Open Access

Paeonol induces the apoptosis of the SGC‑7901 gastric cancer cell line by downregulating ERBB2 and inhibiting the NF‑κB signaling pathway

  • Authors:
    • Jun Fu
    • Luhua Yu
    • Jie Luo
    • Rui Huo
    • Bing Zhu
  • View Affiliations

  • Published online on: May 23, 2018     https://doi.org/10.3892/ijmm.2018.3704
  • Pages: 1473-1483
  • Copyright: © Fu 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

The purpose of the present study was to analyze the association between paeonol and the known genes related to gastric cancer (GC) using bioinformatics methods, and to investigate the role of paeonol in the potential impact on the nuclear factor‑κB (NF‑κB) signaling pathway, in order to provide a theoretical basis for further elucidating the effect of paeonol on cancer cells. Cell viability, morphology and apoptosis were detected using an MTT assay, an inverted microscope, and flow cytometry, respectively. The correlation between drugs and genes was analyzed using the Search Tool for Interactions of Chemicals (STITCH) gene‑drug interaction network. The expression levels of related mRNA and proteins were determined using reverse transcription‑quantitative polymerase chain reaction analysis and enzyme‑linked immunosorbent assay. The changes in protein expression were examined using western blot analysis. The correlation network between target genes directly affected by paeonol and known GC genes was determined by analyzing the association between the compounds and genes recorded in the STITCH database. The GC‑related epidermal growth factor receptor 2 (ERBB2) gene was at the core position of the paeonol interaction network and may be an important potential target gene for the effect of paeonol on cancer cells. The effect of paeonol on the viability of the SGC‑7901 GC cell line was detected using an MTT assay, which showed that the inhibitory effect occurred in a time‑ and dose‑dependent manner. The observations of cell morphology demonstrated that the cells were floating, abnormal in shape, had unclear boundaries and were sparse in arrangement following paeonol treatment. Flow cytometry indicated that paeonol significantly accelerated the apoptotic rate of the SGC‑7901 GC cells. The examination of clinical samples suggested that ERBB2 was expressed at a high level in GC samples, and was significantly downregulated following the addition of paeonol. The western blot analysis revealed that downregulating ERBB2 affected the activation of the NF‑κB signaling pathway, thereby upregulating the pro‑apoptotic factor B‑cell lymphoma‑associated X protein. Taken together, paeonol significantly downregulated ERBB2 and inhibited the activation of the NF‑κB signaling pathway, thereby inhibiting the proliferation of SGC‑7901 cells and inducing apoptosis.

Introduction

Gastric cancer (GC) is one of the four most common types of cancer and one of the three major causes of cancer-associated mortality in the world. Each year, 2% of all new cases of GC are diagnosed in the United States, with 42.6% in China (13). In the late stage of GC, the average survival rate of patients is usually <12 months (4). As there is no obvious clinical manifestation in early GC, the majority of patients are diagnosed with advanced GC. At this time, the optimal period for surgery has been missed (5). Therefore, it is crucial to investigate the biomarkers and drug targets in the development of GC (6). Currently, the most common biomarkers in GC include carcinoembryonic antigen, tumor antigen 19–9 and tumor antigen 72–4 (79). By examining the changes of biomarkers, an effective and early diagnosis for GC can be made. In addition to surgical treatment of GC, chemotherapy is also one of the major treatments for GC. However, its effect is not satisfactory due to the serious side effects caused by chemotherapy. Therefore, the development of more effective chemotherapeutic drugs for GC has attracted increasing attention.

At present, in the development of novel chemotherapeutic drugs, it is generally accepted that natural products are a viable option, which can serve as an important potential source of novel drugs in cancer chemotherapy. There are various sources of natural products, and the antitumor potential of pharmaceutical plants has been closely examined. For example, the spindle formation of cells is inhibited by paclitaxel, mitosis is suspended by the failure of spindle formation, and cell apoptosis is caused by the interruption of mitosis. The cell death-inducing function towards cancer cells by paclitaxel occurs in this way (10). Studies have shown that paclitaxel can effectively promote the apoptosis of tumor cells and, therefore, be involved in the treatment of breast cancer (11). In addition to paclitaxel, paeonol extracted from the root bark of peony has various pharmacological and physiological effects, including sedation, hypnosis, anti-inflammatory and antitumor effects (1214). Studies on paeonol have indicated that it can prevent the division of multiple cancer cell lines and inhibit the growth of xenograft tumor model in the body (1517). For example, in lung cancer cell lines, paeonol and platinum compounds can synergistically inhibit the growth of tumor cells and induce the apoptosis of tumor cells (18). In addition, the growth status of the HepG2 hepatocellular carcinoma cell line treated with paeonol was reduced, compared with that of the control group (13). In addition, paeonol can inhibit cell cycle and promote the apoptosis of cancer cells in this manner. Paeonol also has a significant inhibitory effect on esophageal cancer and GC, and can induce the apoptosis of GC cells (15,19,20). A preliminary study on the cell-death mechanism of paeonol suggested that the increase in the ratio of inducing factor B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax) and inhibitory factor Bcl-2 is the main reason for paeonol promoting cell apoptosis. A study on these two genes indicate that their expression can control cell apoptosis mediated by mitochondria (21). It has also been demonstrated that paeonol is beneficial for novel drugs in the treatment of GC chemotherapy. At present, the mechanism of paeonol has been understood preliminarily, however, the targets of paeonol on GC cells remain to be elucidated. The identification of drug targets is vital for understanding the mechanism of drugs. Consequently, investigation of the targets of paeonol in the treatment of cancer is particularly important.

