Open Access

Ursolic acid inhibits the metastasis of colon cancer by downregulating ARL4C expression

  • Authors:
    • Mengzhe Zhang
    • Fenfen Xiang
    • Yipeng Sun
    • Rongrong Liu
    • Qian Li
    • Qing Gu
    • Xiangdong Kang
    • Rong Wu
  • View Affiliations

  • Published online on: December 18, 2023     https://doi.org/10.3892/or.2023.8686
  • Article Number: 27
  • Copyright: © Zhang 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

Ursolic acid (UA), a natural pentacyclic triterpenoid, is known to exhibit various biological activities and anticancer effects. However, the underlying anticancer mechanism is not fully understood to date. The present study aimed to investigate the antimetastatic effect of UA through ADP‑ribosylation factor like GTPase 4C (ARL4C) in colon cancer. A lung metastasis model of colon cancer in nude mice was established through tail vein injection. A Cell Counting Kit‑8 assay was used to investigate the proliferation of colon cancer cells. Transwell assays were used to detect cell migration and invasion. The expression levels of proteins including ARL4C, matrix metallopeptidase 2 (MMP2), phosphorylated (p)‑AKT and p‑mTOR were measured using western blot analysis. Immunohistochemistry was used to determine the protein expression level in tissues. ARL4C ubiquitination levels were analysed using immunoprecipitation and western blotting. The results indicated that UA inhibits the metastasis of colon cancer in vivo and in vitro. The expression of ARL4C in human colon cancer tissue was significantly higher than that in adjacent tissues and its high expression level was associated with lymph node metastases and tumour stage. UA treatment significantly decreased ARL4C and MMP2 protein levels and inhibited the AKT/mTOR signalling pathway. Overexpression of ARL4C reversed the inhibitory effect of UA on the invasion and migration of HCT‑116 and SW480 cells, as well as the expression and secretion of MMP2 protein. In addition, UA and an AKT signalling pathway inhibitor (LY294002) induced the ubiquitination of the ARL4C protein, which was reversed by a proteasome inhibitor (MG‑132). Collectively, it was revealed in the present study that UA served as a novel solution to relieve colon cancer metastasis by inducing the ubiquitination‑mediated degradation of ARL4C by modulating the AKT signalling pathway. Thus, UA may be a promising treatment option to prolong the survival of patients with colon cancer metastasis.

Introduction

Colon cancer is one of the most common gastrointestinal tumours and it is the second leading overall cause of cancer-related mortality (1). Previous studies have revealed that 20–25% of patients with colon cancer present with metastatic disease at the time of diagnosis (2). Although treatments such as surgery, chemotherapy, radiotherapy and immunotherapy can increase the survival rate, metastasis and relapse remain the leading causes of death in most patients with colon cancer (3,4). Thus, it is necessary to understand the underlying molecular mechanisms of colon cancer metastasis and develop new molecular markers and drugs for improving patient diagnosis and treatment.

ADP-ribosylation factor (ARF) like GTPase 4C (ARL4C), a small GTP binding protein, belongs to the ARF subfamily of proteins (5). Previous studies have demonstrated that ARL4C modulates filopodium formation and cell migration (6,7), participates in cytoskeleton rearrangement (8) and intracellular vesicle transport (9). In addition, as the target gene of the Wnt/β-catenin and EGF/Ras signalling pathways, ARL4C is a regulator coordinating carcinogenesis and epithelial tubule formation (10). There is evidence that ARL4C enhances the stem-like characteristics of glioblastoma cells and is markedly increased in patients with high-grade glioma (11). Patients with glioma and low ARL4C levels have longer overall survival and progression-free survival (11). High expression of ARL4C is reported to be a poor prognostic factor in gastric cancer and is related to the depth of tumour invasion and peritoneal metastasis (12). Moreover, ARL4C silencing resulted in cell cycle arrest and increased apoptosis in gastric cancer cells through inhibition of MDM2 protein expression in the p53 pathway (13). The growth of primary and metastatic liver tumours in mice can be effectively reduced by subcutaneous injection of oligonucleotide drugs targeting ARL4C (14). These findings indicated that targeting ARL4C may be an effective and precise cancer treatment for tumours.

Currently, Chinese herbal medicine is regarded as an important source for the development of new anticancer drugs (15). With its high efficiency and low toxicity, it has gradually become the focus of attention. Ursolic acid (UA) is a triterpenoid compound in natural plants that is widely employed in Traditional Chinese Medicine, such as Hedyotis diffusa, Gardenia and Prunella vulgaris. UA possesses antioxidant, anti-inflammatory and antitumorigenic biological activities (16). In the case of cancer treatment, UA exhibits preventive and therapeutic effects in several different cancer types, including breast cancer (17), lung cancer (18), oral squamous cell carcinoma (19), prostate cancer (20) and colorectal cancer (21). It was reported that UA promoted apoptosis, autophagy and chemosensitivity to gemcitabine in pancreatic cancer (22). Moreover, UA was shown to inhibit the metastasis of prostate cancer to the lung and liver by downregulating the expression of C-X-C motif chemokine receptor 4 (CXCR4) in the prostate tissue of TRAMP mice (23). Mechanistically, the anticancer effect of UA was associated with the inhibition of cell proliferation (24) and invasion (25), reversal of drug resistance (26), induction of apoptosis (27) and regulation of the tumour microenvironment and cancer immunity (28). Concerning colon cancer, several studies have investigated the effects and mechanisms of UA in cell proliferation, angiogenesis and apoptosis (2931). However, the effect of UA on colon cancer invasion and migration and its corresponding molecular mechanism remains unclear. The present study aimed to investigate the effect of UA on invasion and migration in vitro and in vivo and revealed the pharmacological effect and mechanism of UA on ARL4C expression. The present study may provide a potential molecular target and novel insights into UA, in the effective treatment of colon cancer metastasis.

