Open Access

PEA15 promotes liver metastasis of colorectal cancer by upregulating the ERK/MAPK signaling pathway

Retraction in: /10.3892/or.2023.8574

  • Authors:
    • Bo Tang
    • Wenjin Liang
    • Yong Liao
    • Zeming Li
    • Yan Wang
    • Chao Yan
  • View Affiliations

  • Published online on: October 25, 2018     https://doi.org/10.3892/or.2018.6825
  • Pages: 43-56
  • Copyright: © Tang 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

Liver metastasis is one of the major causes of death in patients with colorectal cancer, and although treatment has improved recently, the long‑term survival rate of patients has not improved significantly. In the present study, we used immunohistochemistry to determine that phosphoprotein enriched in astrocytes‑15 kDa (PEA15) was highly expressed in colorectal cancer tissues and liver metastatic cancer tissues. It was also highly expressed in metastatic colorectal cancer patients compared to non‑metastatic patients. Through clinicopathological data of patients with liver metastasis of colorectal cancer, we found that high expression of PEA15 was positively correlated with TNM staging, liver metastasis and poor prognosis of colorectal cancer patients. Using confocal immunofluorescence microscopy, western blotting and cell proliferation, migration and invasion assays, we also determined that PEA15 could promote cancer cell proliferation in vitro and in vivo, epithelial mesenchymal transition (EMT) and the characteristics of cancer stem cells in vitro, thus promoting the abilities of invasion and migration. In addition, we revealed that PEA15 promoted the liver metastasis of colorectal cancer cells in a xenograft tumor metastasis model. In addition, concerning the mechanism, we used gene chip analysis to determine that PEA15 upregulated the ERK/MAPK signaling pathway in colorectal cancer cells. Therefore, we concluded that PEA15 may be a potential biomarker for liver metastasis of colorectal cancer therapy. Collectively, PEA15 promoted the development of liver metastasis of colorectal cancer through the ERK/MAPK signaling pathway.

Introduction

Colorectal cancer is the third most common cancer in the world and its annual fatality rate ranks third (1,2). The main reason leading to the death of patients with colorectal cancer is distant invasion and metastasis such as liver, lung and bone metastasis, while liver metastasis of colorectal cancer is the most common (3). Although the diagnosis and treatment strategies for liver metastasis of colorectal cancer have greatly improved in recent years, to date, the overall survival rate of patients has not significantly improved. Therefore, exploring the mechanism of liver metastasis of colorectal cancer can provide effective guidance for potential clinical interventions, thus improving the long-term prognosis of patients.

PEA15 is a 15-kDa-sized small molecule protein containing an NH2 terminal death effector domain and a COOH terminal irregular structure distributed in the cytoplasm. Its DNA sequence is located on chromosome 1q21-22 and it is highly conserved in mammalian gene sequences (4,5). PEA-15 is widely expressed in human tissues and participates in the interaction between various proteins. It can regulate cell apoptosis, proliferation and glucose metabolism by acting on the key functions of cellular effectors in vivo (68). At present, it has been revealed that PEA-15, through its death-effector domain (DED) and ERK1/2-targeted binding, inhibited ERK1/2 phosphorylation and nucleus transfer, thus playing an anti-apoptotic role (9). Some studies have demonstrated that PEA15 is highly expressed in tumors such as liver (10), lung (11), breast (12) and ovarian cancer (13). However, it has a dual role in the regulation of tumors (14). It has been revealed that phosphorylation of PEA15 phosphorylated ERK and promoted the proliferation and invasion of hepatocellular carcinoma cells (10), whereas the unphosphorylated state of PEA15 inhibited the phosphorylation of ERK and EGFR, thus inhibiting proliferation, invasion and metastasis of breast (12) and ovarian cancer (13). However, whether PEA15 plays a role in liver metastasis of colorectal cancer is still unknown.

In the present study, we revealed that PEA15 was highly expressed in colorectal cancer tissues and metastatic liver tissues. High expression of PEA15 was positively correlated with TNM stage, liver metastasis and indicated a poor prognosis. In vitro and in vivo experiments confirmed that PEA15 promoted the proliferation, invasion, migration and EMT of colorectal cancer cells. To determine its mechanism, we used gene chip analysis and found that PEA15 regulated the ERK/MAPK signaling pathway to promote the invasion and migration of colorectal cancer cells. Concurrently, xenograft tumor experiments also confirmed that PEA15 promoted liver metastasis of colorectal cancer cells in vivo.

Materials and methods

Experimental reagents and antibodies

Lipofectamine 2000 liposomes and TRIzol reagent were purchased from Invitrogen (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Primary antibodies for PEA15 (cat. no. ab133217), PLD1 (cat. no. ab50695), ERK (cat. no. ab184699), p38 (cat. no. ab170099), MAP2K6 (cat. no. ab154901), JNK (cat. no. ab47337), PRKX (cat. no. ab109389) and FLNB (cat. no. ab97457) proteins were purchased from Abcam (Cambridge, MA, USA). Primary antibodies for E-cadherin (cat. no. #14472), N-cadherin (cat. no. 13116), vimentin (cat. no. 5741) and β-actin (cat. no. 3700) were purchased from Cell Signaling Technology (Danvers, MA, USA). Other reagents were purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany).