The process of cancer occurrence and subsequent tumor migration and diffusion is complicated, and several genes are involved in the process. Currently, with advances in molecular biology and genomics methods, genes involved in tumorigenesis have been increasingly investigated, and the data in associated databases are being constantly enriched. How to identify the target genes of drugs from these complicated data is a major problem in scientific research. The correlation between drugs and genes can be predicted using bioinformatics, and the association between genes and the potential target genes of drug action can be identified quickly and effectively (22,23). This method provides a novel approach for investigations the mechanism of drug action. In the present study, the potential target genes of paeonol associated with GC were investigated by combining the method of pre-analysis with experiments (22,23). Using bioinformatics, a potential target gene of paeonol, epidermal growth factor receptor 2 (ERBB2), in the treatment of GC was identified and it was found that the nuclear factor (NF)-κB signaling pathway may be associated with the role of paeonol. The effects of paeonol on ERBB2 and the NF-κB signaling pathway were preliminarily examined in subsequent experiments, providing a theoretical basis for further elucidating the mechanism underlying the effect of paeonol on cancer cells.

Materials and methods

Bioinformatics retrieval

The paeonol drug and its targets were searched and analyzed using the Search Tool for Interactions of Chemicals (STITCH; http://stitch.embl.de/) tool. GC-related genes were searched using PolySearch2 (http://polysearch.cs.ualberta.ca/). The shortest path between the genes was calculated using Dijkstra's algorithm, thereby predicting the potential genes. GC-related genes were subsequently analyzed using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING; version 10.5; https://string-db.org/).

Cell culture

The SGC-7901 GC cell line was obtained from the Shanghai Cell Bank of Chinese Academy of Sciences (Shanghai, China). The cells were cultured with RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), which contained 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), and incubated in a 5% CO2 incubator at 37°C. The culture medium was replaced every 2–3 days. Following culture, SGC-7901 cells in the logarithmic growth phase were used for transfection experiments. Cell transfection was performed using Lipofectamine 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and morphology was observed under an inverted microscope (×300).

Tissue samples

The GC and adjacent tissues were collected from patients (age, 40–80; 14 females and 36 males) the Department of Gastrointestinal Surgery of The First Affiliated Hospital of Bengbu Medical College (Bengbu, China) between January and December 2013. The First Affiliated Hospital of Bengbu Medical College provided ethical approval for the study and written informed consent was obtained from all subjects. No patients had received chemotherapy or radiotherapy prior to surgery. Tissues were obtained during tumor resection surgery and stored in liquid nitrogen and stored at −80°C.

Detection of cell viability by MTT assay

Paeonol was purchased from Aladdin Industrial Corporation (Shanghai, China) and diluted to different concentrations in DMSO (0, 0.05, 0.1, 0.2, 0.4, 0.6 and 0.8 mg/ml). Cells were seeded (5×103 cells/well) in a 96-well plate and were treated for 24 and 48 h at 37°C by paeonol at different concentrations separately. The treated cells were then screened using the MTT assay. Into each well, 20 µl MTT (5 mg/ml) was added and incubated for 4 h. The medium was carefully aspirated, and 200 µl dimethyl sulfoxide was added into each well and incubated for 10 min at 37°C or oscillated for 10 min until the blue-violet crystals were completely dissolved. The absorbance value was determined at 570 nm using a microplate reader and a cell growth curve was then drawn.

Flow cytometry

Annexin V-FITC/PI double staining was performed to detect cell apoptosis. Following 0.25% trypsin digestion, a single cell suspension was produced, and the cell density was adjusted to 5×105–1×106/ml. The cells were washed with 1 ml PBS three times and centrifuged for 10 min at 157 × g at 4°C, followed by discarding of the culture medium. Subsequently, the cells were resuspended with 200 µl binding buffer and then incubated with Annexin V-FITC (10 µl) and PI (5 µl) for 15 min in the dark at room temperature. Following incubation, 300 µl of binding buffer was added and flow cytometry was performed. The light source was a 488 nm fluorine ion laser. Following excitation, FITC emitted green fluorescence and PI emitted red fluorescence.

Detection of the expression of ERBB2 using an enzyme-linked immunosorbent assay (ELISA)

The protein expression of ERBB2 was detected using ELISA with the Thermo Fisher ELISA kit according to the manufacturer's protocol (Thermo Fisher Scientific, Inc.).