Materials and methods

Cell culture and reagents

Human colon cancer cell lines, HCT-116 and SW480, were purchased from the The Cell Bank of Type Culture Collection of The Chinese Academy of Sciences. The cells were cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.) containing 10% FBS (Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 g/ml streptomycin (HyClone™; Cytiva) at 37°C in a 5% CO2 incubator. UA (Selleck Chemicals) was dissolved in dimethyl sulfoxide (Sigma-Aldrich; Merck KGaA) and stored at −20°C. LY294002, rapamycin and MG-132 were purchased from Selleck Chemicals.

Cell proliferation assays

HCT-116 and SW480 cells were seeded in 96-well plates at a density of 1×104 cells/well and incubated overnight at 37°C. The cells were treated with various concentrations of UA (0, 5, 10, 15, 20, 25, 30 and 40 µmol/l) for 24 or 48 h. Subsequently, serum-free medium containing 10% Cell Counting Kit-8 (Dojindo Laboratories, Inc.) reagent was added and incubated at 37°C for 2 h. The absorbance was measured at 450 nm with a microplate reader.

Transwell migration and invasion assays

For the migration analysis, HCT-116 and SW480 cells (5×104 cells in RPMI-1640 serum-free medium) pretreated with different concentrations of UA were evenly seeded in the Transwell upper chamber. A total of 500 µl of medium containing 10% FBS was added to the lower chamber. After 36 h of incubation at 37°C, the migrating cells were fixed with 4% paraformaldehyde at room temperature for 30 min and stained with crystal violet solution at room temperature for 10 min. The number of cells in three random fields was recorded using a light microscope (Leica Microsystems GmbH). Regarding the invasion analysis, 100 µl Matrigel (BD Biosciences) were placed on the bottom of the Transwell upper chamber in advance and incubated at 37°C for 6 h. The remaining steps were the same as those for the migration analysis.

Tissue samples

Human tissue samples were obtained from a tissue microarray (cat. no. HCol-Ade060CS-01) provided by Shanghai Xinchao Biological Technology Co., Ltd., including 40 colon cancer tissue samples and 40 adjacent normal tissues. The present study was approved (approval no. PTEC-A-2021-29-1) by the Ethics Committee of Putuo Hospital affiliated to Shanghai University of Traditional Chinese Medicine (Shanghai, China).

Immunohistochemistry

The mouse lung tissue was fixed in 4% paraformaldehyde (Ruibaohe Biotechnology, Inc.) at room temperature for 24 h, and then was dehydrated in an automatic dehydrator. After being embedded in paraffin, the tissue was sliced into 5-µm-thick slices. Subsequently, immunohistochemical staining was performed according to the manufacturer's instructions. The slides were placed in 100% xylene for dewaxing and were then rehydrated in the graded ethanol series (70, 80, 90, 95, and 100%; Shanghai Lingfeng Chemical Reagent Co., Ltd.). For heat-mediated antigen retrieval, the slides were heated at 100°C for 15 min and 50°C for 6 min in citrate buffer (0.01 mmol/l, pH 6.0). Tissue sections were incubated with endogenous peroxygenase inhibitor (cat. no. PV-9001; Beijing Zhongshan Golden Bridge Biotechnology Co., Ltd.) for 10 min at room temperature and subsequently with 5% BSA (cat. no. ST025; Beyotime Institute of Biotechnology) at 37°C for 30 min. The sections were incubated with primary antibody against ARL4C (1:200; cat. no. ab122025, Abcam) at 4°C overnight and then incubated with a secondary, horseradish peroxidase (HRP)-labelled antibody at 37°C for 30 min. DAB staining solution was added (Fuzhou Maixin Biotechnology, Inc.) and the sections were incubated at room temperature for 3 min. Finally, the slides were counterstained with hematoxylin at room temperature for 10 sec, washed with running water, soaked in deionized water for 30 min and dried overnight at room temperature. The slides were observed and images were captured under a light microscope (BX43; Olympus Corporation). Analysis of the pathological changes was conducted using Image-Pro Plus software (v 6.0; Media Cybernetics, Inc.).

Western blot analysis

Cells were lysed with the use of RIPA buffer (Beyotime Institute of Biotechnology) containing a protease inhibitor on ice for 15 min. Cell lysates were subjected to centrifugation at 8,051 × g for 15 min at 4°C and the supernatant was drawn. The total protein was quantified using a BCA kit (Beyotime Institute of Biotechnology). A total of 20 µg protein was separated using 10% SDS-PAGE and transferred onto PVDF membranes (MilliporeSigma). The membranes were blocked with 5% non-fat milk at room temperature for 2 h and then incubated with primary antibodies against β-actin (1:5,000; cat. no. ab6276, Abcam), ARL4C (1:1,000; cat. no. ab122025, Abcam) and matrix metallopeptidase 2 (MMP2) (1:1,000; cat. no. ab92536, Abcam), p-AKT (1:1,000; cat. no. 4060S, Cell Signaling Technology, Inc.) and p-mTOR (1:1,000; cat. no. 5536, Cell Signaling Technology, Inc.) overnight at 4°C. The membranes were washed three times with TBST buffer containing 0.05% Tween-20 (cat. no. ST1726, Beyotime Institute of Biotechnology) and incubated with anti-rabbit IgG, HRP-conjugated secondary antibody (1:1,000; cat. no. 7074, Cell Signaling Technology, Inc.) for 2 h at room temperature. Finally, an enhanced chemiluminescence reagent (Bio-Rad Laboratories, Inc.) was added to visualize the immunoreactive bands. The band intensity was analysed with ImageJ software (version 1.51J8; National Institutes of Health) using β-actin as the loading control.