Patient and samples

The present study included 40 cases of colorectal cancer and corresponding clinicopathological data, which contained 19 cases of liver metastasis tissue specimens. These specimens were all paraffin specimens and obtained from the Department of General Surgery of the Affiliated Hospital of Guilin Medical University from August 2000 to August 2016 and all the specimens were clinically diagnosed by the Department of Pathology. All clinical cases and specimen tissues were reviewed and approved by the Ethics Committee of Guilin Medical University, and informed consents were signed by the patients according to the Declaration of Helsinki. Clinicopathological data included sex, age, tumor diameter, tumor differentiation, TNM staging and liver metastasis. Follow-up of patients was performed until March 2018.

Immunohistochemistry

The surgically resected paraffin specimens were collected from the Department of Pathology of the Affiliated Hospital of Guilin Medical University and were prepared into 4-mm thick slices. According to the EnVision two-step procedure (14), dewaxing and hydration were carried out, after EDTA buffer high pressure antigen retrieval. Subsequently a primary antibody against PEA15 (Abcam) with a dilution of 1:100 was added, and the specimens were incubated at room temperature for 1 h, then washed and incubated at room temperature for 0.5 h with a secondary antibody (MaxVision™ HRP-polymer anti-Rabbit IHC Kit, cat. no. KIT-5006, MXB Biotechnologies, Fuzhou, China) followed by washing with phosphate-buffered saline (PBS) buffer. A DAB Horseradish Peroxidase Color Development kit (Beyotime Institute of Biotechnology, Haimen, China) was used for color development and scored using the Olympus X71 inverted microscope (Olympus Corp., Tokyo, Japan). Based on the staining intensity, samples were divided into the following grades: 0, <10% of tumor cells were positively stained; 1+, 11–25% of tumor cells were positively stained; 2+, 26–50% of tumor cells were positively stained; and 3+, >50% of tumor cells were positively stained. Immunohistochemical analysis and scoring were conducted by two researchers, respectively.

Western blotting

Cell samples were collected and lysis buffer [50 mM Tris-HCl, 137 mM NaCl, 10% glycerol, 100 mM sodium orthovanadate, 1 mM phenylmethylsulfonyl fluoride (PMSF), 10 mg/ml aprotinin, 10 mg/ml leupeptin, 1% Nonidet P-40, and 5 mM protease inhibitor cocktail; pH 7.4] was added to extract total proteins. Then the protein concentration was assessed by BCA (bicinchoninic acid) kit (Beyotime Institute of Biotechnology) and the appropriate amount of loading buffer was added. Protein sample (30 µg) was added to 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis system (SDS-PAGE) for electrophoresis separation and then transferred to polyvinylidene difluoride (PVDF) membranes (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Then, 5% non-fat milk was used to incubate the PVDF membranes, and peroxidase-labeled secondary antibodies (HRP-labeled goat anti-rabbit IgG; cat. no. A0208; Beyotime Institute of Biotechnolgy) at a 1:10,000 were used for incubation for 1 h at room temperature. Subsequently, chemiluminescence was used to display the imprinting, and the results were analyzed using the Tannon 5200 chemiluminescent imaging system (Tanon Science and Technology, Shanghai, China).

Quantitative reverse transcription polymerase chain reaction (qRT-PCR)

The collected cells and tissues were added to TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.) and total RNA was extracted according to the RNAiso Plus kit instructions (Takara Bio, Inc., Otsu, Japan). The RNA concentration was assessed using the NanoDrop spectrophotometer (Thermo Fisher Scientific, Inc.). The FastQuant cDNA First-Strand Synthesis kit (Tiangen Biotech, Co., Ltd., Beijing, China) was used for reverse transcription. A qRT-PCR kit FastStart Universal SYBR Green Master Mix (Rox) (Roche Diagnostics GmbH, Mannheim, Germany) was used to produce the reaction system and amplification according to the manufacturer's instructions. The reaction cycling conditions were performed as follows: 1 cycle at 95°C for 15 min, followed by 40 cycles at 95°C for 10 sec and at 60°C for 60 sec, and then relative mRNA expression was analyzed using the comparative quantification cycle (Cq) method followed by normalization to β-actin expression (Applied Biosystems; Thermo Fisher Scientific, Inc.) (15). The primers used for amplification were as follows: PEA15 forward, 5′-GTTCTGTAGTCAACCACCAT-3′ and reverse, 5′-ACCAACAACATCACCCTT-3′; β-actin forward, 5′-TCTTCATGGTGCTGGGAG-3′) and reverse, 5′-AATGAGCGGTTCCGTTGC-3′ which was used as the internal control.