Western blot analysis

Cells at the logarithmic growth phase were selected and washed once with PBS. Fresh protein was extracted following cell lysis in 200 µl of 1% sodium dodecyl sulfate (SDS) in an ice bath. Quantitative detection of proteins was performed using a Pierce protein determination kit. Briefly, following separation by 12% SDS-polyacrylamide gel electrophoresis, 50–100 µl protein was transferred onto polyvinyl difluoride membranes and sealed for 1 h in Tris-buffered saline-Tween 20 (TBST) solution with 5% evaporated milk. Subsequently, phosphoinositide 3-kinase (PI3K; cat. no. SAB4502195; Sigma-Aldrich; EMD Millipore, Billerica, MA, USA; 1:1,000), phosphorylated (p-)PI3K (Tyr607; cat. no. ab182651; Abcam, Cambridge, MA, USA; 1:1,000), Akt (cat. no. SAB4500800; Sigma-Aldrich; EMD Millipore; 1:500), p-Akt (Thr308; cat. no. 05–802R; 1:200; Sigma-Aldrich; EMD Millipore) were used for cell incubation at 4°C overnight. β-actin (cat. no. LCA01; Auragene, Changsha, China; 1:15,000) was incubated with the membrane at room temperature for 2 h, followed by incubation with horseradish peroxidase-conjugated goat anti-mouse or goat anti-rabbit secondary antibodies # (cat nos. SA001 and SA009, respectively; Auragene, Changsha, China; 1:10,000) for 1.5 h at room temperature following TBST rinsing (5 min, three times). Finally, the membranes were washed with TBST (10 min, three times) and visualized using a chemiluminescent substrate (ECL™, GE Healthcare Life Sciences, Uppsala, Sweden). ImageJ 1. 39U software (National Institutes of Health, Bethesda, MD, USA) was used to analyze the optical density value of the target strips. β-actin served as the internal reference, and the experiment was repeated five times.

Immunocytochemical staining

The cells in logarithmic growth phase were selected and stained using standard immunostaining procedures. The p65 protein antibody (cat. no. 8242; 1:100) was purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). The staining with DAB and Warp Red was then observed under a light microscope (24).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

The method used for the RT-qPCR assay was as previously described (18). Total RNA was extracted using TRIzol reagent (Takara Biotechnology Co., Ltd., Dalian, China) and cDNA was synthesized using a PrimeScript™ RT reagent kit (Takara Biotechnology Co., Ltd.) according to the manufacturer's instructions. PCR was performed using a SYBR Premix Ex Taq™ II kit (Takara Biotechnology Co., Ltd.) with the following thermocycling conditions: 95°C for 30 sec, followed by 40 cycles of 95°C for 5 sec and 60°C for 30 sec. The primers of genes were as follows: Interleukin (IL)8, forward 5′-GGCTCTCTTGGCAGCCTTCCTG-3′ and reverse 5′-CTTCTCCACAACCGTCTCACCC-3′; IL17A, forward 5′-CTGTGTCTCTGATGCTGTTGC-3′ and reverse 5′-GTGGAACGGTTGAGGTAGTCT-3′; ERBB2, forward 5′-CTTTTGGGGCCAAACCTTAT-3′ and reverse 5′-CTAGTGGGACGCGGACAT-3′; IL1B, forward 5′-ATGATGGCTTATTACAGTGGCAA-3′ and reverse 5′-GTCGGAGATTCGTAGCTGGA-3′. The 2−ΔΔCq method was used to analyze the relative changes in gene expression (25).

Statistical analysis

All data were analyzed using SPSS 13.0 (SPSS, Inc., Chicago, IL, USA. Enumeration data were assessed using the χ2 test. Using a normality test, measurement data with a normal distribution are expressed as the mean ± standard deviation, whereas measurement data without a normal distribution are expressed as the median M (Q1, Q3). Comparisons between two groups were made using Student's t-test, and one-way analysis of variance was used for more than two groups, followed by Tukey's post-hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Target genes of paeonol

In total, seven genes interacting with paeonol were obtained from the STITCH database (Fig. 1), namely caspase (CASP)3, CASP8, intercellular adhesion molecule 1, prostaglandin-endoperoxide synthase 2 (PTGS2), vascular cell adhesion molecule 1 (VCAM1), tyrosinase (TYR), and CASP10. These genes were directly affected by paeonol and were recognized as the direct targets of paeonol.

GC-related genes

Using 'gastric cancer' as a keyword, the related genes were selected from the literature using the disease gene database PolySearch2. Ultimately, a total of 11 GC-related genes were identified (Table I). These genes were known molecules and molecular mechanisms of GC, therefore, the effect on GC is anticipated to be mediated by these genes. Via STRING analysis of the cluster of these GC-related genes, the association network of the ERBB2, cadherin 1 (CDH1), IL1B, IL8, IL17A and transforming growth factor-β receptor 2 (TGFBR2) genes was formed (Fig. 2A). Through the detection of the expression levels of mRNAs, ERBB2, IL1B, IL8 and IL17A were significantly upregulated in GC tissues, compared with those in adjacent tissues (Fig. 2B).

Table I

Known gastric cancer-related hot genes using PolySearch2.

Table I

Known gastric cancer-related hot genes using PolySearch2.

Z-scoreR-scoreEntity IDNameHits
22.971,120PS20265ERBB247
21.121,035PS17336CDH154
16.12805PS00155IL1B8
10.79560PS19669DSG220
8.07435PS00191IL86
6.44360PS01903LTB8
6.00340PS13455TSC118
5.46315PS12239TGFBR22
5.18302PS06478PGA514
5.13300PS10230SLC6A22
5.02295PS00221IL17A3

[i] ERBB2, epidermal growth factor receptor 2; CDH1, cadherin 1; IL, interleukin; DSG2, desmoglein 2; LTB, lymphotoxin β; TSC1, tuberous sclerosis 1; TGFBR2, transforming growth factor-β receptor 2; PGA5, pepsinogen 5, group I (pepsinogen A); SLC6A2, solute carrier family 6 member 2.