Construction of the ARL4C lentivirus

To generate the ARL4C-overexpressing cell lines (ARL4C-OE), the 2nd generational system was used in the lentivirus transfection experiment. Briefly, the ARL4C coding sequence was cloned into the GV358 (11.6 kb) lentiviral vector (Shanghai GeneChem Co., Ltd.) to form recombinant plasmids. Subsequently, the recombinant GV358, Helper 1.0 and Helper 2.0 plasmids were co-transfected into 293T cells at 37°C. The quantity of plasmids, Helper 1.0 (packaging) and Helper 2.0 (envelope) were 20, 15 and 10 µg, respectively. After 72 h, the culture supernatant was harvested, concentrated and purified (82,700 × g for 2 h at 4°C). The concentration of lentivirus was 1.5×109 TU/ml. The HCT-116 and SW480 cells (5×104) were then seeded on a 12-well plate wherein cells were transfected with lentivirus containing human ARL4C-Flag cDNA or lentiviral vectors (Ubi-MCS-SV40-puromycin; cat. no. con254; Shanghai GeneChem Co., Ltd.) for 24 h at 37°C (multiplicity of infection=10). Subsequently, the transfection solution was replaced with complete medium, and the stable strain selection experiment was carried out after culturing for 96 h with puromycin at 37°C. The concentration of puromycin for induction was 5 µg/ml determined in the preliminary experiment and that for maintenance was 2 µg/ml. As the lentiviral vector contains Flag elements instead of enhanced green fluorescent protein (EGFP), western blot analysis detecting Flag antibody and PCR detecting ARL4C gene expression were conducted for determining the success of the transfection. The time interval between transduction and subsequent experimentation was ~2 weeks. The stable strain expressing ARL4C, and the negative control were named ARL4C-OE and ARL4C-NC respectively.

ELISA analysis

According to the manufacturer's instructions, the secretion of MMP2 in cell supernatant was detected using an ELISA kit (cat. no. orb1494431, Biorbyt, Ltd.).

In vivo model of lung metastasis

The male BALB/c-nude mice (5 weeks old; weighing 15–18 g; n=18) used in the present study were purchased from the Shanghai SLAC Laboratory Animal Co., Ltd. The mice were randomly divided into three groups with 6 mice in each group as follows: Control, UA (10 mg/kg), UA (20 mg/kg). According to institutional guidelines, the mice were raised in a specific pathogen-free level barrier system [22°C; 50% humidity; 12:12 light/dark cycle; food and water were freely available (ad libitum)]. HCT-116 cells (5×106) were injected into the lateral tail vein of male nude mice to establish the lung metastatic model of colon cancer. After 1 week, the mice were injected intraperitoneally with different concentrations of UA (10 and 20 mg/kg) every other day for 4 weeks. After 6 weeks, all of the mice were sacrificed by cervical dislocation and the lung tissues were fully excised. In addition, the number of lung metastases was measured. The lung samples were processed into paraffin-embedded sections and stained with haematoxylin and eosin (H&E) to visualize the presence of metastasis. All animal experimental protocols were reviewed and approved (approval no. TJ-HB-LAC-2020-92) by the Animal Experimental Ethics Committee of Shanghai Tongfeng Huiji Biological Medicine Technology Co., Ltd. (Shanghai, China).

H&E staining

H&E staining was performed using an H&E Staining Kit (cat. no. G1120, Beijing Solarbio Science & Technology Co., Ltd.). Briefly, the steps of tissue fixation and slicing were same as those described for immunohistochemistry. After dewaxing and rehydrating, the sections were stained with hematoxylin for 2 min and with differentiation solution for 10 sec at room temperature. Subsequently, eosin staining was carried out for 30 sec. Finally, the sections were treated with dehydration, transparency and sealing. Images were captured using a light microscope at the magnification of ×40 and ×100.

The Cancer Genome Atlas (TCGA) data analysis

TCGA data analysis was performed using the publicly accessible UALCAN database (https://ualcan.path.uab.edu/index.html). The mRNA values of ARL4C were collected from healthy (normal) individuals and colon cancer patients, and the mRNA levels of ARL4C related to lymph node metastasis and tumor staging in patients with colon cancer were analyzed based on the TCGA dataset.

In vitro protein ubiquitination assay

HCT-116 and SW480 cells with ARL4C-OE and ARL4C-NC were treated with LY294002 (15 µmol/l) or UA (20 µmol/l) for 8 h at 37°C. The cells were lysed with 200 µl Pierce™ IP Lysis Buffer (cat. no. 87787, Thermo Fisher Scientific, Inc.) on ice for 15 min. Subsequently, the samples were immunoprecipitated with anti-Flag magnetic beads (cat. no. B26101, Selleck Chemicals) overnight at 4°C to pull down ARL4C protein. The immunoprecipitated beads were then washed three times with PBST containing 0.5% Tween-20 for 5 min at room temperature. The beads with 50 µl SDS-PAGE sample loading buffer (cat. no. P0015A, Beyotime Institute of Biotechnology) were then boiled for 5 min at 100°C. The resulting complexes were subjected to immunoblotting with anti-ubiquitin antibody (1:100; cat. no. ab134953; Abcam).

