Pin2/TRF1‑binding protein X1 inhibits colorectal cancer cell migration and invasion in vitro and metastasis in vivo via the nuclear factor‑κB signaling pathway

Retraction in: /10.3892/or.2023.8477

  • Authors:
    • Tao Jiang
    • Haiqing Li
    • Ranran Jiang
    • Hailong Li
    • Pingfu Hou
    • Yansu Chen
    • Jin Bai
    • Jun Song
  • View Affiliations

  • Published online on: July 13, 2018     https://doi.org/10.3892/or.2018.6570
  • Pages: 1533-1544
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pin2/TRF1‑binding protein X1 (PinX1) functioned as a potent inhibitor of telomerase, which was also widely considered to be a sufficient tumor suppressor. Previous studies have demonstrated that PinX1 expression was reduced in several types of cancer and was associated with poor overall survival. However, little is known regarding the role of PinX1 in colorectal cancer (CRC). The present study investigated PinX1 expression via immunostaining of CRC tissue microarrays consisting of tumor and adjacent non‑cancerous tissues (ANCT) from 568 patients. PinX1 expression was significantly lower in CRC tissues than in ANCT. Decreased PinX1 expression was revealed to be associated with lymph node metastasis, distant metastasis and advanced Tumor‑Node‑Metastasis stage, as well as a poorer overall and disease‑free survival. Furthermore, Cox regression analysis determined that a decreased PinX1 expression was an independent prognostic marker for patients with CRC. In an in vitro assay, PinX1 markedly restricted CRC cell migration and invasion. Additionally, the present study revealed that PinX1 could hinder the activity of matrix metalloproteinase 2 (MMP2) through nuclear factor (NF)‑κB‑dependent transcription to further suppress the migration and invasion ability of CRC cells through western blot analysis and a gelatin zymography assay. In vivo studies verified that PinX1 could suppress CRC metastasis, as well as the expression of MMP2 and NF‑κB p65. These results suggested that PinX1 can serve as an independent prognostic factor for patients with CRC and that it may function as a tumor metastasis suppressor in the progression of CRC though negatively regulating the NF‑κB/MMP2 signaling pathway.

Introduction

Colorectal cancer (CRC) is the third leading cause of cancer-associated mortality worldwide (1,2). Notably, owing to the increasing participation in colonoscopy screening, the incidence and mortality rates of CRC have declined by ~3% per year in males and females in recent years (1). However, recurrence and metastasis continue to be the main factors in the long-term survival and prognosis of patients with CRC, but the precise mechanism of this remains unclear. Furthermore, clinically effective treatment protocols for inhibiting tumor recurrence and metastasis are lacking. Therefore, identifying metastasis-associated genes in CRC and discovering the potential mechanism of this disease are of great significance to reduce the rates of recurrence and metastasis in order to alleviate the symptoms and enhance the quality of life of patients with CRC.

The PinX1 gene is located on chromosome 8p23, where a loss of heterozygosity is frequently observed in various types of human malignancy (3,4). Additionally, the PinX1 gene encodes a 45-KDa nucleolar protein comprising 328 amino acids (5), known as Pin2/TRF1-binding protein ×1 (PinX1), which is characterized by its function as a potent inhibitor of telomerase by binding human telomerase reverse transcriptase (6,7). Additional studies have reported that PinX1 was downregulated in breast, stomach, renal and ovarian carcinoma, and that a decrease in PinX1 expression was associated with CRC progression and that it may serve as an independent prognostic marker (712). Our previous studies have reported that PinX1 functions as a tumor suppressor in breast cancer and leads to a decrease or an increase in the invasion and metastasis abilities of breast cancer and clear cell type renal cell carcinomas through inhibiting cell migration and invasion (10,11). However, the role of PinX1 in the occurrence and development of CRC remains unclear. Therefore, it is worth investigating the biological functions of PinX1 and the potential mechanism of this in the development of CRC.

To assess the function of PinX1 in CRC, a tissue microarray (TMA) of CRC was used to analyze the association between PinX1 expression, and the survival and clinicopathological parameters of patients with CRC. Additionally, our in vitro and in vivo studies have revealed that PinX1 suppresses the migration and invasion of CRC by repressing the activity and expression of matrix metalloproteinase 2 (MMP2) in a nuclear factor (NF)-κB pathway-dependent manner. These results highlighted that PinX1 acted as an inhibitory factor of tumor metastasis in the improvement of CRC and suggested that PinX1 serves as a novel prognostic marker and a potential therapeutic target in patients with CRC.

Materials and methods

Patient information and specimen collection

A total of 568 patients with CRC were retrospectively enrolled from the Affiliated Hospital of Xuzhou Medical University (Jiangsu, China). All the patients had received a definitive diagnosis of CRC and subsequently underwent radical surgery (including abdominoperineal resection and low anterior resection, depending on the distance between the anus and the tumor, as well as the patient's nutritional status and other underlying diseases) at the Affiliated Hospital of Xuzhou Medical University between April 2010 and March 2015. The present study was approved by the Ethics Committee of the Affiliated Hospital of Xuzhou Medical University and all patients or their families provided written informed consent. Cancer tissues and adjacent para-carcinoma tissues (APCT) were obtained from the Department of Pathology, Affiliated Hospital of Xuzhou Medical University. The tissues were fixed with 10% formalin at room temperature for 24 h and embedded into tissue blocks with paraffin. Each patient for whom general information and clinicopathological parameters were obtained from the Medical Records Department of the Affiliated Hospital of Xuzhou Medical University had complete follow-up records.