Interaction network between paeonol and GC-related genes

To investigate the association between paeonol and GC-related genes, the correlations between the target genes directly affected by paeonol and the known GC genes were analyzed through gene-gene interaction networks in the STRING database. Furthermore, the shortest paths between these genes were calculated using Dijkstra's algorithm. The interaction network between the compounds and genes was constructed, as shown in Fig. 3.

None of the seven target genes directly affected by paeonol were known GC-related genes and had not been found to be involved in GC, however, they were closely related with the known GC-related genes. Through the calculation of the connection degree between these target genes and the known genes (Table I), it was found that the target genes of paeonol have a wide range of connections with the known genes, suggesting that paeonol was likely to affect the pathogenesis of GC by acting on these genes. In addition, according to the core degree and connectivity, several genes (Table II) were at the core of the connection in the whole interaction network, indicating their importance in GC and reflecting that the mechanism underlying the effect of paeonol in GC involved indirectly affecting these core genes. The GC-related core genes with indirect associations were at the core of paeonol, including ERBB2, CDH1, C-X-C motif chemokine ligand 8 (CXCL8; IL8), IL17A, IL1B and TGFBR2, among which ERBB2 was the most important gene in the GC-related genes (Z-score, 22.97; R-score, 1,120; Table I) and paeonol core genes, indicating that it may be a potential factor in paeonol affecting GC. However, there are few reports on the association between paeonol and ERBB2.

Table II

Core molecules in the interaction network of paeonol and genes.

Table II

Core molecules in the interaction network of paeonol and genes.

GeneK-core valueDegree
CASP3936
CASP8934
CDH1930
CXCL8928
ERBB2928
ICAM1932
IL17A926
IL1B926
PTGS2932
TGFBR2924
VCAM1926

[i] CASP, caspase; CDH1, cadherin 1; CXCL8, C-X-C motif chemokine ligand 8; ERBB2, epidermal growth factor receptor 2; ICAM1, intercellular adhesion molecule 1; IL, interleukin; PTGS2, prostaglandin-endoperoxide synthase 2; TGFBR2, transforming growth factor-β receptor 2; VCAM 1, vascular cell adhesion molecule 1.

Enrichment analysis of signaling pathways of genes affected by paeonol

The genes found to be directly or indirectly affected by paeonol were subjected to enrichment analysis of signaling pathways (Fig. 4). The results demonstrated that the genes affected by paeonol were mainly concentrated in the Cytokine-receptor interaction, tumor necrosis factor (TNF) and NF-κB signaling pathways. Among these, the ERBB2 and NF-κB signaling pathway have been reported frequently in breast cancer, and their role in GC requires further investigation.

Paeonol inhibits the growth of SGC-7901 cells

The cells were treated with 0, 0.05, 0.1, 0.2, 0.4, 0.6 and 0.8 mg/ml paeonol and detected using an MTT assay following 24 and 48 h, respectively. The results showed that, with the increase in the dose of paeonol, the inhibitory effect on cell viability was increased. There were no significant differences in the inhibitory effects at the same concentration between the different time points. The half maximal inhibitory concentration (IC50) of paeonol was 0.395 mg/ml at 48 h, as assessed by the MTT assay. This suggested that the inhibitory effect of paeonol on SGC-7901 cells was significantly dose-dependent (Fig. 5).

Paeonol disrupts the normal morphology of SGC-7901 cells

Following treatment with paeonol, the morphological changes of the cells were observed under an inverted microscope (×300). As shown in Fig. 6, without the addition of paeonol, the cells were clear in shape, were well arranged and floating cells were almost absent. Following the addition of paeonol, cell morphology was significantly altered. With the increase in the concentration of paeonol, the morphological changes of the cells were more marked. Following the addition of 0.4 mg/ml paeonol, the cells appeared blurred, abnormal in shape and sparse in arrangement, and there were several floating cells. This indicated that the concentration of paeonol significantly affected the morphology of the GC cells.

Paeonol promotes the apoptosis of SGC-7901 cells

Following treatment with paeonol, the death of GC cells at different concentrations was measured. In the control group, without the addition of paeonol, there were fewer dead cells and apoptotic rate was low. Following the addition of paeonol, the apoptotic rate of the cells was increased (Fig. 7), particularly following the addition of 0.4 mg/ml paeonol, with which the rate of apoptosis was significantly increased. Flow cytometry showed that the apoptotic number was notably increased, which confirmed the above results. Following the addition of paeonol, the rate of cell apoptosis was apparently increased. The higher the concentration of paeonol was, the higher the rate of cell apoptosis was, showing a significant dose-effect association (P<0.05).

