y, Lau HY, Huang J, Puneet P and Chevali L: Pathophysiology of acute pancreatitis. Pancreatology. 5:132–144. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Gao B, Zhang X, Huang Y, Yang Z, Zhang Y, Zhang W, Gao ZH and Xue D: Coding and non-coding gene regulatory networks underlie the immune response in liver cirrhosis. PLoS One. 12:e01741422017. View Article : Google Scholar : PubMed/NCBI

36 

Gao B, Shao Q, Choudhry H, Marcus V, Dong K, Ragoussis J and Gao ZH: Weighted gene co-expression network analysis of colorectal cancer liver metastasis genome sequencing data and screening of anti-metastasis drugs. Int J Oncol. 49:1108–1118. 2016. View Article : Google Scholar : PubMed/NCBI

37 

de Dios I, Uruñuela A, Pinto RM, Orfao A and Manso MA: Cell-cycle distribution of pancreatic cells from rats with acute pancreatitis induced by bile-pancreatic obstruction. Cell Tissue Res. 300:307–314. 2000. View Article : Google Scholar : PubMed/NCBI

38 

Zhang B, Li SL, Xie HL, Fan JW, Gu CW, Kang C and Teng MJ: Effects of silencing the DUSP1 gene using lentiviral vector-mediated siRNA on the release of proinflammatory cytokines through regulation of the MAPK signaling pathway in mice with acute pancreatitis. Int J Mol Med. 41:2213–2224. 2018.PubMed/NCBI

39 

Cao MH, Xu J, Cai HD, Lv ZW, Feng YJ, Li K, Chen CQ and Li YY: p38 MAPK inhibition alleviates experimental acute pancreatitis in mice. Hepatobiliary Pancreat Dis Int. 14:101–106. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Ma R, Yuan F, Wang S, Liu Y, Fan T and Wang F: Calycosin alleviates cerulein-induced acute pancreatitis by inhibiting the inflammatory response and oxidative stress via the p38 MAPK and NF-κB signal pathways in mice. Biomed Pharmacother. 105:599–605. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Sandoval J, Pereda J, Pérez S, Finamor I, Vallet-Sánchez A, Rodríguez JL, Franco L, Sastre J and López-Rodas G: Epigenetic regulation of early- and late-response genes in acute pancreatitis. J Immunol. 197:4137–4150. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Woo SM, Min KJ, Kim S, Park JW, Kim DE, Chun KS, Kim YH, Lee TJ, Kim SH, Choi YH, et al: Silibinin induces apoptosis of HT29 colon carcinoma cells through early growth response-1 (EGR-1)-mediated non-steroidal anti-inflammatory drug-activated gene-1 (NAG-1) up-regulation. Chem Biol Interact. 211:36–43. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Yan L, Wang Y, Liang J, Liu Z, Sun X and Cai K: miR-301b promotes the proliferation, mobility, and epithelial-to-mesenchymal transition of bladder cancer cells by targeting EGR1. Biochem Cell Biol. 95:571–577. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Leu SY, Kuo LH, Weng WT, Lien IC, Yang CC, Hsieh TT, Cheng YN, Chien PH, Ho LC, Chen SH, et al: Loss of EGR-1 uncouples compensatory responses of pancreatic β cells. Theranostics. 10:4233–4249. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Billah M, Ridiandries A, Rayner BS, Allahwala UK, Dona A, Khachigian LM and Bhindi R: Egr-1 functions as a master switch regulator of remote ischemic preconditioning-induced cardioprotection. Basic Res Cardiol. 115:32019. View Article : Google Scholar : PubMed/NCBI

46 

Fichtner-Feigl S, Strober W, Kawakami K, Puri RK and Kitani A: IL-13 signaling through the IL-13alpha2 receptor is involved in induction of TGF-beta1 production and fibrosis. Nat Med. 12:99–106. 2016. View Article : Google Scholar

47 

Hogan KA, Ravindran A, Podolsky MA and Glick AB: The TGFβ1 pathway is required for NFκB dependent gene expression in mouse keratinocytes. Cytokine. 64:652–659. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Ho LC, Sung JM, Shen YT, Jheng HF, Chen SH, Tsai PJ and Tsai YS: Egr-1 deficiency protects from renal inflammation and fibrosis. J Mol Med (Berl). 94:933–942. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Solovey A, Somani A, Belcher JD, Milbauer L, Vincent L, Pawlinski R, Nath KA, Kelm RJ Jr, Mackman N, O'Sullivan MG, et al: A monocyte-TNF-endothelial activation axis in sickle transgenic mice: Therapeutic benefit from TNF blockade. Am J Hematol. 92:1119–1130. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Bi JG, Zheng JF, Li Q, Bao SY, Yu XF, Xu P and Liao CX: MicroRNA-181a-5p suppresses cell proliferation by targeting Egr1 and inhibiting Egr1/TGF-β/Smad pathway in hepatocellular carcinoma. Int J Biochem Cell Biol. 106:107–116. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Li DM, Zhong M, Su QB, Song FM, Xie TG, He JH, Wei J, Lu GS, Hu XX and Wei GN: Active fraction of polyrhachis vicina rogers (AFPR) suppressed breast cancer growth and progression via regulating EGR1/lncRNA-NKILA/NF-κB axis. Biomed Pharmacother. 123:1096162020. View Article : Google Scholar : PubMed/NCBI

52 

Pang Z, Raudonis R, McCormick C and Cheng Z: Early growth response 1 deficiency protects the host against pseudomonas aeruginosa lung infection. Infect Immun. 88:e00678–19. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Xiao C, Nemazee D and Gonzalez-Martin A: MicroRNA control of B cell tolerance, autoimmunity and cancer. Semin Cancer Biol. 64:102–107. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Liu J, Cao L, Feng Y, Li Y and Li T: miR-338-3p inhibits TNF-α-induced lipogenesis in human sebocytes. Biotechnol Lett. 39:1343–1349. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Zhao L, Liu YW, Yang T, Gan L, Yang N, Dai SS and He F: The mutual regulation between miR-214 and A2AR signaling plays an important role in inflammatory response. Cell Signal. 27:2026–2034. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Tay Y, Rinn J and Pandolfi PP: The multilayered complexity of ceRNA crosstalk and competition. Nature. 505:344–352. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Thomson DW and Dinger ME: Endogenous microRNA sponges: Evidence and controversy. Nat Rev Genet. 17:272–283. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2020
Volume 22 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gao B, Zhang X, Xue D and Zhang W: Effects of Egr1 on pancreatic acinar intracellular trypsinogen activation and the associated ceRNA network. Mol Med Rep 22: 2496-2506, 2020
APA
Gao, B., Zhang, X., Xue, D., & Zhang, W. (2020). Effects of Egr1 on pancreatic acinar intracellular trypsinogen activation and the associated ceRNA network. Molecular Medicine Reports, 22, 2496-2506. https://doi.org/10.3892/mmr.2020.11316
MLA
Gao, B., Zhang, X., Xue, D., Zhang, W."Effects of Egr1 on pancreatic acinar intracellular trypsinogen activation and the associated ceRNA network". Molecular Medicine Reports 22.3 (2020): 2496-2506.
Chicago
Gao, B., Zhang, X., Xue, D., Zhang, W."Effects of Egr1 on pancreatic acinar intracellular trypsinogen activation and the associated ceRNA network". Molecular Medicine Reports 22, no. 3 (2020): 2496-2506. https://doi.org/10.3892/mmr.2020.11316