Cell culture and cell transfection

Colorectal cancer cells (HT-29, Caco-2, COLO 205, HCT-116, RKO, SW-620 and SW-480) were purchased from the Cell Bank of Chinese Academy of Sciences (Shanghai, China). Dulbecco's modified Eagle's medium (DMEM; Life Technologies; Thermo Fisher Scientific, Inc.) was used for HT-29, Caco-2, HCT-116 and RKO and Roswell Park Memorial Institute-1640 medium (RPMI-1640) (Life Technologies; Thermo Fisher Scientific, Inc.) was used for COLO 205, SW-620 and SW-480. All cell culture media contained 10% fetal calf serum (FCS; Life Technologies; Thermo Fisher Scientific, Inc.) and were cultured in a 37°C incubator with 5% CO2/95% air. The procedure aforementioned was used for the preparation and transfection of ERK1/2 siRNA and ERK1/2 plasmid (16). For primary colorectal cancer cells, 16 colorectal cancer specimens (aged 35–70 years) were collected from patients who underwent colorectal cancer resection at the Affiliated Hospital of Guilin Medical University from October 2014 to October 2016. Specimens were obtained with the approval of the Medical Ethics Committee of the Affiliated Hospital of Guilin Medical University. Fresh tumor tissue was cut and incubated with type IV collagenase (Sigma-Aldrich; Merck KGaA) for 5–10 min at 37°C and a single cell suspension was filtered using a 100-mm cell strainer (BD Biosciences, Franklin Lakes, NJ, USA). Cell viability was detected via staining using Trypan blue (Sigma-Aldrich; Merck KGaA), and CD45+ cells in fresh tumor tissue were removed using the CD45 removal kit (Depletion kit; Miltenyi Biotec, Bergisch Gladbach, Germany).

Cell proliferation experiments

The prepared cell suspension was seeded in a 96-well plate (100 µl/well) with a cell density of 2×103 cells/well, and 10 µl of Cell Counting kit-8 (CCK-8) solution (Dojindo Laboratories, Kumamoto, Japan) was added to each well after the predefined different culture time. The plates were placed in an incubator for ~1 h and then placed in a microplate reader (SpectraMax plus 384; Molecular Devices, San Jose, CA, USA) to assess the absorbance at 450 nm.

Plate cloning experiment

The prepared cell suspension was seeded on a 6-well plate at 600 cells/well. Cell colonies were formed after being cultured for ~2 weeks. Cells were fixed with 4% paraformaldehyde for 20 min and stained overnight with 1% crystal violet (cat. no. G1062; Solarbio, Tokyo, Japan). After three washes in PBS, the images of the results were observed and captured using a fluorescence microscope (IX71; Olympus Corporation) and the number of colonies/well was calculated using ImageJ (National Institutes of Health, Bethesda, MD, USA).

Experiments of cell invasion and migration

A Transwell chamber (8 µm; BD Biosciences) (the chamber for invasion detection was coated with Matrigel, and the chamber for migration detection was without Matrigel) was placed into a 24-well plate with ~600 µl medium containing 10% FBS which was added to the bottom chamber. The prepared cell suspension was seeded in a Τranswell chamber at 1,000 cells/well. One or two days later, the cell preparation in the small chamber was observed under a fluorescence microscope (IX71; Olympus Corporation). After an appropriate number of cells had passed through the cell pores, the cells were fixed with 4% paraformaldehyde and stained with 1% crystal violet (cat. no. G1062; Solarbio) and images were captured.

Confocal immunofluorescence microscopy

Cells were seeded on a special slide (Costar, Manassas, VA, USA). After 24 h, the cells on the slides were washed with PBS and 4% paraformaldehyde was added to fix the cells. After the cell membrane was permeabilized with 0.5% Triton X-100, the cells were blocked by adding 10% FBS for 30 min at room temperature. The primary antibody was added for incubation overnight at 4°C, and then the cells were washed three times with PBS, followed by the addition of a fluorescein isothiocyanate (FITC)-labeled secondary antibody (cat. no. A-21063; Invitrogen; Thermo Fisher Scientific, Inc.) and incubation for 1 h in a dark chamber. After washing three times with PBS, the cells were stained with DAPI for 5 min and analyzed with the Carl Zeiss Confocal Imaging System (LSM 780; Carl Zeiss, Jena, Germany).

Detection of colorectal cancer stem cell characteristics

A prepared primary colorectal cancer single cell suspension was counted (Luna II; Logos Biosystems, Anyang, Korea), and 1×106 cells were added into tumor stem cell culture medium suspension for culture, and shaken 1 time every 6 h on the 1st and 2nd days to prevent cell adherence. When the cell spheres became larger and the brightness decreased, the structure was loosened. Accutase enzyme (Thermo Fisher Scientific, Inc.) was used for digestion passage. The parental cells and the passaged cells were cultured in a 6-well plate at 1×105 cells/well for approximately 6 days, the conglomeration rate was calculated. The surface antigen (EPCAM and CD133) of the primary colorectal cancer cells was sorted by flow cytometry, and the proportion of side population cells was counted and analyzed.

Construction of cell lines of stably upregulated and downregulated PEA15

Construct retroviruses were coated with vector pBabe-puro-PEA15 and with vector pSuper-retro- puro-PEA15-shRNA. The extraction of retroviruses and transfection of tumor cells were performed as previously described (17) and the expression of PEA15 was verified using qRT-PCR and western blotting.