In the present study, 327 males and 241 females were recruited. The mean age of the patients was 61.7 years (range, 21–91 years), and the majority of the patients received a pathological diagnosis of adenocarcinoma (559/568). There were 88, 391 and 82 cases with poorly-, moderately- and well-differentiated cancer, respectively; 209 patients with lymph node metastasis; and 24 cases with distant metastasis. The data of the remaining patients were lost to follow-up. The Tumor-Node-Metastasis stage was graded according to the American Joint Committee on Cancer staging system, 299 patients were classified as having stage I and II disease, and 193 patients were classified as having stage III and IV disease (13). The data of the remaining patients were lost to follow-up. The survival time was defined as the time period between surgery and mortality or the last follow-up (December 1, 2016).

Tissue microarray (TMA) and immunohistochemistry (IHC)

Duplicate 1.5-mm diameter cores were punched from the paraffin block of CRC and processed into a TMA. The streptavidin-peroxidase (SP) method was applied for IHC using a standard SP kit (OriGene Technologies, Inc., Beijing, China). Prior to immunostaining, the TMA was heated for 2 h at 70°C, followed by deparaffinization, washing with xylene and rehydration in a graded ethanol series. Endogenous peroxidases were inhibited by 3% hydrogen peroxide for 30 min at room temperature. A standard antigen retrieval method was performed by heat-induced epitope retrieval by heating the TMA slides immersed in retrieval solution (10 mM sodium citrate buffer, pH 6.0) at 100°C for 6 min in a pressure boiler. Following boiling, the slides remained in the pressure boiler, were initially cooled to 90°C and were then cooled to room temperature. The slides were subsequently incubated with a polyclonal rabbit anti-PinX1 antibody (1:50; cat. no. NBP2-32265; Novus Biologicals, LLC, Littleton, CO, USA) at 4°C overnight, and known immunostaining-positive/negative slides served as positive and negative controls.

Evaluation of immunostaining

Positive expression of KIF4A was identified by brown staining using a fluorescence microscope (Nikon ECLIPSE 80i; Nikon Corporation, Tokyo, Japan) at magnifications of ×100 and ×400. NIS-Elements F 4.00.00 software (Nikon Instruments, Inc., Melville, NY, USA) was used to acquire and analyze images. Positive PinX1 immunostaining is observed primarily in the nucleus and partially in the cytoplasm. Two blinded pathologists individually evaluated the scores of PinX1 staining. The scores of PinX1 staining were ranked according to the immunoreactive score (IRS), which is determined by multiplying the scores of staining intensity by the percentage of positive cells. The PinX1 staining intensity was graded as 0, 1, 2 or 3, corresponding to negative, weak, moderate and strong. The percentage of positive cells was also graded into four categories: 1, 0–25%; 2, 26–50%; 3, 51–75%; and 4, 76–100%. The PinX1 staining was characterized as negative (IRS, 0), weak (IRS, 1–3), moderate (IRS, 4–6) or strong (IRS, 8–12). By applying receiver operating characteristic (ROC) curve analysis, an optimum cut-off value for IRS was determined, where IRS 0–3 and 4–12 were categorized as low and high PinX1 expression, respectively.

Cell culture and transfection

The human colorectal cancer HCT116 and SW480 cell lines were obtained from (American Type Culture Collection, Manassas, VA, USA). Cells were cultured in Dulbecco's modified Eagle's medium (High glucose; Thermo Fisher Scientific, Inc., Waltham, MA, USA) with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) at 37°C in a 5% CO2 incubator. Prior to transfection, HCT116 and SW480 cells were grown to 50% confluence. Recombinant lentivirus of PinX1 and its lv3-control retrovirus (Shanghai GenePharma Co., Ltd., Shanghai, China) were used for infecting HCT116 and SW480 cells according to the manufacturer's protocol. In each 60×15 mm cell culture dish, 40 µg PinX1 small interfering (si)RNA, NF-κB-p65 siRNA or negative control siRNA (Shanghai GenePharma Co., Ltd.) were transfected using 8 µl siLentFect Lipid Reagent (Bio-Rad Laboratories, Inc., Hercules, CA, USA). A total of 48 h after transfection, the cells were used for subsequent experimentation according to the manufacturer's protocol. The siRNAs sequences were as follows: siPinX1, GAGCCACAGAUCAUAUUAATT; siNF-κB-p65, CCCUAUCCCUUUACGUCAUTT; and siCtrl, UUCUCCGAACGUGUCACGUTT.

Gelatin zymography assay

Gelatin zymography was performed as previously described (10). At 36 h after transfection, the cells were incubated in serum-free DMEM (Invitrogen; Thermo Fisher Scientific, Inc.) for 24 h. Following absorption and concentration of the supernatant medium with centrifugal filters (EMD Millipore, Billerica, MA, USA) at 7,500 × g for 20 min at 4°C, the protein samples were mixed with 2× SDS-PAGE non-reducing buffer (P0015B; Beyotime Institute of Biotechnology, Haimen, China) at a 1:1 ratio. Next, 50 µl of the mixed sample was loaded onto a 10% polyacrylamide gel containing 0.1% gelatin (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). Next, the gels were washed twice in eluent buffer (2.5% Triton X-100, 50 mM Tris-HCl, 5 mM CaCl2 and 1 µM ZnCl2, pH 7.6) for 30 min at room temperature; equilibrated twice in developing buffer (50 mM Tris-HCl, 5 mM CaCl2 and 1 µM ZnCl2, pH 7.6) for 20 min at room temperature; and finally put in incubation buffer (50 mM Tris-HCl, 5 mM CaCl2, 1 µM ZnCl2, 0.02% Brij and 0.2 M NaCl) at 37°C for 40 h. Next, the gels were incubated with staining buffer (0.05% Coomassie blue G-250 in 45% methanol, 10% acetic acid and 30% methanoic acid) for 3 h and then washed with destaining buffer (45% methanol and 10% acetic acid) until clear bands appeared. The images were obtained using a gel imaging system (Bio-Rad Laboratories, Inc.) and the activities of MMPs were measured by densitometric analysis using ImageJ 1.45s software (National Institutes of Health, Bethesda, MD, USA).

