Open Access

N‑myc downstream‑regulated gene 1 inhibits the proliferation and invasion of hepatocellular carcinoma cells via the regulation of integrin β3

  • Authors:
    • Yan Song
    • Guangping Wu
    • Mingyang Zhang
    • Qianqian Kong
    • Juan Du
    • Yabing Zheng
    • Longtao Yue
    • Lili Cao
  • View Affiliations

  • Published online on: March 27, 2017     https://doi.org/10.3892/ol.2017.5924
  • Pages: 3599-3607
  • Copyright: © Song 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

N-myc downstream-regulated gene 1 (NDRG1) is a multifunctional protein associated with carcinogenesis and tumor progression. The function of NDRG1 in hepatocellular carcinoma (HCC) cells remains controversial. The present study investigated the role of NDRG1 in HCC as well as its molecular mechanism using a range of techniques, including western blot analysis, cellular proliferation test, wound healing assay and Transwell assay. In HCC, the levels of NDRG1 expression were highest in the cytoplasm, followed by the membrane, and were lowest in the nucleus. NDRG1 was revealed to inhibit the proliferation and invasion of BEL7402 cells, which facilitated the hypothesis that NDRG1 expression levels may be lower in cell line with a high metastatic potential compared with those in cell lines with a low metastatic potential. However, the present study identified that NDRG1 expression was higher in detached BEL7402 cells and MHCC‑97H cells compared with that in attached BEL7402 cells and MHCC‑97L cells. Thus, this finding was contrary to what was expected, suggesting that NDRG1 overexpression in the HCC with a high metastatic potential may be the compensatory mechanism. The human HCC BEL7402 cell line demonstrated a significant increase in the capability of motility, invasion and cellular proliferation following NDRG1‑short hairpin RNA transfection. Integrin β3 (ITGB3) protein expression was increased in NDRG1‑downregulated BEL7402 cells and SMMC7721 cells compared with that in the control cells. The present study suggested that NDRG1 may be a potential anti‑tumor target for the treatment of patients with HCC. A potential mechanism for these roles of NDRG1 is by regulating ITGB3 expression; however, this requires additional investigation.

Introduction

Liver cancer is the fifth most frequently diagnosed type of cancer worldwide in males; however, it is the second most common cause of cancer-associated mortality. In women, liver cancer is the seventh most commonly diagnosed type of cancer and the sixth leading cause of cancer-associated mortality (1). Hepatocellular carcinoma (HCC) is the most common form of liver cancer, and accounts for ~70–85% of all cases (1). Patients with HCC typically exhibit few early symptoms, and thus, have low early diagnostic rates. Cases are usually confirmed at a late stage, which is past the most opportunistic time for surgery (2). Due to poor prognosis, metastasis and recurrence are likely to occur following surgery, the 5-year survival rate for patients with HCC is 30–40% (3). Current efforts are focused on identifying reliable biomarkers to predict HCC occurrence and development.

The N-myc downstream-regulated gene 1 (NDRG1; also termed Drg1, cap43, RTP, Rit2 and PROXY-1) belongs to the NDRG gene family. The NDRG1 gene is positioned at chromosome location 8q24.2 (4) or 8q24.3 (5). The NDRG1 gene has 60,085 bp, including 16 exons and 15 introns, and encodes a 2997-bp RNA with a 1182-bp region that translates into the NDRG1 protein (4). The NDRG1 protein is ~42,835 Da in length, and consists of 394 amino acids (6,7). In normal liver tissue, the NDRG1 protein is generally expressed in biliary epithelial cells but not in hepatocytes (8). In addition, NDRG1 protein staining is not affected by the condition of the liver (such as hepatitis or cirrhosis) or the type of infecting hepatitis virus (9).

In hepatoma cells, NDRG1 expression has been observed to be significantly increased compared with normal hepatocytes, and NDRG1 protein was generally expressed in the cytoplasm and membrane, but rarely in the nucleus (810). However, a previous in vitro study revealed that DNA damage increased the levels of NDRG1 nuclear expression (11). Previously, NDRG1 has been defined as an anti-tumor gene in several cancers due to its involvement in tumor invasion, metastasis and proliferation (1216). However, theories regarding the function of NDRG1 vary across different studies. For example, a previous study stated that NDRG1 suppressed tumor growth in HCC (17). By contrast, other studies have suggested that NDRG1 exerted a stimulating effect on HCC (18,19). Therefore, the present study utilized a series of experiments in order to clarify the function of the NDRG1 gene in different HCC cell lines in vitro, and used a polymerase chain reaction (PCR) array test to determine downstream associated genes that may also be regulated by NDRG1. The present study identified that knockdown of NDRG1 expression resulted in the upregulation of several genes, including integrin β3 (ITGB3).