In vivo tumor growth and metastasis

Forty BALB/c 8-week-old male nude mice weighing 18 g-20 g (20 were used for tumor growth while the other 20 were used for a tumor metastasis model) were purchased from the Animal Experimental Center of Guilin Medical University and all animals were used in accordance with the operating instructions and approved by the Animal Care Committee of Guilin Medical University, with room temperature atmosphere and ad libitum. Tumor cell lines in logarithmic growth phase were collected and washed with PBS to prepare a cell suspension. The prepared cell suspension (2×106 cells/ml) was subcutaneously injected into the right groin area of 8-week-old nude mice. The tumor volume was assessed weekly. Nude mice were sacrificed by cervical dislocation. Twenty-eight days after surgery, the tumors were excised and the tumor weights were recorded. For the tumor metastasis model, the cells were collected and washed with PBS and then prepared into a cell suspension. The cell suspension with a concentration of 1×107 cells/ml was injected into the hepatic portal vein of nude mice. All mice were sacrificed by cervical dislocation 60 days after inoculation.

Gene chip analysis

The quality of total RNA was assessed using the Agilent 2100 Bioanalyzer (Agilent Technologies, Inc., Santa Clara, CA, USA) and the NanoDrop ND-1000 spectrophotometer. RNA expression was then analyzed using the Affymetrix HU U133 plus 2.0 array according to the manufacturer's instructions. The raw data was normalized by the Robust Multiarray Average (RMA) of expression control platform software (Affymetrix; Thermo Fisher Scientific, Inc.). Scatter plot analysis of the gene with significant changes in PEA15 in the downregulation group and the control group to screen the genes with upregulation or downregulation of expression ≥3 times was performed. Gene Cluster v3.0 software cluster (Stanford University, Stanford, CA, USA) was used for cluster analysis, and Java TreeView v1.1.4r3 software (Alok Saldanha; http://jtreeview.sourceforge.net/) was used for thermal image visualization.

Statistical analysis

Experimental data were statistically analyzed using GraphPad Prism 5 (GraphPad Software, Inc., San Diego, CA, USA) and SPSS v.18.0 (SPSS Inc., Chicago, IL, USA). The Chi-square test (χ2) was used to evaluate the association between the expression of PEA15 and the clinicopathological features of the patients. Comparisons between different groups were undertaken using the Student's two-tailed t-test. Correlation between PEA15 expression with BRAF and RAS mutation in colon tissues were assessed using Spearman's rank correlation test. Multigroup comparisons of the means were carried out by one-way analysis of variance (ANOVA) test with post hoc contrasts by Student-Newman-Keuls test. The Kaplan-Meier method was used to estimate the correlation between the expression of PEA15 and the 5-year survival rate of patients after diagnosis. The log-rank test was used to compare the survival curves of different groups. P<0.05 was considered to indicate a statistically significant difference.

Results

PEA15 is highly expressed in colorectal cancer and metastatic liver cancer tissues and positively correlated with tumor TNM stage

We randomly collected 40 cases of colorectal cancer and the corresponding liver metastasis tissues with immunohistochemical detection. The results revealed that the expression level of PEA15 in colorectal cancer and metastatic liver cancer tissues was significantly higher than the corresponding paracancerous tissues. In addition, the expression level of PEA15 in liver cancer tissues was significantly higher than that in non-metastatic liver cancer tissues (Fig. 1A-C). Notably, we found that the expression of PEA15 was also significantly increased in endangium of liver metastasis (Fig. 1Aa-f), indicating that PEA15 may be involved in the hematogenous metastasis of tumors. Combined with clinicopathological data of patients with liver metastasis of colorectal cancer, we analyzed the correlation between the expression of PEA15 and the clinicopathological features of patients. After mapping the ROC curve of PEA15 expression (Fig. 1D) and determining the division point of high or low PEA15 expression, we divided the patients into a PEA15-high expression group and a low expression group. The statistical results revealed that the expression of PEA15 was correlated with TNM stage (P<0.05) and liver metastasis (P<0.001) (Table I).

Table I.

Correlations between PEA15 expression and clinicopathological parameters in 40 colorectal carcinoma patients.

Table I.

Correlations between PEA15 expression and clinicopathological parameters in 40 colorectal carcinoma patients.

PEA15 expression

VariablesLow (21)High (19)TotalP-value
Age (years) 0.548
  <409 (47)10 (53)19
  ≥4012 (57)9 (43)21
Sex 0.125
  Male7 (39)11 (61)18
  Female14 (64)8 (36)22
Tumor diameter (cm) 0.062
  <55 (33)10 (67)15
  ≥516 (64)9 (36)25
Tumor differentiation 0.292
  Well3 (33)6 (67)9
  Moderate8 (50)8 (50)16
  Poor10 (67)5 (33)15
TNM stage 0.028a
  I–II6 (33)12 (67)18
  III–IV15 (68)7 (32)22
Liver metastasis <0.001a
  No17 (81)4 (19)21
  Yes4 (21)15 (79)19

a Significant P-value.

PEA15 is positively correlated with the clinical prognosis of patients with liver metastasis of colorectal cancer and the BRAF mutation

Through follow-up of 5-year survival time with patients after being clinically diagnosed, a Kaplan-Meier survival curve was used to analyze the correlation between the expression level of PEA15 and clinical prognosis, and the overall survival time (median, 13 vs. 32 months; P<0.001) and disease-free survival time (median, 11 months vs. 28 months; P<0.05) of patients with liver metastasis of colorectal cancer and low PEA15 expression were significantly better than those of the PEA15-overexpression group (Fig. 1E and F). In addition, in patients with liver metastatis, the total survival time (median, 13 months vs. 15 months, P<0.01) of the PEA15-low expression group was longer than that of the PEA15-high expression group (Fig. 1G). Multivariate Cox regression analysis revealed that PEA15 overexpression (relative risk=3.698; P=0.016) could be used as a relatively independent predictor of poor prognosis in patients with liver metastases of colorectal cancer (Table II). Furthermore, we detected and analyzed the correlation between the high expression of PEA15 and the mutation rates of RAS and BRAF in colorectal cancer tissues. The results revealed that the high expression of PEA15 was positively correlated with BRAF mutation but not with RAS mutation (Fig. 2A and B).