Western blot analysis

Western blot analysis was performed as previously described (10). The HCT116 and SW480 cell lines were collected and lysed in radioimmunoprecipitation assay buffer (50 mM Tris/HCl, pH 7.4; 150 mM NaCl; 1% NP-40; 0.1% SDS) containing protease inhibitors (10 µg/ml leupeptin, 10 µg/ml pepstatin A, 10 µg/ml aprotinin and 1 mM 4-[2-aminoethyl] benzenesulphonyl fluoride) for 30 min on ice. Next, the cell lysate was harvested and centrifuged at 15,000 × g for 10 min at 4°C. Protein concentration was evaluated using an Enhanced bicinchoninic acid protein assay kit (P0010; Beyotime Institute of Biotechnology). Proteins (20–40 µg) were separated on 10% SDS gels and transferred onto polyvinylidene fluoride membranes (GE Healthcare Life Sciences, Little Chalfont, UK). The membranes were incubated in blocking buffer composed of 5% skimmed milk in Tris-buffered saline with Tween-20 (TBST) on a shaking Table at room temperature for 2 h. Next, the blocked membranes were incubated overnight at 4°C with the following primary antibodies: Rabbit anti-PinX1 (1:1,000; NBP2-32265; Novus Biologicals, LLC), rabbit anti-MMP2 (1:500; GTX104577; GeneTex, Inc., Irvine, CA, USA), anti-MMP-9 (1:500; GTX100458; GeneTex, Inc.), rabbit anti-TMP metallopeptidase inhibitor 1 (TIMP-1; 1:1,000; D10E6; Cell Signaling Technology, Inc., Danvers, MA, USA), anti-TIMP-2 (1:1,000; D1887; Cell Signaling Technology, Inc.), anti-NF-κB p65 (1:1,000; 8242P; Cell Signaling Technology, Inc.) and anti-phosphorylated-NF-κB p65 antibodies (1:1,000; 3033S; Cell Signaling Technology, Inc.), as well as mouse anti-GAPDH (Cell Signaling Technology, Inc.), which acted as an internal control for the quantity of target protein. Following washing three times with PBS with Tween 20 on a shaking Table for 5 min, membranes were incubated with secondary goat anti-mouse or anti-rabbit antibodies (1:10,000; anti-mouse cat. no. SA00001-1; anti-rabbit cat. no. SA00001-2; Proteintech Group, Inc., Chicago, IL, USA) for 1 h at room temperature and the signals were identified using the Tanon 6600 Luminescent Imaging Workstation (Tanon Science & Technology Co., Ltd., Dalian, China). ImageJ 1.45s software (National Institutes of Health) was used for densitometric analysis.

Cell migration and invasion assay

The migration and invasion assays were performed using modified two-chamber plates with 8-µm pores. The Transwell filter coated with Matrigel (BD Biosciences, Franklin Lakes, NJ, USA) was applied for the invasion assay. A total of 15×105 cells into the upper chamber with serum-free DMEM (Invitrogen; Thermo Fisher Scientific, Inc.) while DMEM (Invitrogen; Thermo Fisher Scientific, Inc.) containing 20% fetal calf serum (Invitrogen; Thermo Fisher Scientific, Inc.) was simultaneously added to the lower chamber. For the migration and invasion assays, the process was terminated after 24 and 36 h of incubation at 37°C, respectively. The cells were removed from the upper chamber using swabs, and the cells that had crossed the membrane were fixed in 4% paraformaldehyde at room temperature for 20 min and stained at room temperature with crystal violet for 15 min, prior to images being captured and cells being counted under an inverted microscope at ×200 magnification in 5 random fields (DP80; Olympus Corporation, Tokyo, Japan).

Cell proliferation assay

Cell Counting kit (CCK)-8 analysis was performed to determine the effect of PinX1 on cell proliferation. Approximately 4×103 cells were seeded into each well of 96-well plates and CCK-8 solution was added 24, 48, 72 and 96 h afterwards. Cells were incubated at 37°C for 1 h after 10 µl CCK-8 solution was added. The absorbance was measured at 450 nm.

Animals and tail intravenous assay of metastasis

Female BALB/c nude mice (16–17 g; 6 weeks old) were purchased from the Shanghai Laboratory Animal Center (Shanghai, China) for studies approved by the Animal Care Committee of Xuzhou Medical College (Xuzhou, China). The nude mice were maintained in a controlled environment with controlled temperature (24–25°C), humidity (50–70%) and (light, 07:00; dark, 22:00). The water and mouse feed were sterilized by uperization and were freely available. The nude mice were randomly divided into two groups: PinX1 short hairpin RNA (shPinX1) and Control short hairpin RNA (shCtrl), with each group consisting of 10 mice. The mice were intravenously injected with 2.0×106 HCT116 cells in 200 µl PBS through the tail vein. After 45 days, the two groups of mice were sacrificed following anesthesia, prior to the occurrence of pathological mortality, and the lungs and liver were dissected and fixed with 10% formalin at room temperature for 24 h for metastatic nodule counting and further histopathological analysis and hematoxylin-eosin staining at room temperature for 2 min of 4-µm paraffin-embedded sections. The number of metastatic nodules on the surfaces of the lungs and liver of animals in each group was counted by visual inspection using a stereoscopic dissecting microscope at ×100 magnification in 5 randomly selected fields.