Paeonol significantly downregulates the expression of ERBB2

ERBB2 is a known GC-related gene, which is expressed at a high level in GC tissues. In the early bioinformatics analysis, it was found that ERBB2 may be a potential target of paeonol. Therefore, following treatment with different concentrations of paeonol, the expression of ERBB2 in the SGC-7901 GC cell line was detected. The results (Fig. 8A and B) showed that, following the addition of paeonol, the protein expression of ERBB2 in the SGC-7901 cell line was significantly decreased as the concentration of paeonol increased. There was a dose-effect between the two. This suggested that ERBB2 was a potential target of paeonol in the SGC-7901 GC cell line.

Paeonol inhibits activation of the AKT signaling pathway

Following the phosphorylation of PI3K and the further activation of Akt, the Akt gene is activated by the phosphorylation of threonine at position 308 and serine at position 473. Akt and p-Akt in the GC SGC-7901 cell line were detected using western blot analysis. The results (Fig. 9) demonstrated that, following treatment with paeonol, the expression of p-Akt in the GC cells was significantly decreased, whereas no significant change was observed in the expression of Akt. The same was observed for PI3K and p-PI3K. This suggested that paeonol effectively inhibited the activation of the AKT signaling pathway.

Paeonol promotes cell apoptosis by inhibiting the nuclear transformation of NF-κB

The expression of p65 in the nucleus and cytoplasm was detected using western blot analysis. The results (Fig. 10A and B) indicated that, following treatment with paeonol in the GC cell lines, the protein expression of p65 was decreased in the nucleus and increased in the cytoplasm, which also indicated that paeonol inhibited the activation of the NF-κB signaling pathway. The expression of the downstream target gene Bcl-2 in the NF-κB pathway was also detected. The results indicated that, following treatment with different concentrations of paeonol, the gene expression of Bcl-2 was decreased, whereas the gene expression of Bax was increased; these differences were significant. These results suggested that paeonol regulated the ratio of Bax/Bcl-2 by affecting the NF-κB signaling pathway, thereby influencing the apoptosis of the cancer cells.

Discussion

At present, bioinformatics is one of the most common methods for identifying the target molecules of drugs. In the present study, it was found that the GC-related gene ERBB2 was associated with the target gene of paeonol by the STITCH database and Dijkstra's algorithm. ERBB2, also known as HER2, is a receptor of human epidermal growth factor. It has been demonstrated that the expression of PTGS2 is directly affected by the expression of HER2. Vadlamudi et al (26) found that the inhibition of HER2 resulted in a decrease in the expression of PTGS2, whereas the activation of HER2 activated the promoter of PTGS2 and promoted the expression of PTGS2. Of note, one of the direct target genes of paeonol in the treatment of tumor diseases is PTGS2; it was found that, following a decrease in the expression level of PTGS2, the viability of GC cells was markedly reduced (27). Following treatment of tumor cells with paeonol, the expression of PTGS2 was significantly reduced, thus the effect of paeonol on the tumor was achieved by inhibiting the expression of PTGS2 (28). These existing reports indicated that ERBB2 was likely to be a potential target of paeonol, which also confirmed the accuracy of the screening performed in the present study. Through examining the function of ERBB2 and its associated mechanism, it was possible to understand the way in which paeonol functions in the antitumor process.

Investigations on the mechanism of paeonol have shown that paeonol affects the expression of certain inflammation-related cytokines, including TNF-α, IL-1β and IL-6. For example, following treatment with paeonol, the expression levels of TNF-α, IL-1β and IL-6 were significantly decreased (2931). However, certain chronic inflammatory diseases can lead to cancer in certain tissues (32). For example, the chronic inflammatory response caused by asbestos has been associated with mesothelioma (33). These results suggest that the expression of inflammatory factors was directly associated with the development of tumors. Paeonol may exert an antitumor effect by influencing the inflammatory factors. Of note, activation of the NF-κB signaling pathway is one of the major factors involved in the normal expression of inflammatory cytokines. NF-κB is an inflammatory transcription factor and, when activated, the expression of inflammatory cytokines is increased (34). Studies have found that NF-κB can improve the transcription and expression of TNF-α and IL-1β, and TNF-α, IL-1β and IL-6 can significantly accelerate the development of cancer cells (35,36). In addition, it has been reported that NF-κB is the core causal factor in tumors resulting from inflammation (37). These results demonstrated that the activation of NF-κB is critical in the growth of cancer cells. Furthermore, it has been reported that NF-κB has a significant positive effect on the migration and proliferation of tumor cells (38,39). These previous reports indicated that paeonol may be involved in causing the death of cancer cells in the treatment of GC by acting on the NF-κB signaling pathway. However, whether there is a target of paeonol between paeonol and the NF-κB signaling pathway remained to be elucidated.