Table II.

Multivariate analysis with a Cox proportional hazards regression model.

Table II.

Multivariate analysis with a Cox proportional hazards regression model.

Univariate analysisMultivariate analysis


VariablesRR95% CIP-valueRR95% CIP-value
Age (years) (<40 vs. >40)0.5210.420–0.6470.7600.6710.520–0.8640.078
Sex (male vs. female)1.6381.318–2.0340.7820.7530.592–0.9570.082
Tumor diameter (cm) (<5 vs. >5)1.5091.217–1.8720.3253.3372.599–4.2840.095
Tumor differentiation (well vs. poor)1.1440.616–2.1240.3700.5440.421–0.7030.061
TNM stage (I–II vs. III–IV)0.8070.657–0.9910.041a0.6330.499–0.8030.003a
Liver metastasis (No vs. yes)7.7585.598–10.75 <0.001a7.4105.181–10.60.001a
PEA15 (Positive vs. negative)4.4343.356–5.858 <0.001a3.6982.636–5.1890.016a

{ label (or @symbol) needed for fn[@id='tfn2-or-41-01-0043'] } RR, relative risk; CI, confidence interval.

a Significant P-value.

PEA15 promotes the proliferation of colorectal cancer cells in vivo and in vitro

To investigate the regulatory effect of PEA15 on colorectal cancer cell lines, we first selected seven colorectal cancer cell lines (HT-29, Caco-2, HCT-116, RKO, COLO-205, SW-620 and SW-480). The expression of PEA15 in different cell lines was detected by PCR and western blotting, respectively. The results revealed that the expression level of PEA15 was significantly increased in colorectal cancer cell lines with metastatic potential (Fig. 3A-C). To evaluate the role of PEA15 in cell proliferation and tumor growth, we selected the HT-29 and RKO cell lines that produce PEA15 at high levels and proceeded to knockdown its expression using short hairpins RNAs (PEA15-shRNA plasmid). We also selected the SW-480 cell line to transfect with the PEA15 plasmid as aforementioned (Fig. 3D and E). The plate cloning results revealed that the colony formation of HT-29 and RKO cells after PEA15 downregulation was significantly less than that of the normal and negative control group (Fig. 4A and B), but the colony formation of SW-480 cells after PEA15 upregulation was significantly more than that of the normal and negative control group (Fig. 4C). The CCK-8 kit assay results revealed that the proliferation ability of HT-29 and RKO cells was significantly weakened after PEA15 downregulation (Fig. 4D and E) compared with normal group and negative control group, while the proliferation of SW-480 cells increased after PEA15 upregulation (Fig. 4F). In vivo, it was also confirmed that downregulation of PEA15 significantly reduced the tumor weight in nude mice, whereas upregulation of PEA15 increased the tumor weight (Fig. 4G and H).

PEA15 promotes the invasion and migration of colorectal cancer cells in vitro and participates in the regulation of EMT

According to Transwell chamber invasion and migration experiments, the invasion and migration abilities of HT-29 and RKO cells was evidently decreased after downregulation of PEA15 (Fig. 5A and B), while in SW-480 cells, these abilities were enhanced after PEA15 was upregulated (Fig. 5C). Subsequently, we examined the effect of PEA15 on the expression of EMT-related proteins in colorectal cancer cells by immunofluorescence confocal microscopy and western blotting. The results revealed that after PEA15 downregulation, the expression of E-cadherin in HT-29 and RKO cells increased, while the expression of N-cadherin and vimentin decreased (Fig. 6A and B); while in SW-480 cells, the opposite was observed after PEA15 was downregulated (Fig. 6C). The aforementioned results indicated that PEA15 promoted the invasion and migration of tumor cells by regulating the EMT of colorectal cancer cells.

PEA15 induces stem cell-like features of colorectal cancer cells

In order to investigate whether PEA15 regulated the stem cell characteristics of colorectal cancer cells, we detected the ability of spheroid formation of SW-480 cells and RKO cells by upregulation or downregulation of PEA15. The results revealed that with the upregulation of PEA15, the ability of spheroid formation of SW-480 cells and RKO cells was enhanced. In contrast, the ability of spheroid formation of both cells weakened after PEA15 downregulated (Fig. 7A). Subsequently, we used flow cytometry to detect the proportion of surface cancer antigen marker molecules (EPCAM and CD133) of the colorectal cancer cells and the ratio of side population cells. The experimental results revealed that the high expression of PEA15 in colorectal cancer cells exhibited more of EPCAM+ and CD133+ (Fig. 7B). Side population (SP) cell ratio analysis revealed that the proportion of SP increased in SW-480 cells after PEA15 was upregulated, whereas the proportion of SP in RKO cells decreased after PEA15 was downregulated (Fig. 7C). Therefore, the aforementioned results indicated that PEA15 could regulate the stem cell characteristics of colorectal cancer cells.