Statistical analysis

All statistical analyses were performed using SPSS 20.0 software (IBM Corp., Armonk, NY, USA). Based on the paired Wilcoxon signed-rank test, the significance of PinX1 expression between cancer tissues and ANCT was evaluated. The association between PinX1 expression and the clinicopathological parameters of the patients with CRC was examined by the χ2 test. The Kaplan-Meier curve method and the log-rank test were implemented to assess the association between PinX1 expression and patient survival. Univariate and multivariate Cox regression analysis was used for estimating the crude hazard ratios (HRs) and 95% confidence intervals (CIs) of the HRs. Two-way analysis of variance and Dunnett's test were conducted to assess differences between treatment groups. Data are presented as the mean ± standard deviation. P<0.05 was considered to indicate a statistically significant difference.

Results

PinX1 expression is downregulated in CRC tissues

To detect the PinX1 protein expression in CRC tissues and the ANCT, immunostaining analysis of colorectal TMA consisting of a total of 568 pairs of samples was performed. Following removal of the samples lost due to antigen retrieval, 515 CRC tissues, 528 ANCT and 507 matched samples were obtained. As shown in Fig. 1A, it was revealed that PinX1 expression was mainly distributed in the cell nucleus and partially distributed in the cytoplasm. Furthermore, 507 pairs of CRC and ANCT samples were compared using a paired Wilcoxon signed-rank test, and the data revealed that PinX1 protein expression was significantly decreased in cancer tissues, compared with ANCT (P<0.001; Fig. 1B).

Association between PinX1 expression and clinicopathological parameters of CRC

As samples with IRS 0–3 and IRS 4–12 were classified as low and high PinX1 expression, respectively, the low and high expression rates of 515 CRC samples were 46.4% (239/515) and 53.6% (276/515), respectively (Table I). Next, the association between PinX1 expression and the clinicopathological parameters of CRC was evaluated using Fisher's exact test, and the data demonstrated that low PinX1 expression was significantly associated with lymph node metastasis (P=0.021; Fig. 1C), distant metastasis (P=0.030; Fig. 1D) and advanced TNM stage (P=0.014; Fig. 1E). Furthermore, there was no noTable significance in the association between PinX1 expression, and age, sex, depth of the invasion, tumor diameter and differentiation (Table I).

Table I.

Association between PinX1 expression and clinicopathological features in patients with colorectal cancer.

Table I.

Association between PinX1 expression and clinicopathological features in patients with colorectal cancer.

PinX1 expression (n=515)

VariableLow (%)High (%) P-valuea
All patients239 (100)276 (100)
Age, years 1.000
  ≤60140 (59)161 (58)
  >6099 (41)115 (42)
Sex 0.050
  Male126 (53)170 (62)
  Female113 (47)106 (38)
Depth of invasionb 0.747
  T1/T253 (22)57 (21)
  T3/T4216 (78)219 (79)
Lymph node metastasis 0.021
  N0141 (59)190 (69)
  N1/N2/N398 (41)86 (31)
Distant metastasis 0.030
  M0226 (95)271 (98)
  M113 (5)5 (2)
TNM stage 0.014
  I–II133 (56)187 (66)
  III–IV106 (44)93 (34)
Tumor diameterc 0.366
  ≤5 cm174 (73)195 (71)
  >5 cm65 (27)81 (29)
Differentiationd 0.267
  Poor37 (18)43 (14)
  Moderate/high169 (82)259 (86)

a Two-sided Fisher's exact tests

b The data of 1 patient could not be assessed

c The data of 2 patients were lost

d The data of 7 patients with CRC were lost. PinX1, Pin2/TRF1-binding protein ×1; TNM, Tumor-Node-Metastasis.

Low PinX1 expression contributes toward a poor prognosis in patients with CRC

The Kaplan-Meier survival curve and log-rank test revealed that low PinX1 expression was associated with overall and disease-free survival of patients with CRC (Fig. 2; P=0.001 and P=0.017, respectively). Furthermore, univariate Cox regression analysis indicated that PinX1 expression was significantly associated with overall and disease-free survival in patients with CRC (P=0.001 and P=0.009, respectively; Table II). Furthermore, the independent prognostic value of PinX1 expression in CRC was confirmed using the multivariate Cox regression model, and the data revealed that the expression of PinX1 could serve as an independent prognostic marker for overall survival with P=0.001 (HR=0.57; 95% CI, 0.41–0.79) and disease-free survival with P=0.017 (HR=0.53; 95% CI, 0.31–0.90; Table III).

Table II.

Univariate Cox regression analysis of PinX1 expression and clinicopathological variables predicting the survival of 515 patients with colorectal cancer.

Table II.

Univariate Cox regression analysis of PinX1 expression and clinicopathological variables predicting the survival of 515 patients with colorectal cancer.