In the screening experiments described in the present study, it was found that ERBB2 may be a potential target of paeonol. The results of follow-up experiments showed that, following treatment of GC cells with paeonol, the protein content of ERBB2 was significantly reduced. The higher the concentration of paeonol, the lower the protein content of ERBB2. This suggested that the gene expression of ERBB2 was significantly affected by the negative regulation of paeonol, with a higher concentration of paeonol causing more marked negative regulation. The examination of ERBB2 in the present study showed that a high expression of ERBB2 increased the proliferation of cells and activated NF-κB (40). In addition, when the expression level of NF-κB was increased, NF-κB was able to bind with the promoter of ERBB2, which further promoted the expression of ERBB2 (41). On investigating the regulatory mechanism of ERBB2 on NF-κB, it was found that the PI3K/Akt pathways located downstream of ERBB2 were involved in the regulation of NF-κB activity (4244). The PI3K/Akt signaling pathways have been found to be involved in several cell responses, including cell proliferation and protein synthesis, which are closely associated with inflammation caused by viral infection (45,46). It was found that the increased gene expression of ERBB2 induced PI3K and Akt kinase activity, and increased the expression of NF-κB. The inhibition of PI3K and Akt led to a decrease in NF-κB activity (43). These reports suggested that ERBB2 had an activating effect on PI3K/Akt signaling pathways, which further activated downstream NF-κB by this activation. The activation of NF-κB caused activation of downstream inflammatory cytokines, ultimately affecting tumor cells. In the present study, following treatment of GC cells with paeonol, the GC cells showed marked cell death, the protein expression level of PI3K/Akt was significantly decreased, and NF-κB appeared to be inhibited. In addition, the protein content of apoptosis-promoting factor was increased, whereas the protein content of the inhibitor was decreased. These results demonstrated that paeonol inhibited ERBB2, and the inhibition of ERBB2 had an inhibitory effect on the PI3K/Akt signaling pathways, ultimately affecting NF-κB. This effect finally promoted the death of cancer cells.

Using bioinformatics methods, including the analysis of gene-drug associations and STRING analysis, the ERBB2 gene was screened from the GC-related genes. There was a close association between the ERBB2 gene and the known targets of paeonol, suggesting that ERBB2 may be a potential target of paeonol. Subsequently, cytology experiments showed that paeonol significantly inhibited the expression of ERBB2. The protein contents of PI3K/Akt and NF-κB downstream of ERBB2 were decreased. The above results suggested that paeonol affected NF-κB by regulating ERBB2, thereby exerting anticancer effects. Taken together, these results confirmed that ERBB2 is a target of paeonol. However, only SGC-7901 cells were used as a model to verify the results, and using additional cell lines may be beneficial to confirm the conclusions of the present study.

In conclusion, ERBB2, a GC-related gene predicted by bioinformatics, was expressed at high levels in GC tissues, compared with that in adjacent tissues. Paeonol significantly downregulated ERBB2, thereby inhibiting the activation of the AKT/PI3K pathways. The signal transduction of the nuclear translocation of NF-κB was also affected, which finally resulted in the upregulation of the downstream Bax gene inducing the death of SGC-7901 cells. This suggested that paeonol may affect SGC-7901 cells through the regulation of ERBB2.

Acknowledgments

The authors would like to acknowledge the helpful reviewer comments received on this manuscript.

Funding

This study was supported by the Natural Science Fund of Bengbu Medical College (grant no. BYKY1632ZD).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

JF and LY designed the study and collected the data. JL performed the statistical analysis. RH interpreted the data. BZ prepared the manuscript.

Ethics approval and consent to participate

The First Affiliated Hospital of Bengbu Medical College provided ethical approval for the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Global Burden of Disease Cancer Collaboration; Fitzmaurice C, Dicker D, Pain A, Hamavid H, Moradi-Lakeh M, MacIntyre MF, Allen C, Hansen G, Woodbrook R, et al: The global burden of cancer 2013. JAMA Oncol. 1:505–527. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T and Thun MJ: Cancer statistics, 2008. CA Cancer J Clin. 58:71–96. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Khushalani N: Cancer of the esophagus and stomach. Mayo Clin Proc. 83:712–722. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Zhang XY and Zhang PY: Gastric cancer: Somatic genetics as a guide to therapy. J Med Genet. 54:305–312. 2017. View Article : Google Scholar

5 

Toiyama Y, Tanaka K, Kitajima T, Shimura T, Imaoka H, Mori K, Okigami M, Yasuda H, Okugawa Y, Saigusa S, et al: Serum angiopoietin-like protein 2 as a potential biomarker for diagnosis, early recurrence and prognosis in gastric cancer patients. Carcinogenesis. 36:1474–1483. 2015.PubMed/NCBI

6 

Guo Y, Shi J, Zhang J, Li H, Liu B and Guo H: Polypeptide N-acetylgalactosaminyltransferase-6 expression in gastric cancer. Onco Targets Ther. 10:3337–3344. 2017. View Article : Google Scholar :

7 

Lee JC, Lee SY, Kim CY and Yang DH: Clinical utility of tumor marker cutoff ratio and a combination scoring system of preoperative carcinoembryonic antigen, carbohydrate antigen 19-9, carbohydrate antigen 72-4 levels in gastric cancer. J Korean Surg Soc. 85:283–289. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Tocchi A, Costa G, Lepre L, Liotta G, Mazzoni G, Cianetti A and Vannini P: The role of serum and gastric juice levels of carcinoembryonic antigen, CA19.9 and CA72.4 in patients with gastric cancer. J Cancer Res Clin Oncol. 124:450–455. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Zheng TH, Zhao JL and Guleng B: Advances in molecular biomarkers for gastric cancer. Crit Rev Eukaryot Gene Exp. 25:299–305. 2015. View Article : Google Scholar