PEA15 promotes the development of colorectal cancer by regulating the MAPK signaling pathway

In order to further explore the mechanism of PEA15 in the regulation of liver metastasis of colorectal cancer, we compared the gene expression differences of HT-29 cells between the control group and the PEA15-downregulation group by gene chip analysis (the data is not currently available in GEO). Scatter plots were used to analyze the significantly changed genes in the PEA15-downregulation group and the control group. More than 400 genes with upregulation or downregulation change of more than 3 times were selected. Gene microarray analysis revealed that these genes were involved in a variety of signaling pathways, including the MAPK signaling pathway, the TGF-β signaling pathway, the PI3K/AKt signaling pathway, the stem cell regulatory pathway and the HIF-1 signaling pathway, while the most noticeable difference gene change was in the MAPK signaling pathway (Fig. 8A). These results indicated that PEA15 affected the development of colorectal cancer cells mainly through the regulation of the MAPK signaling pathway. We further detected the expression of MAPK signaling pathway-related proteins before and after PEA15 downregulation by western blotting. The results revealed that the expression of PEA15, PLD1, ERK, MAP2K6, c-Myc and PRKX proteins decreased and the expression of FLNB protein increased in RKO cells after downregulation of PEA15, while the expression of p38 and JNK proteins exhibited no significant changes (Fig. 8B). Furthermore, we downregulated ERK1/2 and found that the proliferation, invasion and migration abilities of RKO cells were all suppressed (Fig. 9A and B). Moreover, the results revealed that the expression of MAP2K6, c-Myc and PRKX proteins decreased while the expression of FLNB protein increased in RKO cells after downregulation of ERK1/2 (Fig. 9C). The aforementioned results indicated that PEA15 regulated the development of colorectal cancer by regulating the ERK/MAPK signaling pathway.

PEA15 promotes the formation of metastases of colorectal cancer cells in vivo

In order to verify the effect of PEA15 on the metastatic potential of colorectal cancer cells in vivo, we upregulated PEA15 in the RKO cell line and injected it into the hepatic portal vein of nude mice. After 60 days of inoculation, nude mice were sacrificed by cervical dislocation, and the liver metastasis foci were recorded. The results revealed that the foci number of liver metastases significantly increased after PEA15 upregulation (Fig. 10A and B). Meanwhile, immunohistochemistry revealed that the expression of PEA15 in liver tissues was significantly higher in the PEA15-upregulated colorectal cancer cell group (Fig. 10C). The aforementioned results confirmed that PEA15 promoted the development of liver metastasis of colorectal cancer by regulating the ERK/MAPK signaling pathway (Fig. 11).

Discussion

A previous study revealed that PEA15 is a key member of the anti-apoptotic family that inhibits apoptosis by restraining the formation of the death-inducing signal complex (DISC) and blocking the cascade activation of caspases (18). The present study confirmed that PEA15 promoted the proliferation of colorectal cancer cells in vitro and in vivo. Notably, we also found that the expression of PEA15 in colorectal cancer with liver metastasis was significantly higher than that without metastasis. In addition, the clinical TNM stage and BRAF gene mutation were positively correlated with PEA15 expression. These results indicated that PEA15 may be involved in the regulation of liver metastasis of colorectal cancer.

Current studies have revealed that EMT of colorectal cancer cells is a key factor in the distant metastasis of colorectal cancer (19). The present study revealed that when PEA15 was downregulated, the expression of E-cadherin in colorectal cancer cells increased, however, V-cadherin and vimentin decreased, and the invasion and migration capacities of colorectal cancer cells were significantly weakened. Tumor stem cell characterization test results also revealed that with downregulation of PEA15, the ability of cell formation became weaker and the proportion of side population cells decreased. While with upregulation of PEA15, the opposite results were obtained. Therefore, we conclude that PEA15 regulated EMT to promote liver metastasis of colorectal cancer and induced stem cell characteristics of colorectal cancer cells.

Current studies have also found that the MAPK signaling pathway may be involved in the regulation of colorectal cancer of liver metastasis (20). The MAPK signaling pathway is one of the most important regulatory pathways, and its signal-related proteins are widely distributed in the nucleus, cytoplasm, mitochondria and Golgi participating in regulating the various life activities of the body (21). The ERK/MAPK signaling pathway can regulate cell proliferation and invasion, promote the EMT process and finally promote the development of colorectal cancer (22,23). However, phosphorylation of two sites, Ser-104 and Ser-116 in PEA15 can promote tumor invasion and migration (24). The present study revealed that the expression of PEA15, PLD1, ERK, MAP2K6 and PRKX was downregulated, and the expression of FLNB was upregulated, while the expression of p38 and JNK was not markedly changed after PEA15 downregulation. In addition, downregulation of ERK1/2 supressed the proliferation, invasion and migration abilities of RKO cells. Therefore, we concluded that PEA15 regulated the ERK/MAPK signaling pathway. Research has shown that phosphorylated PEA15 can phosphorylate ERK in combination with PLD1, thereby exerting a tumor-promoting effect (25). In gliomas, Ser116 phosphorylation of PEA-15 regulates the ERK/MAPK signaling axis and antagonizes glioma cell death due to glucose deficiency (26). In addition, PEA15 could regulate PRKX to promote tumor epithelial cell proliferation, invasion and vascular structure formation (27). Our results also revealed that the expression of PLD1 and PRKX proteins decreased when PEA15 was downregulated, and PEA15 expression was positive in vascular endothelium in metastatic liver tissues. These results confirmed that PEA15 promoted the invasion and migration of colorectal cancer cells through the ERK/MAPK signal axis, and promoted EMT of colorectal cancer cells and generation of stem cell characteristics, thereby contributing to the occurrence of liver metastasis of colorectal cancer.