Statistical analysis

SPSS 21.0 software (IBM Corp.) was used for statistical analysis. All data were obtained from three independently replicated experiments and are expressed as the mean ± standard deviation. Comparisons between two groups were performed using an unpaired Student's t-test or the Mann-Whitney U test, while comparisons among ≥3 groups were performed using either one- or two-way ANOVA followed by the Fisher's least significant difference (LSD) test. P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of UA on the proliferation, migration and invasion of colon cancer cells

First, the present study investigated the effect of UA on the proliferation of HCT-116 and SW480 cells. By increasing the UA concentration, the proliferation of HCT-116 and SW480 cells gradually decreased at 24 or 48 h (Fig. 1A and B) and the pharmacological effect of the drug at 48 h was improved compared with that at 24 h. The IC50 value of UA for the HCT-116 cell line at 48 h was 20.47 µM while for the SW480 cell line it was 25.18 µM. Thus, three concentrations lower than this dose were selected in the subsequent experiment. Subsequently, Transwell assays were performed to verify the inhibitory effect of UA on the migration and invasion of colon cancer cells. The results revealed that UA inhibited the migration and invasion of HCT-116 and SW480 cells in a dose-dependent manner (Fig. 1C and D). These findings indicated that UA significantly inhibited the proliferation, migration and invasion of HCT-116 and SW480 cells.

UA inhibits the lung metastasis of colon cancer in vivo

The present study aimed to evaluate the effect of UA on colon cancer metastasis in vivo. To achieve this, a lung metastatic model of colon cancer was established by injecting HCT-116 cells into the tail vein of male BALB/c-nude mice. The organs of the lung were excised after 6 weeks and the number of lung metastatic tumours was measured. The findings indicated that UA could relieve the weight loss caused by colon cancer on day 42 (Fig. 2A). A significant decrease in the number of lung metastases was observed after administering low (10 mg/kg) and high (20 mg/kg) doses of UA, particularly at 20 mg/kg (Fig. 2B and C). Similarly, the presence of lung metastases from colon cancer was confirmed using H&E staining and a significant improvement in lung metastases was observed after UA treatment (Fig. 2D). Taken together, these results demonstrated that UA could inhibit the lung metastasis of colon cancer in vivo.

UA inhibits the expression of ARL4C in colon cancer in vitro and in vivo

A previous study reported that high expression of ARL4C is associated with poor prognosis, depth of tumour invasion and peritoneal metastasis (10). Therefore, UA may inhibit metastasis by targeting ARL4C. First, the present study analyzed the expression of ARL4C in normal samples and samples from patients with colon cancer by employing the UALCAN database from The Cancer Genome Atlas (TCGA). In Fig. 3A, the expression of ARL4C was revealed to be increased in colon cancer tissues compared with normal tissues. Subsequently, the association between ARL4C expression and nodal metastatic status or individual cancer stages was analysed using data from TCGA database. The data indicated that the expression level of ARL4C was positively associated with the number of lymph node metastases (Fig. 3B) and cancer stage (Fig. 3C). To further determine the role of ARL4C in human colon cancer tissue, the protein expression of ARL4C in 40 paired human colon cancer specimens was detected through immunohistochemistry. The results revealed that ARL4C was highly expressed in colon cancer tissues compared with the adjacent tissues (Fig. 3D and E). The effects of UA on the expression of ARL4C in HCT-116 and SW480 cells were then evaluated. Western blot analyses demonstrated that the expression level of ARL4C was significantly decreased after UA treatment (Fig. 3F-H). In addition, the protein expression of MMP2 was also decreased (Fig. 3F-H). Similarly, the effect of UA on the expression of ARL4C in mouse lung metastases was examined. The immunohistochemical results (Fig. 3I and J) revealed that the expression of ARL4C was higher in lung tissues not treated with UA while it was significantly decreased after treatment with high-dose UA. These findings indicated that ARL4C was highly expressed in human colon cancer tissue and was associated with local lymph node metastasis and tumour stage. UA effectively inhibited colon cancer metastasis in vivo and in vitro and the mechanism may be associated with the inhibition of ARL4C expression.

Overexpression of ARL4C reverses UA-inhibited migration and invasion

To explore whether the UA-inhibited migration and invasion was dependent on the ARL4C protein, ARL4C-overexpressing HCT-116 and SW480 cell lines were constructed using lentivirus transfection. The transfection efficiency was verified using western blot analysis (Fig. 4A-C). Subsequently, Transwell assays revealed that overexpression of ARL4C reversed the inhibitory effect of UA on cell migration and invasion, compared with cells only treated with UA (Fig. 4D and E). To rule out any toxic effect of UA in determining the decrease in cell migration and invasion, the assays were also performed at the 90% viability (IC10) concentration of UA. The results in Fig. S1 showed that low concentrations of UA inhibited cell migration and invasion, while overexpression of ARL4C reversed the inhibitory effect of UA. Western blot analysis revealed that the protein expression of ARL4C and MMP2 in the UA group was lower than that in the ARL4C overexpression group combined with the UA treatment (Fig. 4F and H). As MPP2 is a type of secreted protein (32), the secreted MPP2 was detected using ELISA with the cell supernatant. As expected, UA treatment decreased the secretion of MPP2, which was reversed by ARL4C overexpression (Fig. 4G and I). These findings indicated that the pharmacological effect of UA on colon cancer invasion and migration was mediated through the downregulation of ARL4C protein expression.

UA promotes ubiquitination of the ARL4C protein through the AKT/mTOR pathway

It was reported that the AKT/mTOR pathway regulated the expression of ARL4C in glioblastoma and lung cancer (33,34). Therefore, the present study further investigated whether the inhibitory effect of ARL4C by UA is associated with the inhibition of AKT signalling activation. First, ARL4C protein expression was decreased in the presence of the AKT inhibitor, LY294002 (Fig. 5A-C), or the mTOR inhibitor, rapamycin (Fig. 5D-F). In addition, UA significantly inhibited the phosphorylation of AKT and mTOR in HCT-116 and SW480 cells (Fig. 5G-J).

However, the mechanism by which AKT regulates ARL4C expression still requires investigation. As the stability of ARL4C is controlled by ubiquitination in hippocampal morphogenesis (35), MG-132, a proteasome inhibitor, was utilized. The results demonstrated that MG-132 reversed the decreased expression of the ARL4C protein caused by UA or LY294002 (Fig. 6A-F). Finally, using an immunoprecipitation experiment, it was observed that the ubiquitination level of ARL4C was increased after the administration of UA or LY294002 in HCT-116 and SW480 cells (Fig. 6G). Overall, these findings indicated that UA increased the ubiquitination of ARL4C by inhibiting the AKT pathway.