ITGB3, also termed platelet glycoprotein III and cluster of differentiation 61, belongs to the integrin family. Within this family of cell surface receptors and adhesion molecules, ITGB3 regulates cellular proliferation, migration, cell survival and cell morphology, performs a main role in the processes of cell adhesion and movement, and can affect tumor growth and metastasis (20). However, the exact role of ITGB3 during tumor growth and metastasis requires additional studies. The integrin αvβ3 and αIIbβ3 are members of the ITGB3 family. Integrin αvβ3 is mainly expressed on the surface of endothelial cells, smooth muscle cells, monocytes and platelets (21). Integrin αvβ3 has been observed to affect tumor angiogenesis and is also strongly expressed in malignant tumor angiogenic endothelial cells (22). Integrin αvβ3 could promote tumor cell growth and angiogenesis in melanoma and breast cancer (23,24) and blocking integrin αvβ3 resulted in reduced proliferation and invasion in ovarian cancer (25). Furthermore, integrin αIIbβ3 is mainly expressed in platelets and megakaryocytes, and it has been observed to regulate platelet interacting with tumor cells, which might help to tumor metastatic spread (20). Currently, correlation studies between NDRG1 and ITGB3 have not been reported. In the present, study it was shown that knockdown of NDRG1 expression resulted in the upregulation of ITGB3. However, the regulatory mechanism between NDRG1 gene expression and ITGB3 function requires additional investigation.

Materials and methods

Tissue samples

The tissue microarray (Hliv-HCC150cs-01) was purchased from Shanghai Outdo Biotech Co., Ltd. (Shanghai, China), and included 75 specimens of cancerous HCC tissue and corresponding adjacent degenerative tissue. Table I describes the patient characteristics of the study population.

Table I.

Characteristics of the study population (n=75).

Table I.

Characteristics of the study population (n=75).

Categorical variablesPatients, n (%)
Gender
  Female11 (14.7)
  Male64 (85.3)
Age, years (mean ± standard deviation)52.5±10.5
AJCC cancer staging
  I23 (30.7)
  II26 (34.7)
  III23 (30.7)
  IV3 (4.0)

[i] AJCC, American Joint Committee on Cancer.

Cell lines and culture

The human HCC BEL7402 and SMMC7721 cell lines, the human colon carcinoma SW480 and SW620 cell lines, a cell line with a high metastatic potential (MHCC-97H) and a cell line with a low metastatic potential (MHCC-97L) were all obtained from the Cell Bank of Type Culture Collection of Chinese Academy of Sciences (Shanghai, China). The BEL7402, SMMC7721, SW480 and SW620 cell lines were maintained in Roswell Park Memorial Institute (RPMI)-1640 medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA). The MHCC-97H and MHCC-97L cell lines were maintained in Dulbecco's modified Eagle's medium (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% (v/v) fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and 1% antibiotic solution (Gibco™ Penicillin-Streptomycin; Thermo Fisher Scientific, Inc.) and incubated at 37°C in a humidified incubator under 5% CO2. The detach BEL7402 cells mimic the metastatic cancer cells in the vascular system. Access method of attach and detach BEL7402 cells was used according to our previous description (26).

Immunohistochemistry (IHC)

The IHC staining method was used according to a previous description (27).

Plasmid construction and transfection

Three candidate NDRG1 knockdown plasmids were purchased from Shanghai Genechem Co., Ltd. (Shanghai, China). The BEL7402 cells were transfected with the short hairpin (sh) RNA plasmid using the jetPRIME® transfection reagent (Polyplus-transfection, Illkirch, France) according to the manufacturer's protocol. The shRNA sequence was 5′-CTCTAAACAACCCTGAGAT-3′, and was designed according to the sequence provided under the GenBank accession number NM_001258432.1 (28). The shRNA sequence was inserted into the eukaryotic expression vector GV102 (GeneChem Co., Ltd., Shanghai, China). The manufacturer's protocol was followed for the transfection, using non-transfected and untreated cells as controls. Total RNA was extracted 48 h post-transfection, and total protein was extracted 72 h post-transfection. Western blot analysis and quantitative (q) PCR were used to validate the effects of NDRG1 downregulation in transfected HCC cells.