Overall survivalDisease-free survival


VariableaHR (95% CI)P-valueHR (95%CI)P-value
PinX10.60 (0.42–0.79)0.0010.55 (0.33–0.91)0.019
Age1.20 (0.88–1.65)0.2501.46 (0.89–2.41)0.135
Sex1.40 (1.03–1.92)0.0341.97 (1.19–3.25)0.010
LNM1.30 (0.95–1.78)0.1071.97 (1.19–3.25)0.008
Distant metastasis2.86 (1.39–5.85)0.0043.39 (1.23–9.39)0.019
TNM stage1.45 (1.06–1.98)0.0212.01 (1.22–3.31)0.006
Differentiation0.83 (0.72–0.96)0.0120.75 (0.59–0.94)0.012
Tumor diameter1.30 (0.93–1.82)0.1261.78 (1.07–2.97)0.027
Depth of invasion1.80 (1.45–2.83)0.0115.49 (1.72–17.5)0.004

{ label (or @symbol) needed for fn[@id='tfn5-or-40-03-1533'] } PinX1, Pin2/TRF1-binding protein ×1; HR, hazard ratio; CI, confidence interval; LNM, lymph node metastasis; TNM, Tumor-Node-Metastasis.

a PinX1, low vs. high; Age, ≤60 vs. >60; Sex, male vs. female; LNM, N0 vs. N1, N2 and N3; Depth of invasion, T1-T2 vs. T3-T4; Distant metastasis, M0 vs. M1; Differentiation, moderate and high vs. poor; TNM stage, I–II vs. III–IV; Tumor diameter, ≤5 vs. >5.

Table III.

Multivariate Cox regression analysis models assessing the effects of covariates on overall and disease-free survival in 515 patients with colorectal cancer.

Table III.

Multivariate Cox regression analysis models assessing the effects of covariates on overall and disease-free survival in 515 patients with colorectal cancer.

Overall survivalDisease-free survival


VariableaHR (95% CI)P-valueHR (95% CI)P-value
PinX10.57 (0.41–0.79)0.0010.53 (0.31–0.90)0.017
Age1.20 (0.87–1.65)0.2731.38 (0.83–2.28)0.213
Sex1.45 (1.04–2.01)0.0302.29 (1.35–3.87)0.002
Tumor diameter1.27 (0.89–1.80)0.1741.87 (1.10–3.17)0.020
TNM stage1.48 (1.07–2.04)0.0202.11 (1.27–3.53)0.004
Differentiation1.44 (1.03–2.00)0.0330.74 (0.58–0.94)0.014

{ label (or @symbol) needed for fn[@id='tfn7-or-40-03-1533'] } HR, hazard ratio; CI, confidence interval; PinX1, Pin2/TRF1-binding protein ×1; TNM, Tumor-Node-Metastasis.

a PinX1, low vs. high; Age, ≤60 vs. >60; Sex, male vs. female; Tumor diameter, ≤5 cm vs. >5 cm; Differentiation, moderate and high vs. poor; TNM stage, I–II vs. III–IV.

Silencing of PinX1 promotes CRC cell migration, invasion and proliferation in vitro

The results of the present study demonstrated that low PinX1 expression is associated with a poorer prognosis than high PinX1 expression and may accelerate tumor metastasis in CRC. Therefore, it was further examined whether PinX1 participated in CRC cell migration and invasion in vitro. For the in vitro assay, HCT116 and SW480 cells with stable interference of PinX1 expression were constructed though retroviral interference (Fig. 3A and B).

Notably, cell migration was markedly increased following silencing of PinX1 expression in the HCT116 and SW480 cell lines (Fig. 3C and D). Concurrently, analogous results were observed in the cell invasion assay, demonstrating that cell invasion was also significantly increased (Fig. 3E and F).

The CCK-8 cell proliferation assay revealed that cell proliferation in the HCT116 and SW480 cell lines with PinX1-knockdown was increased compared with that in the cells in the control groups (Fig. 3G and H).

PinX1 inhibits invasion by decreasing the expression and activity of MMP2 in CRC

Previous studies have demonstrated that the MMP family serves an important role in malignancy metastasis, which could degrade the extracellular matrix (ECM) and facilitate the invasion of tumor cells through the basement membrane (14,15). To examine whether PinX1 regulates metastasis through MMPs in CRC cells, the protein expression level and activity of MMPs were detected by western blot analysis and gelatin zymography, respectively. It was revealed that MMP2 expression and activity were increased following PinX1-knockdown in HCT116 and SW480 cells (Fig. 4A and B). However, MMP9 expression did not exhibit a significant change under identical conditions (Fig. 4A). Therefore, we hypothesized that PinX1 could inhibit invasion by decreasing MMP2 expression and activity in CRC cells.

Furthermore, the protein levels of TIMP1 and TIMP2, which are tissue inhibitors of MMP9 and MMP2, respectively, were detected. The results of the present study demonstrated that MMP2 expression varied with PinX1 expression, whereas the expression of TIMP1 and TIMP2 did not present a significant alteration with PinX1 silencing in HCT116 and SW480 cells (Fig. 4A).

PinX1 suppresses MMP2 expression via the NF-κB signaling pathway in CRC

Evidence has indicated that the NF-κB signaling pathways are crucial for tumor development (16) and contribute toward tumor ECM destruction (17,18). NF-κB could regulate MMPs at the transcription level through recognizing the κB sites in the promoters of MMP genes (19). The present study demonstrated that the expression levels of NF-κB-p65 (p65), phosphorylated-NF-κB-p65 (p-p65) and MMP2 were significantly increased following PinX1-knockdown in HCT116 and SW480 cells (Fig. 4C). Therefore, we hypothesized that PinX1 may regulate MMP2 through the NF-κB pathway in CRC.

To further ascertain whether PinX1 affects MMP2 through the NF-κB signaling pathway, p65 siRNA was transfected into PinX1-silenced HCT116 and SW480 cells. Western blot analysis demonstrated that the levels of MMP2, p65 and p-p65 were decreased following p65 interference in CRC cells (Fig. 5A and B). Additionally, the migration and invasion abilities repressed by PinX1-knockdown were markedly reversed by the silencing of p65 (Fig. 5C and D). These data suggested that PinX1 regulates migration and invasion via the NF-κB/MMP2 signaling pathway.