10 

Weaver BA: How Taxol/paclitaxel kills cancer cells. Mol Biol Cell. 25:2677–2681. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Suzuki S, Sakurai K, Adachi K, Nagashima S, Hara Y, Amano S, Enomoto K and Makishima M: Examination of the response rate of paclitaxel and bevacizumab therapy for metastatic advanced breast cancer according to the lymphopenia grade. Gan To Kagaku Ryoho. 42:1249–1251. 2015.In Japanese. PubMed/NCBI

12 

Li H, Dai M and Jia W: Paeonol attenuates high-fat-diet-induced atherosclerosis in rabbits by anti-inflammatory activity. Planta Med. 75:7–11. 2009. View Article : Google Scholar

13 

Chunhu Z, Suiyu H, Meiqun C, Guilin X and Yunhui L: Antiproliferative and apoptotic effects of paeonol on human hepatocellular carcinoma cells. Anticancer Drugs. 19:401–409. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Zhang LH, Xiao PG and Huang Y: Recent progresses in pharmacological and clinical studies of paeonol. Zhongguo Zhong XI Yi Jie He Za Zhi. 16:187–190. 1996.In Chinese. PubMed/NCBI

15 

Sun GP, Wan X, Xu SP, Wang H, Liu SH and Wang ZG: Antiproliferation and apoptosis induction of paeonol in human esophageal cancer cell lines. Dis Esophagus. 21:723–729. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Sun GP, Wang H, Xu SP, Shen YX, Wu Q, Chen ZD and Wei W: Anti-tumor effects of paeonol in a HepA-hepatoma bearing mouse model via induction of tumor cell apoptosis and stimulation of IL-2 and TNF-alpha production. Eur J Pharmacol. 584:246–252. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Wan XA, Sun GP, Wang H, Xu SP, Wang ZG and Liu SH: Synergistic effect of paeonol and cisplatin on oesophageal cancer cell lines. Dig Liver Dis. 40:531–539. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Xu SP, Sun GP, Shen YX, Peng WR, Wang H and Wei W: Synergistic effect of combining paeonol and cisplatin on apoptotic induction of human hepatoma cell lines. Acta Pharmacol Sin. 28:869–878. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Wang F, Sun JY, Zhu YH, Liu NT, Wu YF and Yu F: MicroRNA-181 inhibits glioma cell proliferation by targeting cyclin B1. Mol Med Rep. 10:2160–2164. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Li N, Fan LL, Sun GP, Wan XA, Wang ZG, Wu Q and Wang H: Paeonol inhibits tumor growth in gastric cancer in vitro and in vivo. World J Gastroenterol. 16:4483–4490. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Sarada SK, Himadri P, Ruma D, Sharma SK, Pauline T and Mrinalini: Selenium protects the hypoxia induced apoptosis in neuroblastoma cells through upregulation of Bcl-2. Brain Res. 1209:29–39. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Laenen G, Thorrez L, Bornigen D and Moreau Y: Finding the targets of a drug by integration of gene expression data with a protein interaction network. Mol Biosyst. 9:1676–1685. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Ma W, Yang D, Gu Y, Guo X, Zhao W and Guo Z: Finding disease-specific coordinated functions by multi-function genes: Insight into the coordination mechanisms in diseases. Genomics. 94:94–100. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Ewald JA and Jarrard DF: Decreased Skp2 expression is necessary but not sufficient for therapy-induced senescence in prostate cancer. Transl Oncol. 5:278–287. 2012. View Article : Google Scholar : PubMed/NCBI

25 

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

26 

Vadlamudi R, Mandal M, Adam L, Steinbach G, Mendelsohn J and Kumar R: Regulation of cyclooxygenase-2 pathway by HER2 receptor. Oncogene. 18:305–314. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Elder DJ, Halton DE, Crew TE and Paraskeva C: Apoptosis induction and cyclooxygenase-2 regulation in human colorectal adenoma and carcinoma cell lines by the cyclooxygenase-2-selective non-steroidal anti-inflammatory drug NS-398. Int J Cancer. 86:553–560. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Li M, Tan SY and Wang XF: Paeonol exerts an anticancer effect on human colorectal cancer cells through inhibition of PGE2 synthesis and COX-2 expression. Oncol Rep. 32:2845–2853. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Chen N, Liu D, Soromou LW, Sun J, Zhong W, Guo W, Huo M, Li H, Guan S, Chen Z and Feng H: Paeonol suppresses lipopoly-saccharide-induced inflammatory cytokines in macrophage cells and protects mice from lethal endotoxin shock. Fundam Clin Pharmacol. 28:268–276. 2014. View Article : Google Scholar

30 

Fu PK, Wu CL, Tsai TH and Hsieh CL: Anti-inflammatory and anticoagulative effects of paeonol on LPS-induced acute lung injury in rats. Evid Based Complement Alternat Med. 2012:8375132012. View Article : Google Scholar : PubMed/NCBI

31 

Li X, Zhou Y, Yu C, Yang H, Zhang C, Ye Y and Xiao S: Paeonol suppresses lipid accumulation in macrophages via upregulation of the ATP-binding cassette transporter A1 and downregulation of the cluster of differentiation 36. Int J Oncol. 46:764–774. 2015. View Article : Google Scholar