In summary, the present study confirmed that PEA15 is highly expressed in colorectal and metastatic liver tissues. We also determined for the first time that high expression of PEA15 was positively correlated with tumor TNM stage and liver metastasis, and acted as a relatively independent predictor of poor prognosis in patients with liver metastasis of colorectal cancer. Concurrently, the present study revealed that PEA15 not only regulated proliferation ability, but also the invasion and migration capacities, as well as EMT characteristics, and induced the stem cell characteristics of cancer cells. In relation to its mechanism, we determined for the first time that the PEA15/ERK/MAPK signaling pathway induced invasion and migration by regulating the EMT of colorectal cancer cells and induced tumor stem cell characteristics.

Acknowledgements

We wish to particularly acknowledge the patients enrolled in this study for their participation, and the Department of Pathology and Physiopathology, Guilin Medical University, for its collaboration in providing the human samples and the clinical information used in this study with the appropriate ethics approval.

Funding

The present study was supported in part by The National Natural Science Foundation of China (no. 81560393), the Guangxi Science Fund for Distinguished Young Scholars Program (no. 2016GXNSFFA380003), the Natural Science Foundation of Guangxi (no. 2015jjDA40010) and the Scientific Research and Technology Development Project for Guilin (no. 20140310-2-2).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

BT, YW and CY conceived and designed the experiments. BT and WL performed the experiments. BT, WL, YL, and ZL analyzed the data. YW, CY supervised the whole experimental work and revised the manuscript. BT and WL wrote the paper. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

All clinical cases and specimen tissues were reviewed and approved by the Ethics Committee of Guilin Medical University, and informed consents were signed by the patients according to the Declaration of Helsinki. The animal experiments were approved by the Animal Care Committee of Guilin Medical University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Wu Z, Wei D, Gao W, Xu Y, Hu Z, Ma Z, Gao C, Zhu X and Li Q: TPO-induced metabolic reprogramming drives liver metastasis of colorectal cancer CD110+ tumor-initiating cells. Cell Stem Cell. 17:47–59. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Renault F, Formstecher E, Callebaut I, Junier MP and Chneiweiss H: The multifunctional protein PEA-15 is involved in the control of apoptosis and cell cycle in astrocytes. Biochem Pharmacol. 66:1581–1588. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Danziger N, Yokoyama M, Jay T, Cordier J, Glowinski J and Chneiweiss H: Cellular expression, developmental regulation, and phylogenic conservation of PEA-15, the astrocytic major phosphoprotein and protein kinase C substrate. J Neurochem. 64:1016–1025. 1995. View Article : Google Scholar : PubMed/NCBI

6 

Farina B, Doti N, Pirone L, Malgieri G, Pedone EM, Ruvo M and Fattorusso R: Molecular basis of the PED/PEA15 interaction with the C-terminal fragment of phospholipase D1 revealed by NMR spectroscopy. Biochim Biophys Acta. 1834:1572–1580. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Nagarajan A, Dogra SK, Liu AY, Green MR and Wajapeyee N: PEA15 regulates the DNA damage-induced cell cycle checkpoint and oncogene-directed transformation. Mol Cell Biol. 34:2264–2282. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Fiory F, Formisano P, Perruolo G and Beguinot F: Frontiers: PED/PEA-15, a multifunctional protein controlling cell survival and glucose metabolism. Am J Physiol Endocrinol Metab. 297:E592–E601. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Mace PD, Wallez Y, Egger MF, Dobaczewska MK, Robinson H, Pasquale EB and Riedl SJ: Structure of ERK2 bound to PEA-15 reveals a mechanism for rapid release of activated MAPK. Nat Commun. 4:16812013. View Article : Google Scholar : PubMed/NCBI

10 

Quintavalle C, Hindupur SK, Quagliata L, Pallante P, Nigro C, Condorelli G, Andersen JB, Tagscherer KE, Roth W, Beguinot F, et al: Phosphoprotein enriched in diabetes (PED/PEA15) promotes migration in hepatocellular carcinoma and confers resistance to sorafenib. Cell Death Dis. 8:e31382017. View Article : Google Scholar : PubMed/NCBI