PinX1 negatively regulates CRC cell metastasis in vivo

To further investigate the role of PinX1 in CRC metastasis in vivo, HCT116 cells infected with shCtrl or shPinX1 were injected into two groups of nude mice via the tail vein. A total of 45 days later, the mice were sacrificed, the lungs and livers were dissected and fixed with 10% formalin for metastatic nodule counting and further histopathological analysis.

Hematoxylin-eosin staining revealed that the randomly selected metastatic foci were present in the livers, rather than in the lungs (Fig. 6A). Extensive micro-metastases were detected in the livers of the mice injected with HCT116-shPinX1 cells (Fig. 6B). Furthermore, statistical analysis revealed that the number of metastatic foci was markedly increased in the shPinX1 group compared with that in the shCtrl group (Fig. 6C).

Immunohistochemical staining of metastatic nodules in the liver demonstrated that the expression levels of MMP2 and p65 in the shPinX1 group were increased compared with those in the shCtrl group (Fig. 6D). These results further confirmed our in vitro conclusions.

Discussion

The present study investigated the roles of PinX1 in human CRC by combining PinX1 immunostaining with the retrospective cohorts of 515 patients with CRC. The results revealed that low PinX1 expression was significantly associated with tumor metastasis to distant organs or lymph nodes and advanced TNM stage (Table I). Prognostic analysis demonstrated that low levels of PinX1 were associated with poorer overall and disease-free survival rates (Fig. 2A and B). Cox regression analysis revealed that low PinX1 expression acts as an independent adverse prognostic indicator for patients with CRC (Tables II and III). These results supported the possible inhibitory effects of PinX1 on colorectal tumor metastasis and its potential as an independent indicator for the treatment of patients with CRC. However, how PinX1 regulates the metastasis of CRC remains unclear; therefore, the present study investigated the potential mechanisms of regulating CRC metastasis.

The in vitro assay revealed that the migration and invasion of CRC cells was markedly increased following knockdown of PinX1 (Fig. 3). Furthermore, the migration and invasion of tumor cells has crucial effects on tumor metastasis, and acquiring such a capacity is typically a vital step in tumor metastasis. Therefore, tumor cells would move to the basement membrane to combine with the corresponding receptors and degrade the ECM (20,21). It is known that the MMP family of proteolytic enzymes degrades the ECM and basement membrane, which serves an important role in facilitating the invasion of tumor cells through the basement membrane barrier to result in infiltration and metastasis (14,15,22). The individual MMPs, MMP2 and MMP9, are the major enzymes in the degradation of the basement membrane and ECM (23). Previous studies have reported that increased expression of MMP2 and MMP9 contributed toward a poorer prognosis for patients with CRC, and participated in the process of CRC metastasis (24,25). The present study demonstrated that PinX1 could suppress the expression and activity of MMP2 but not those of MMP9 (Fig. 4A and B). Therefore, we hypothesized that PinX1 may inhibit the migration and invasion of CRC by regulating MMP2 expression.

A vital mechanism for the regulation of the activity of MMPs draws support from binding to the specific endogenous tissue inhibitors of metalloproteinases (TIMPs) (26). Among the TIMPs, TIMP1 and TIMP2 are indicated as specific tissue inhibitors of MMP9 and MMP2, respectively (27). The results of the present study indicated that following PinX1-knockdown, the expression of TIMP1 and TIMP2 did not significantly change in CRC cells, suggesting that the PinX1 gene is not regulated through TIMP2 and that other mechanisms regulate MMP2 to affect the ability of CRC cell migration and invasion in vitro. Therefore, this specific mechanism requires further investigation.

Recent studies have indicated that the NF-κB pathway is important for tumor development and that it is involved in stimulating cell proliferation, inhibiting apoptosis, and increasing metastasis and angiogenesis (16), including CRC (28). NF-κB comprises different protein dimers that bind to a common sequence motif known as the κB site, which was identified in the promoters of genes that encode MMPs (1719). NF-κB is constitutively expressed in cells as a heterodimer, comprising a p50 DNA-binding subunit and the p65 trans-activating subunit (29). Previous studies have reported that the N-terminal Gly-rich patch (G-patch) of PinX1 is a key nucleic acid binding domain that combines with the C-terminus of the NF-κB-repression factor (NRF) (30). NRF, a nuclear inhibitor of NF-κB, can constrain the transcriptional activity of NF-κB proteins by protein-protein interactions (31). Therefore, we hypothesized that PinX1 can also inhibit the transcriptional activity of NF-κB proteins by direct protein-protein interactions through its G-patch domain. The results of the present study demonstrated that p65-siRNA efficiently inhibited the upregulation of MMP2 expression induced by PinX1-knockdown (Fig. 5A and B). Furthermore, the enhanced migration and invasion resulting from PinX1-knockdown were also suppressed by p65-siRNA in CRC cells (Fig. 5C and D). Therefore, these results suggested that PinX1 may control the NF-κB/MMP2 signaling pathway for the regulation of the migration and invasion of CRC cells. However, the molecular mechanism of how PinX1 regulates the NF-κB/MMP2 signaling pathway was not investigated; therefore, future studies will focus on investigating whether PinX1 could function as a transcription factor and regulate NF-κB/MMP2 at the transcription level.