Discussion

The occurrence of colon cancer metastasis causes poor prognosis and increased mortality in patients with colon cancer. Therefore, identifying specific molecules and highly effective drugs is key to improving the survival rate. The present study demonstrated that UA significantly inhibited the migration and invasion of colon cancer cells by inhibiting the AKT signalling pathway and increasing the ubiquitination of the ARL4C protein.

Several previous studies have confirmed that ARL4C is involved in tumour growth and metastasis (36,37). The function and molecular mechanism of ARL4C have been revealed to be significantly different in different tumour types. For instance, ARL4C recruits IQ motif containing GTPase activating protein 1 (IQGAP1) and its downstream effector MMP14 to invasive pseudopodia in pancreatic cancer cells, which induces extracellular matrix degradation. This process is an important condition for the invasion of pancreatic cancer cells (38). Another study demonstrated that the downregulation of ARL4C significantly inhibits the proliferation, migration and invasion of clear cell renal cell carcinoma cells. Moreover, ARL4C promoted the progression of clear cell renal cell carcinoma and the occurrence of epithelial-mesenchymal transformation (EMT) (39). By contrast, other studies have showed that ARL4C is a tumour suppressor in ovarian and breast cancer and low expression of ARL4C was associated with poor prognosis in these patients (40,41). The present study revealed through TCGA database analysis that ARL4C was highly expressed in patients with colon cancer compared with healthy individuals. Moreover, the high expression of ARL4C was closely associated with colon cancer metastasis. Subsequently, the expression level of ARL4C was assessed in human colon cancer tissues. The findings of the present study revealed that the expression of ARL4C was higher in colon cancer tissues than in paracancerous tissues. This is consistent with the results revealed in a study by Chen et al (42). In addition, the present study demonstrated that overexpression of ARL4C can promote the invasion and metastasis of colon cancer cells and significantly increase the expression and secretion of MMP2 protein. These results indicated that ARL4C may be a potential target for colon cancer therapy.

UA is an effective anticancer agent originating from natural sources that is widely distributed in a large variety of traditional medicinal herbs, vegetables and fruits. Previous studies have reported that UA plays a role in cancer cell proliferation, metastasis, apoptosis and angiogenesis, which is due to its wide regulation of intracellular signalling pathways, including Wnt/β-catenin (43), the Hippo signalling pathway (44), the PI3K/AKT pathway (45) and STAT3 (21). For example, UA was revealed to inhibit the proliferation of colon cancer HT-29 cells and induce cancer cell apoptosis by inhibiting the EGFR/MAPK pathway (46). UA can target multiple signalling pathways to inhibit angiogenesis in colorectal cancer (31). The present study investigated the potential anticancer mechanism of UA in colon cancer metastasis both in vitro and in vivo. To the best of the authors' knowledge, it was determined for the first time that UA significantly suppresses colon cancer metastasis by downregulating the expression of ARL4C in vivo and in vitro. In addition, the overexpression of ARL4C reversed the inhibitory effect of UA. Thus, decreasing ARL4C expression is one of the mechanisms of action of UA in the treatment of colon cancer metastasis.

The AKT/mTOR pathway is a central node of several signalling pathways and is frequently deregulated in several types of human cancers (47). Activation of the AKT signalling pathway can promote angiogenesis, EMT and metastasis of tumour cells (48). Deng et al (49) reported that the activated PI3K/AKT/mTOR signalling pathway can induce EMT and enhance the expression of cancer stem cell markers in ovarian cancer. Zhang et al (50) reported that the PI3K/AKT/mTOR signalling pathway is an important regulator of the proliferation, migration and invasion of LoVo colon cancer cells. Previous studies demonstrated that the regulation of ARL4C expression is involved in multiple carcinogenic signalling pathways, such as Wnt/β-catenin (39), p53 (13), RAF1-MEK/ERK (51) and AKT/mTOR (34). In the present study, AKT or mTOR inhibitors regulated ARL4C protein expression in HCT-116 and SW480 colon cancer cells, which further identifies the critical role of AKT in regulating the ARL4C protein. Moreover, UA significantly inhibited the activation of the AKT/mTOR signalling pathway, which reveals the role of AKT in the antitumour effect of UA in colon cancer. In addition, MG-132 reversed the decreased expression of ARL4C protein caused by UA or LY294002, and UA and AKT signalling pathway inhibitors could induce the ubiquitination level of ARL4C. These data indicated that UA promotes the ubiquitination-mediated degradation of ARL4C through the AKT signalling pathway, which further clarifies the antitumour mechanism of UA.

The present study demonstrated that ARL4C is highly expressed in colon cancer tissues and promotes the occurrence of colon cancer metastasis. The AKT signalling pathway could regulate ARL4C expression and its ubiquitination. Furthermore, UA decreased ARL4C expression and inactivated the AKT signalling pathway, thus inhibiting the migration and invasion of colon cancer cells. Collectively, UA served as a novel solution to relieve colon cancer metastasis by inducing the ubiquitination-mediated degradation of ARL4C by modulating the AKT signalling pathway. UA may be a promising treatment option to prolong the survival of patients with colon cancer metastasis.

Supplementary Material

Supporting Data

Acknowledgements

We would like to thank Mrs Zixi Chen and Mrs Xiaoxiao Li from Shanghai University of Traditional Chinese Medicine Affiliated Putuo Hospital (Shanghai, China) for their professional assistance and support in revising the manuscript.