32 

Demaria S, Pikarsky E, Karin M, Coussens LM, Chen YC, El-Omar EM, Trinchieri G, Dubinett SM, Mao JT, Szabo E, et al: Cancer and inflammation: Promise for biological therapy. J Immunother. 33:335–351. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Yang H, Bocchetta M, Kroczynska B, Elmishad AG, Chen Y, Liu Z, Bubici C, Mossman BT, Pass HI, Testa JR, et al: TNF-alpha inhibits asbestos-induced cytotoxicity via a NF-kappaB-dependent pathway, a possible mechanism for asbestos-induced oncogenesis. Proc Natl Acad Sci USA. 103:10397–10402. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Kumar A and Yu FS: Toll-like receptors and corneal innate immunity. Curr Mol Med. 6:327–337. 2006. View Article : Google Scholar : PubMed/NCBI

35 

Fan Y, Mao R and Yang J: NF-kappaB and STAT3 signaling pathways collaboratively link inflammation to cancer. Protein Cell. 4:176–185. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Gray GK, McFarland BC, Nozell SE and Benveniste EN: NF-κB and STAT3 in glioblastoma: Therapeutic targets coming of age. Expert Rev Neurother. 14:1293–1306. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Wang SW and Sun YM: The IL-6/JAK/STAT3 pathway: Potential therapeutic strategies in treating colorectal cancer (Review). Int J Oncol. 44:1032–1040. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Sethi G, Sung B and Aggarwal BB: Nuclear factor-kappaB activation: From bench to bedside. Exp Biol Med (Maywood). 233:21–31. 2008. View Article : Google Scholar

39 

Shishodia S and Aggarwal BB: Nuclear factor-kappaB activation: A question of life or death. J Biochem Mol Biol. 35:28–40. 2002.

40 

Galang CK, García-Ramírez J, Solski PA, Westwick JK, Der CJ, Neznanov NN, Oshima RG and Hauser CA: Oncogenic Neu/ErbB-2 increases ets, AP-1, and NF-kappaB-dependent gene expression, and inhibiting ets activation blocks Neu-mediated cellular transformation. J Biol Chem. 271:7992–7998. 1996. View Article : Google Scholar : PubMed/NCBI

41 

Cao N, Li S, Wang Z, Ahmed KM, Degnan ME, Fan M, Dynlacht JR and Li JJ: NF-kappaB-mediated HER2 overexpression in radiation-adaptive resistance. Radiat Res. 171:9–21. 2009. View Article : Google Scholar : PubMed/NCBI

42 

Guo G, Wang T, Gao Q, Tamae D, Wong P, Chen T, Chen WC, Shively JE, Wong JY and Li JJ: Expression of ErbB2 enhances radiation-induced NF-kappaB activation. Oncogene. 23:535–545. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Pianetti S, Arsura M, Romieu-Mourez R, Coffey RJ and Sonenshein GE: Her-2/neu overexpression induces NF-kappaB via a PI3-kinase/Akt pathway involving calpain-mediated degradation of IkappaB-alpha that can be inhibited by the tumor suppressor. PTEN Oncogene. 20:1287–1299. 2001. View Article : Google Scholar

44 

Zhou BP, Hu MC, Miller SA, Yu Z, Xia W, Lin SY and Hung MC: HER-2/neu blocks tumor necrosis factor-induced apoptosis via the Akt/NF-kappaB pathway. J Biol Chem. 275:8027–8031. 2000. View Article : Google Scholar : PubMed/NCBI

45 

Diehl N and Schaal H: Make yourself at home: Viral hijacking of the PI3K/Akt signaling pathway. Viruses. 5:3192–3212. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Tian J, Zhang X, Wu H, Liu C, Li Z, Hu X, Su S, Wang LF and Qu L: Blocking the PI3K/AKT pathway enhances mammalian reovirus replication by repressing IFN-stimulated genes. Front Microbiol. 6:8862015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 42 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fu J, Yu L, Luo J, Huo R and Zhu B: Paeonol induces the apoptosis of the SGC‑7901 gastric cancer cell line by downregulating ERBB2 and inhibiting the NF‑κB signaling pathway. Int J Mol Med 42: 1473-1483, 2018
APA
Fu, J., Yu, L., Luo, J., Huo, R., & Zhu, B. (2018). Paeonol induces the apoptosis of the SGC‑7901 gastric cancer cell line by downregulating ERBB2 and inhibiting the NF‑κB signaling pathway. International Journal of Molecular Medicine, 42, 1473-1483. https://doi.org/10.3892/ijmm.2018.3704
MLA
Fu, J., Yu, L., Luo, J., Huo, R., Zhu, B."Paeonol induces the apoptosis of the SGC‑7901 gastric cancer cell line by downregulating ERBB2 and inhibiting the NF‑κB signaling pathway". International Journal of Molecular Medicine 42.3 (2018): 1473-1483.
Chicago
Fu, J., Yu, L., Luo, J., Huo, R., Zhu, B."Paeonol induces the apoptosis of the SGC‑7901 gastric cancer cell line by downregulating ERBB2 and inhibiting the NF‑κB signaling pathway". International Journal of Molecular Medicine 42, no. 3 (2018): 1473-1483. https://doi.org/10.3892/ijmm.2018.3704