To further determine the functional effect of PinX1 in CRC metastasis in vivo, an experimental model comprising two groups of nude mice was constructed. Using this model, it was demonstrated that PinX1-knockdown in CRC cells significantly inhibited the formation of metastasis nodules in the livers of nude mice. Further immunohistochemical staining of MMP2 and p65 revealed that the expression levels of MMP2 and p65 in the shPinX1 group were increased compared with those in the shCtrl group (Fig. 6). This observation confirmed that PinX1 suppressed CRC metastasis by inhibiting MMP2 expression and activity via the NF-κB pathway.

In conclusion, the results of the present study suggested that reduced PinX1 expression could be regarded as an independent prognostic factor for patients with CRC and that PinX1 can function as an authentic tumor metastasis suppressor in the progression of CRC by negatively regulating the NF-κB/MMP2 signaling pathway. These findings indicated that PinX1 may be an effective target for targeted therapy of patients with CRC and may serve an important role as a therapeutic target to combat CRC metastasis.

Acknowledgements

Not applicable.

Funding

The present study was funded by grants from the National Natural Science Foundation of China (grant nos. 81472663, 81502280 and 81672845), the Education Department of Jiangsu Province (grant no. 15KJA320006) and the Postgraduate Research & Practice Innovation Program of Jiangsu Province (grant no. SJCX17_0553). This work was also supported by a grant ‘Project of Invigorating Health Care through Science, Technology and Education’, Jiangsu, China (grant no. LGY2017093).

Availability of data and materials

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

Authors' contributions

TJ, JB and JS conceived and designed the experiments; HL, RJ and HL conducted the experiments; YC and PH performed the statistical analysis. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Written informed consent was obtained from all patients and the present study was approved by the Review Board of the Affiliated Hospital of Xuzhou Medical University. The animal studies were approved by the Animal Care Committee of Xuzhou Medical University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

PinX1

Pin2/TRF1-binding protein ×1

CRC

colorectal cancer

ANCT

adjacent non-cancerous tissues

IHC

immunohistochemistry

NF-κB

nuclear factor κB

MMPs

matrix metalloproteinases

TMA

tissue microarray

IRS

immunoreactive score

ROC

receiver operating characteristic

ECM

extracellular matrix

HR

hazard ratio

CI

confidence interval

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Siegel R, Desantis C and Jemal A: Colorectal cancer statistics, 2014. CA Cancer J Clin. 64:104–117. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Becker SA, Zhou YZ and Slagle BL: Frequent loss of chromosome 8p in hepatitis B virus-positive hepatocellular carcinomas from China. Cancer Res. 56:5092–5097. 1996.PubMed/NCBI

4 

Baffa R, Santoro R, Bullrich F, Mandes B, Ishii H and Croce CM: Definition and refinement of chromosome 8p regions of loss of heterozygosity in gastric cancer. Clin Cancer Res. 6:1372–1377. 2000.PubMed/NCBI

5 

Johnson FB: PinX1 the tail on the chromosome. J Clin Invest. 121:1242–1244. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Zhou XZ and Lu KP: The Pin2/TRF1-interacting protein PinX1 is a potent telomerase inhibitor. Cell. 107:347–359. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Kondo T, Oue N, Mitani Y, Kuniyasu H, Noguchi T, Kuraoka K, Nakayama H and Yasui W: Loss of heterozygosity and histone hypoacetylation of the PINX1 gene are associated with reduced expression in gastric carcinoma. Oncogene. 24:157–164. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Kim MS, Kim SS, Yoo NJ and Lee SH: Somatic mutation of PINX1 gene is rare in common solid cancers. APMIS. 120:770–771. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Ma Y, Wu L, Liu C, Xu L, Li D and Li JC: The correlation of genetic instability of PINX1 gene to clinico-pathological features of gastric cancer in the Chinese population. J Cancer Res Clin Oncol. 135:431–437. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Shi M, Cao M, Song J, Liu Q, Li H, Meng F, Pan Z, Bai J and Zheng J: PinX1 inhibits the invasion and metastasis of human breast cancer via suppressing NF-κB/MMP-9 signaling pathway. Mol Cancer. 14:662015. View Article : Google Scholar : PubMed/NCBI

11 

Li HL, Han L, Chen HR, Meng F, Liu QH, Pan ZQ, Bai J and Zheng JN: PinX1 serves as a potential prognostic indicator for clear cell renal cell carcinoma and inhibits its invasion and metastasis by suppressing MMP-2 via NF-κB-dependent transcription. Oncotarget. 6:21406–21420. 2015.PubMed/NCBI

12 

Cai MY, Zhang B, He WP, Yang GF, Rao HL, Rao ZY, Wu QL, Guan XY, Kung HF, Zeng YX, et al: Decreased expression of PinX1 protein is correlated with tumor development and is a new independent poor prognostic factor in ovarian carcinoma. Cancer Sci. 101:1543–1549. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Weiser MR: AJCC 8th Edition: Colorectal Cancer. Ann Surg Oncol. 25:1454–1455. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Yin LL, Chung CM, Chen J, Fok KL, Ng CP, Jia RR, Ren X, Zhou J, Zhang T, Zhao XH, et al: A suppressor of multiple extracellular matrix-degrading proteases and cancer metastasis. J Cell Mol Med. 13:4034–4041. 2009. View Article : Google Scholar : PubMed/NCBI