Funding

The present study was supported from the Scientific Research Fund of Shanghai Sixth People's Hospital Medical Group (grant no. 22-LY-02), the One Hundred Talents Project of Putuo Hospital, Shanghai University of Traditional Chinese Medicine (grant no. 2022-RCQH-03), the Science and Technology Innovation Project of Putuo District Health System (grant no. ptkwws202307) and the National Natural Science Foundation of China (grant no. 82305141).

Availability of data and materials

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

Authors' contributions

XK, RW and MZ conceived and designed the project. FX, YS and MZ completed the experiments and acquired the data. YS and RL wrote the article and prepared the images of the figures. QL and QG analyzed the data. MZ and FX confirm the authenticity of all the raw data. All authors read and approved the final version of this manuscript.

Ethics approval and consent to participate

The animal experimental protocols were reviewed and approved (approval no. TJ-HB-LAC-2020-92) by the Animal Experimental Ethics Committee of Shanghai Tongfeng Huiji Biological Medicine Technology Co., Ltd. (Shanghai, China). All procedures performed in studies involving human samples were approved (approval no. PTEC-A-2021-29-1) by the Ethics Committee of Putuo Hospital affiliated to Shanghai University of Traditional Chinese Medicine (Shanghai, China).

Patient consent to publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Xu W, He Y, Wang Y, Li X, Young J, Ioannidis JPA, Dunlop MG and Theodoratou E: Risk factors and risk prediction models for colorectal cancer metastasis and recurrence: An umbrella review of systematic reviews and meta-analyses of observational studies. BMC Med. 18:1722020. View Article : Google Scholar : PubMed/NCBI

3 

Liang Z, Xie H, Shen W, Shao L, Zeng L, Huang X, Zhu Q, Zhai X, Li K, Qiu Z, et al: The synergism of natural compounds and conventional therapeutics against colorectal cancer progression and metastasis. Front Biosci (Landmark Ed). 27:2632022. View Article : Google Scholar : PubMed/NCBI

4 

Islam MR, Akash S, Rahman MM, Nowrin FT, Akter T, Shohag S, Rauf A, Aljohani ASM and Simal-Gandara J: Colon cancer and colorectal cancer: Prevention and treatment by potential natural products. Chem Biol Interact. 368:1101702022. View Article : Google Scholar : PubMed/NCBI

5 

Burd CG, Strochlic TI and Setty SR: Arf-like GTPases: Not so Arf-like after all. Trends Cell Biol. 14:687–694. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Chiang TS, Wu HF and Lee FS: ADP-ribosylation factor-like 4C binding to filamin-A modulates filopodium formation and cell migration. Mol Biol Cell. 28:3013–3028. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Chiang TS, Lin MC, Tsai MC, Chen CH, Jang LT and Lee FS: ADP-ribosylation factor-like 4A interacts with Robo1 to promote cell migration by regulating Cdc42 activation. Mol Biol Cell. 30:69–81. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Matsumoto S, Fujii S, Sato A, Ibuka S, Kagawa Y, Ishii M and Kikuchi A: A combination of Wnt and growth factor signalling induces ARL4C expression to form epithelial tubular structures. EMBO J. 33:702–718. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Wei SM, Xie CG, Abe Y and Cai JT: ADP-ribosylation factor-like 7 (ARL7) interacts with alpha-tubulin and modulates intracellular vesicular transport. Biochem Biophys Res Commun. 384:352–356. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Matsumoto S, Fujii S and Kikuchi A: ARL4C is a key regulator of tubulogenesis and tumourigenesis as a target gene of Wnt-beta-catenin and growth factor-Ras signalling. J Biochem. 161:27–35. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Chen Q, Fu WJ, Tang XP, Wang L, Niu Q, Wang S, Lin Y, Cao MF, Hu R, Wen HY, et al: ADP-Ribosylation Factor Like GTPase 4C (ARL4C) augments stem-like traits of glioblastoma cells by upregulating ALDH1A3. J Cancer. 12:818–826. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Hu Q, Masuda T, Sato K, Tobo T, Nambara S, Kidogami S, Hayashi N, Kuroda Y, Ito S, Eguchi H, et al: Identification of ARL4C as a peritoneal dissemination-associated gene and its clinical significance in gastric cancer. Ann Surg Oncol. 25:745–753. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Xie N, Pan Y, Wu J, Bai Y, Xiao C, Gao X, Wang J and Liu N: MicroRNA-302s might regulate ARL4C-mediated gastric cancer progression via p53 signaling: Bioinformatics analysis and experiments validation. Onco Targets Ther. 14:2541–2553. 2021. View Article : Google Scholar : PubMed/NCBI

14 

Harada T, Matsumoto S, Hirota S, Kimura H, Fujii S, Kasahara Y, Gon H, Yoshida T, Itoh T, Haraguchi N, et al: Chemically modified antisense oligonucleotide against ARL4C inhibits primary and metastatic liver tumor growth. Mol Cancer Ther. 18:602–612. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Amin AR, Kucuk O, Khuri FR and Shin DM: Perspectives for cancer prevention with natural compounds. J Clin Oncol. 27:2712–2725. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Wozniak L, Skapska S and Marszalek K: Ursolic Acid-A pentacyclic triterpenoid with a wide spectrum of pharmacological activities. Molecules. 20:20614–20641. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Yin R, Li T, Tian JX, Xi P and Liu RH: Ursolic acid, a potential anticancer compound for breast cancer therapy. Crit Rev Food Sci Nutr. 58:568–574. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Castrejon-Jimenez NS, Leyva-Paredes K, Baltierra-Uribe SL, Castillo-Cruz J, Campillo-Navarro M, Hernandez-Perez AD, Luna-Angulo AB, Chacon-Salinas R, Coral-Vazquez RM, Estrada-Garcia I, et al: Ursolic and Oleanolic acids induce mitophagy in A549 human lung cancer cells. Molecules. 24:34442019. View Article : Google Scholar : PubMed/NCBI