11 

Zanca C, Garofalo M, Quintavalle C, Romano G, Acunzo M, Ragno P, Montuori N, Incoronato M, Tornillo L, Baumhoer D, et al: PED is overexpressed and mediates TRAIL resistance in human non-small cell lung cancer. J Cell Mol Med. 12:2416–2426. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Shin M, Lee KE, Yang EG, Jeon H and Song HK: PEA-15 facilitates EGFR dephosphorylation via ERK sequestration at increased ER-PM contacts in TNBC cells. FEBS Lett. 589:1033–1039. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Bartholomeusz C, Rosen D, Wei C, Kazansky A, Yamasaki F, Takahashi T, Itamochi H, Kondo S, Liu J and Ueno NT: PEA-15 induces autophagy in human ovarian cancer cells and is associated with prolonged overall survival. Cancer Res. 68:9302–9310. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Pinato DJ, Tan TM, Toussi ST, Ramachandran R, Martin N, Meeran K, Ngo N, Dina R and Sharma R: An expression signature of the angiogenic response in gastrointestinal neuroendocrine tumours: Correlation with tumour phenotype and survival outcomes. Br J Cancer. 110:115–122. 2014. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

Li F and Mahato RI: RNA interference for improving the outcome of islet transplantation. Adv Drug Deliv Rev. 63:47–68. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Sotelo J, Esposito D, Duhagon MA, Banfield K, Mehalko J, Liao H, Stephens RM, Harris TJ, Munroe DJ and Wu X: Long-range enhancers on 8q24 regulate c-Myc. Proc Natl Acad Sci USA. 107:3001–3005. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Urosevic J, Garcia-Albéniz X, Planet E, Real S, Céspedes MV, Guiu M, Fernandez E, Bellmunt A, Gawrzak S, Pavlovic M, et al: Colon cancer cells colonize the lung from established liver metastases through p38 MAPK signalling and PTHLH. Nat Cell Biol. 16:685–694. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Loboda A, Nebozhyn MV, Watters JW, Buser CA, Shaw PM, Huang PS, Van't Veer L, Tollenaar RA, Jackson DB, Agrawal D, et al: EMT is the dominant program in human colon cancer. BMC Med Genomics. 4:92011. View Article : Google Scholar : PubMed/NCBI

20 

Formisano P, Ragno P, Pesapane A, Alfano D, Alberobello AT, Rea VE, Giusto R, Rossi FW, Beguinot F, Rossi G and Montuori N: PED/PEA-15 interacts with the 67 kD laminin receptor and regulates cell adhesion, migration, proliferation and apoptosis. J Cell Mol Med. 16:1435–1446. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Mor A and Philips MR: Compartmentalized Ras/MAPK signaling. Annu Rev Immunol. 24:771–800. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Fang JY and Richardson BC: The MAPK signalling pathways and colorectal cancer. Lancet Oncol. 6:322–327. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Gu Y, Wang Q, Guo K, Qin W, Liao W, Wang S, Ding Y and Lin J: TUSC3 promotes colorectal cancer progression and epithelial-mesenchymal transition (EMT) through WNT/β-catenin and MAPK signalling. J Pathol. 239:60–71. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Lee J, Bartholomeusz C, Krishnamurthy S, Liu P, Saso H, Lafortune TA, Hortobagyi GN and Ueno NT: PEA-15 unphosphorylated at both serine 104 and serine 116 inhibits ovarian cancer cell tumorigenicity and progression through blocking β-catenin. Oncogenesis. 1:e222012. View Article : Google Scholar : PubMed/NCBI

25 

Sulzmaier F, Opoku-Ansah J and Ramos JW: Phosphorylation is the switch that turns PEA-15 from tumor suppressor to tumor promoter. Small GTPases. 3:173–177. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Eckert A, Böck BC, Tagscherer KE, Haas TL, Grund K, Sykora J, Herold-Mende C, Ehemann V, Hollstein M, Chneiweiss H, et al: The PEA-15/PED protein protects glioblastoma cells from glucose deprivation-induced apoptosis via the ERK/MAP kinase pathway. Oncogene. 27:1155–1166. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Li X, Iomini C, Hyink D and Wilson PD: PRKX critically regulates endothelial cell proliferation, migration, and vascular-like structure formation. Dev Biol. 356:475–485. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2019
Volume 41 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tang B, Liang W, Liao Y, Li Z, Wang Y and Yan C: PEA15 promotes liver metastasis of colorectal cancer by upregulating the ERK/MAPK signaling pathway Retraction in /10.3892/or.2023.8574. Oncol Rep 41: 43-56, 2019
APA
Tang, B., Liang, W., Liao, Y., Li, Z., Wang, Y., & Yan, C. (2019). PEA15 promotes liver metastasis of colorectal cancer by upregulating the ERK/MAPK signaling pathway Retraction in /10.3892/or.2023.8574. Oncology Reports, 41, 43-56. https://doi.org/10.3892/or.2018.6825
MLA
Tang, B., Liang, W., Liao, Y., Li, Z., Wang, Y., Yan, C."PEA15 promotes liver metastasis of colorectal cancer by upregulating the ERK/MAPK signaling pathway Retraction in /10.3892/or.2023.8574". Oncology Reports 41.1 (2019): 43-56.
Chicago
Tang, B., Liang, W., Liao, Y., Li, Z., Wang, Y., Yan, C."PEA15 promotes liver metastasis of colorectal cancer by upregulating the ERK/MAPK signaling pathway Retraction in /10.3892/or.2023.8574". Oncology Reports 41, no. 1 (2019): 43-56. https://doi.org/10.3892/or.2018.6825