15 

DeClerck YA, Mercurio AM, Stack MS, Chapman HA, Zutter MM, Muschel RJ, Raz A, Matrisian LM, Sloane BF, Noel A, et al: Proteases, extracellular matrix, and cancer: A workshop of the path B study section. Am J Pathol. 164:1131–1139. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Karin M, Cao Y, Greten FR and Li ZW: NF-κB in cancer: From innocent bystander to major culprit. Nat Rev Cancer. 2:301–310. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Wang W, Abbruzzese JL, Evans DB and Chiao PJ: Overexpression of urokinase-type plasminogen activator in pancreatic adenocarcinoma is regulated by constitutively activated RelA. Oncogene. 18:4554–4563. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Takeshita H, Yoshizaki T, Miller WE, Sato H, Furukawa M, Pagano JS and Raab-Traub N: Matrix metalloproteinase 9 expression is induced by Epstein-Barr virus latent membrane protein 1 C-terminal activation regions 1 and 2. J Virol. 73:5548–5555. 1999.PubMed/NCBI

19 

Bond M, Fabunmi RP, Baker AH and Newby AC: Synergistic upregulation of metalloproteinase-9 by growth factors and inflammatory cytokines: An absolute requirement for transcription factor NF-kappa B. FEBS Lett. 435:29–34. 1998. View Article : Google Scholar : PubMed/NCBI

20 

Duffy MJ: The biochemistry of metastasis. Adv Clin Chem. 32:135–166. 1996. View Article : Google Scholar : PubMed/NCBI

21 

Price JT, Bonovich MT and Kohn EC: The biochemistry of cancer dissemination. Crit Rev Biochem Mol Biol. 32:175–253. 1997. View Article : Google Scholar : PubMed/NCBI

22 

Xu X, Wang Y, Chen Z, Sternlicht MD, Hidalgo M and Steffensen B: Matrix metalloproteinase-2 contributes to cancer cell migration on collagen. Cancer Res. 65:130–136. 2005.PubMed/NCBI

23 

Li HC, Cao DC, Liu Y, Hou YF, Wu J, Lu JS, Di GH, Liu G, Li FM, Ou ZL, et al: Prognostic value of matrix metalloproteinases (MMP-2 and MMP-9) in patients with lymph node-negative breast carcinoma. Breast Cancer Res Treat. 88:75–85. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Chu D, Zhao Z, Zhou Y, Li Y, Li J, Zheng J, Zhao Q and Wang W: Matrix metalloproteinase-9 is associated with relapse and prognosis of patients with colorectal cancer. Ann Surg Oncol. 19:318–325. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Langers AM, Verspaget HW, Hawinkels LJ, Kubben FJ, van Duijn W, van der Reijden JJ, Hardwick JC, Hommes DW and Sier CF: MMP-2 and MMP-9 in normal mucosa are independently associated with outcome of colorectal cancer patients. Br J Cancer. 106:1495–1498. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Brew K, Dinakarpandian D and Nagase H: Tissue inhibitors of metalloproteinases: Evolution, structure and function. Biochim Biophys Acta. 1477:267–283. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Lambert E, Dassé E, Haye B and Petitfrère E: TIMPs as multifacial proteins. Crit Rev Oncol Hematol. 49:187–198. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Ma J, Mi C, Wang KS, Lee JJ and Jin X: Zinc finger protein 91 (ZFP91) activates HIF-1α via NF-κB/p65 to promote proliferation and tumorigenesis of colon cancer. Oncotarget. 7:36551–36562. 2016.PubMed/NCBI

29 

Lee WR, Chung CL, Hsiao CJ, Chou YC, Hsueh PJ, Yang PC, Jan JS, Cheng YW and Hsiao G: Suppression of matrix metalloproteinase-9 expression by andrographolide in human monocytic THP-1 cells via inhibition of NF-κB activation. Phytomedicine. 19:270–277. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Jianfeng D, Feng J, Chaoneng J, Zhongzhou Z, Shaohua G, Qihan W, Liu W, Gang Y, Yi X and Mao Y: Cloning of the correct full length cDNA of NF-kappaB-repressing factor. Mol Cells. 16:397–401. 2003.PubMed/NCBI

31 

Nourbakhsh M and Hauser H: Constitutive silencing of IFN-beta promoter is mediated by NRF (NF-kappa B-repressing factor), a nuclear inhibitor of NF-kappa B. EMBO J. 18:6415–6425. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 40 Issue 3

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
Jiang T, Li H, Jiang R, Li H, Hou P, Chen Y, Bai J and Song J: Pin2/TRF1‑binding protein X1 inhibits colorectal cancer cell migration and invasion in vitro and metastasis in vivo via the nuclear factor‑κB signaling pathway Retraction in /10.3892/or.2023.8477. Oncol Rep 40: 1533-1544, 2018
APA
Jiang, T., Li, H., Jiang, R., Li, H., Hou, P., Chen, Y. ... Song, J. (2018). Pin2/TRF1‑binding protein X1 inhibits colorectal cancer cell migration and invasion in vitro and metastasis in vivo via the nuclear factor‑κB signaling pathway Retraction in /10.3892/or.2023.8477. Oncology Reports, 40, 1533-1544. https://doi.org/10.3892/or.2018.6570
MLA
Jiang, T., Li, H., Jiang, R., Li, H., Hou, P., Chen, Y., Bai, J., Song, J."Pin2/TRF1‑binding protein X1 inhibits colorectal cancer cell migration and invasion in vitro and metastasis in vivo via the nuclear factor‑κB signaling pathway Retraction in /10.3892/or.2023.8477". Oncology Reports 40.3 (2018): 1533-1544.
Chicago
Jiang, T., Li, H., Jiang, R., Li, H., Hou, P., Chen, Y., Bai, J., Song, J."Pin2/TRF1‑binding protein X1 inhibits colorectal cancer cell migration and invasion in vitro and metastasis in vivo via the nuclear factor‑κB signaling pathway Retraction in /10.3892/or.2023.8477". Oncology Reports 40, no. 3 (2018): 1533-1544. https://doi.org/10.3892/or.2018.6570