19 

Lin CW, Chin HK, Lee SL, Chiu CF, Chung JG, Lin ZY, Wu CY, Liu YC, Hsiao YT, Feng CH, et al: Ursolic acid induces apoptosis and autophagy in oral cancer cells. Environ Toxicol. 34:983–991. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Mu D, Zhou G, Li J, Su B and Guo H: Ursolic acid activates the apoptosis of prostate cancer via ROCK/PTEN mediated mitochondrial translocation of cofilin-1. Oncol Lett. 15:3202–3206. 2018.PubMed/NCBI

21 

Kim K, Shin EA, Jung JH, Park JE, Kim DS, Shim BS and Kim SH: Ursolic acid induces apoptosis in colorectal cancer cells partially via upregulation of MicroRNA-4500 and inhibition of JAK2/STAT3 phosphorylation. Int J Mol Sci. 20:1142018. View Article : Google Scholar : PubMed/NCBI

22 

Lin JH, Chen SY, Lu CC, Lin JA and Yen GC: Ursolic acid promotes apoptosis, autophagy, and chemosensitivity in gemcitabine-resistant human pancreatic cancer cells. Phytother Res. 34:2053–2066. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Shanmugam MK, Manu KA, Ong TH, Ramachandran L, Surana R, Bist P, Lim LH, Kumar AP, Hui KM and Sethi G: Inhibition of CXCR4/CXCL12 signalling axis by ursolic acid leads to suppression of metastasis in transgenic adenocarcinoma of mouse prostate model. Int J Cancer. 129:1552–1563. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Zhang X, Li T, Gong ES and Liu RH: Antiproliferative activity of Ursolic acid in MDA-MB-231 human breast cancer cells through Nrf2 pathway regulation. J Agric Food Chem. 68:7404–7415. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Zhang L, Cai QY, Liu J, Peng J, Chen YQ, Sferra TJ and Lin JM: Ursolic acid suppresses the invasive potential of colorectal cancer cells by regulating the TGF-β1/ZEB1/miR-200c signalling pathway. Oncol Lett. 18:3274–3282. 2019.PubMed/NCBI

26 

Xiang F, Fan Y, Ni Z, Liu Q, Zhu Z, Chen Z, Hao W, Yue H, Wu R and Kang X: Ursolic acid reverses the chemoresistance of breast cancer cells to paclitaxel by targeting MiRNA-149-5p/MyD88. Front Oncol. 9:5012019. View Article : Google Scholar : PubMed/NCBI

27 

Zheng J, Wang SS, Shen KP, Huang XW, Li M, Chen L, Peng X, An HM and Hu B: Ursolic acid potentiated oxaliplatin to induce apoptosis in colorectal cancer RKO cells. Pharmazie. 75:246–249. 2020.PubMed/NCBI

28 

Zhang N, Liu S, Shi S, Chen Y, Xu F, Wei X and Xu Y: Solubilization and delivery of Ursolic acid for modulating tumour microenvironment and regulatory T cell activities in cancer immunotherapy. J Control Release. 320:168–178. 2020. View Article : Google Scholar : PubMed/NCBI

29 

Lin J, Chen Y, Wei L, Shen A, Sferra TJ, Hong Z and Peng J: Ursolic acid promotes colorectal cancer cell apoptosis and inhibits cell proliferation via modulation of multiple signalling pathways. Int J Oncol. 43:1235–1243. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Wang J, Liu L, Qiu H, Zhang X, Guo W, Chen W, Tian Y, Fu L, Shi D, Cheng J, et al: Ursolic acid simultaneously targets multiple signalling pathways to suppress proliferation and induce apoptosis in colon cancer cells. PLoS One. 8:e638722013. View Article : Google Scholar : PubMed/NCBI

31 

Lin J, Chen Y, Wei L, Hong Z, Sferra TJ and Peng J: Ursolic acid inhibits colorectal cancer angiogenesis through suppression of multiple signalling pathways. Int J Oncol. 43:1666–1674. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Ribeiro Vitorino T, Ferraz do Prado A, Bruno de Assis Cau S and Rizzi E: MMP-2 and its implications on cardiac function and structure: Interplay with inflammation in hypertension. Biochem Pharmacol. 215:1156842023. View Article : Google Scholar : PubMed/NCBI

33 

Chen Q, Weng HY, Tang XP, Lin Y, Yuan Y, Li Q, Tang Z, Wu HB, Yang S, Li Y, et al: ARL4C stabilized by AKT/mTOR pathway promotes the invasion of PTEN-deficient primary human glioblastoma. J Pathol. 247:266–278. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Sun Y, Zhang Z, Xiang F, Zhang M, Chen Q, Tang L, Zhu L, Liu J, Meng Z, Hu J, et al: Decreasing ARL4C expression by inhibition of AKT signal in human lung adenocarcinoma cells. Life Sci. 246:1174282020. View Article : Google Scholar : PubMed/NCBI

35 

Han JS, Hino K, Li W, Reyes RV, Canales CP, Miltner AM, Haddadi Y, Sun J, Chen CY, La Torre A and Simo S: CRL5-dependent regulation of the small GTPases ARL4C and ARF6 controls hippocampal morphogenesis. Proc Natl Acad Sci USA. 117:23073–23084. 2020. View Article : Google Scholar : PubMed/NCBI

36 

Kanai R, Uehara T, Yoshizawa T, Kamakura M, Nakajima T, Kinugawa Y, Iwaya M, Asaka S, Kitazawa M, Nagaya T and Ota H: ARL4C is associated with epithelial-to-mesenchymal transition in colorectal cancer. BMC Cancer. 23:4782023. View Article : Google Scholar : PubMed/NCBI

37 

Alkhatib DZR, Thi Kim Truong T, Fujii S, Hasegawa K, Nagano R, Tajiri Y and Kiyoshima T: Stepwise activation of p63 and the MEK/ERK pathway induces the expression of ARL4C to promote oral squamous cell carcinoma cell proliferation. Pathol Res Pract. 246:1544932023. View Article : Google Scholar : PubMed/NCBI

38 

Harada A, Matsumoto S, Yasumizu Y, Shojima K, Akama T, Eguchi H and Kikuchi A: Localization of KRAS downstream target ARL4C to invasive pseudopods accelerates pancreatic cancer cell invasion. Elife. 10:e667212021. View Article : Google Scholar : PubMed/NCBI

39 

Zhang P, Xu Y, Chen S, Wang Z, Zhao L, Chen C, Kang W, Han R, Qiu J, Wang Q, et al: ARL4C regulates the progression of clear cell renal cell carcinoma by affecting the Wnt/β-catenin signaling pathway. J Oncol. 2022:27245152022.PubMed/NCBI

40 

Su D, Katsaros D, Xu S, Xu H, Gao Y, Biglia N, Feng J, Ying L, Zhang P, Benedetto C and Yu H: ADP-ribosylation factor-like 4C (ARL4C), a novel ovarian cancer metastasis suppressor, identified by integrated genomics. Am J Transl Res. 7:242–256. 2015.PubMed/NCBI

41 

Li L, Sun RM and Jiang GQ: ATF3 Demethylation promotes the transcription of ARL4C, which acts as a tumor suppressor in human breast cancer. Onco Targets Ther. 13:3467–3476. 2020. View Article : Google Scholar : PubMed/NCBI

42 

Chen X, Su Z, Wang S and Xu H: Clinical and prognostic significance of ARL4C expression in colorectal cancer. Cancer Biomark. 16:253–257. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Kim JH, Kim YH, Song GY, Kim DE, Jeong YJ, Liu KH, Chung YH and Oh S: Ursolic acid and its natural derivative corosolic acid suppress the proliferation of APC-mutated colon cancer cells through the promotion of beta-catenin degradation. Food Chem Toxicol. 67:87–95. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Kim SH, Jin H, Meng RY, Kim DY, Liu YC, Chai OH, Park BH and Kim SM: Activating hippo pathway via Rassf1 by Ursolic acid suppresses the tumorigenesis of gastric cancer. Int J Mol Sci. 20:47092019. View Article : Google Scholar : PubMed/NCBI

45 

Lee KC, Chen YL, Lin PY and Chuang WL: Ursolic Acid-induced apoptosis via regulation of the PI3K/Akt and MAPK signaling pathways in Huh-7 cells. Molecules. 23:20162018. View Article : Google Scholar : PubMed/NCBI

46 

Shan JZ, Xuan YY, Zheng S, Dong Q and Zhang SZ: Ursolic acid inhibits proliferation and induces apoptosis of HT-29 colon cancer cells by inhibiting the EGFR/MAPK pathway. J Zhejiang Univ Sci B. 10:668–674. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Revathidevi S and Munirajan AK: Akt in cancer: Mediator and more. Semin Cancer Biol. 59:80–91. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Harsha C, Banik K, Ang HL, Girisa S, Vikkurthi R, Parama D, Rana V, Shabnam B, Khatoon E, Kumar AP and Kunnumakkara AB: Targeting AKT/mTOR in oral cancer: Mechanisms and advances in clinical trials. Int J Mol Sci. 21:32852020. View Article : Google Scholar : PubMed/NCBI

49 

Deng J, Bai X, Feng X, Ni J, Beretov J, Graham P and Li Y: Inhibition of PI3K/Akt/mTOR signalling pathway alleviates ovarian cancer chemoresistance through reversing epithelial-mesenchymal transition and decreasing cancer stem cell marker expression. BMC Cancer. 19:6182019. View Article : Google Scholar : PubMed/NCBI

50 

Zhang X, Shi H, Tang H, Fang Z, Wang J and Cui S: miR-218 inhibits the invasion and migration of colon cancer cells by targeting the PI3K/Akt/mTOR signalling pathway. Int J Mol Med. 35:1301–1308. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Fujii S, Ishibashi T, Kokura M, Fujimoto T, Matsumoto S, Shidara S, Kurppa KJ, Pape J, Caton J, Morgan PR, et al: RAF1-MEK/ERK pathway-dependent ARL4C expression promotes ameloblastoma cell proliferation and osteoclast formation. J Pathol. 256:119–133. 2022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2024
Volume 51 Issue 2

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
Zhang M, Xiang F, Sun Y, Liu R, Li Q, Gu Q, Kang X and Wu R: Ursolic acid inhibits the metastasis of colon cancer by downregulating ARL4C expression. Oncol Rep 51: 27, 2024
APA
Zhang, M., Xiang, F., Sun, Y., Liu, R., Li, Q., Gu, Q. ... Wu, R. (2024). Ursolic acid inhibits the metastasis of colon cancer by downregulating ARL4C expression. Oncology Reports, 51, 27. https://doi.org/10.3892/or.2023.8686
MLA
Zhang, M., Xiang, F., Sun, Y., Liu, R., Li, Q., Gu, Q., Kang, X., Wu, R."Ursolic acid inhibits the metastasis of colon cancer by downregulating ARL4C expression". Oncology Reports 51.2 (2024): 27.
Chicago
Zhang, M., Xiang, F., Sun, Y., Liu, R., Li, Q., Gu, Q., Kang, X., Wu, R."Ursolic acid inhibits the metastasis of colon cancer by downregulating ARL4C expression". Oncology Reports 51, no. 2 (2024): 27. https://doi.org/10.3892/or.2023.8686