Western blot analysis

Experiments were conducted at least three times. Total protein was extracted using a radio immunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology, Haimen, China) and separated using SDS-PAGE (12% separating gel and 5% stacking gel). A total of 20 µg of protein was loaded into each well. The target proteins were then transferred onto polyvinylidene fluoride membranes (EMD Millipore, Billerica, MA, USA). Subsequent to being blocked with 5% non-fat milk for 2 h at room temperature, the membranes were incubated with a primary anti-human NDRG1 antibody (catalog no. PA5-18109; dilution, 1:1,000; Pierce, Thermo Fisher Scientific, Inc.) or anti-human ITGB3 antibody (catalog no. 13,166; dilution 1:1,000; Cell Signaling Technology, Inc., Danvers, MA, USA) at 4°C overnight. The membranes were then incubated with a secondary peroxidase-conjugated affiniPure rabbit anti-goat IgG antibody (cat. no. ZB2306; dilution, 1:10,000; Zhong Shan Jin Qiao Inc., Beijing, China) for NDRG1 and goat anti-rabbit IgG antibody (cat. no. ZB2301; dilution, 1:10,000; Zhong Shan Jin Qiao Inc.) for integrin β3 at room temperature for 1 h. Rabbit anti-GAPDH (dilution, 1:1,000; Hangzhou Goodhere Biotechnology Co., Ltd., Hangzhou, China) was used as the control. An enhanced chemiluminescence method was used to visualize western blot results (cat. no. WBLUC0100; EMD Millipore). Protein band densitometry was measured using Image J2x (National Institutes of Health, Bethesda, MA, USA).

qPCR

Experiments were conducted at least three times. BEL7402 cells transfected with empty plasmid vector (BEL7402vec) and BEL7402 cells were as negative controls. Total RNA was extracted from transfected and non-transfected BEL7402 cells using the Quick-RNA extraction reagent (Ambion; Thermo Fisher Scientific, Inc., USA). qPCR was performed using the SYBR Green Premix Ex Taq™ (catalog no. RR420Q; Takara Bio, Inc., Otsu, Japan) following the manufacturer's protocol, using 35 cycles of 94°C for 30 sec and 60°C for 30 sec. The primers for NDRG1 were as follows: Forward, 5′-AACCCACACAGTCACCCTGC-3′ and reverse, 5′-ACTCCACCACGGCATCCACT-3′. The primers for GAPDH were as follows: Forward, 5′-GAGAAGTATGACAACAGCCTCAA-3′ and reverse, 5′-TGAGTCCTTCCACGATACCAA-3′. The 2−∆∆Cq method was used as the normalization method for analysis (29).

PCR array assay

The RT2 Profiler™ PCR Array Human Tumor Metastasis kit was purchased from Qiagen, Inc. (catalog no. 330231; Valencia, CA, USA). The procedures for RNA purification, complementary DNA first strand synthesis and qPCR were conducted according to the manufacturer's protocols. The primers for 84 genes known to be involved in metastasis were provided. The reaction system for qPCR was prepared according to the manufacturer's protocol, using 40 cycles of 95°C for 15 sec and 60°C for 60 sec. Briefly, 20 µl of the total reaction mixture was added to each well of a 96-well plate containing 5 µg RNA. Data analysis was completed using the Web-based PCR Array Data Analysis Software on the Qiagen website (http://pcrdataanalysis.sabiosciences.com/pcr/arrayanalysis.php).

Cellular proliferation test

A cell suspension of 100 µl (20,000 cells) was inoculated in each well of a 96-well plate. Subsequently, cells were cultured at 37°C in a humidified incubator with 5% CO2 overnight. Cellular proliferation was assessed using a Cell Counting Kit-8 assay (CCK-8; Dojindo Molecular Technologies, Inc., Kumamoto, Japan) assay according to the manufacturer's protocol. In briefly, 10 µl of the CCK-8 assay solution was added to each well and incubated at 37°C in a humidified incubator with 5% CO2 for 2 h. Absorbance values were then measured at 450 nm with a spectrophotometer.

Wound healing assay

A total of 1×106 cells were seeded into 35-mm dishes, and when they reached 90% confluence, a scratch was created with a 200-µl pipette tip. Subsequently, cells were cultured in a serum-free RPMI-1640 medium at 37°C in a humidified incubator with 5% CO2 for the next 48 h. Micrographs were captured at 0 and 48 h. Three separate studies were conducted, and Image-Pro Plus version 6.0 (Media Cybernetics, Inc., Rockville, MD, USA) was used for data analysis.

Transwell migration and Matrigel invasion assays

For Matrigel invasion assay, 1×105 cells were seeded in the upper chamber with Matrigel (NO. 356,234; BD Biosciences, USA) in serum-free medium for 20 h as previously described (30). Transwell migration assay was conducted without Matrigel and cultured for 6 h as previously described (30). Subsequently, the membrane was swabbed to remove the cells in the upper chamber, and the membrane was stained with 0.5% crystal violet dissolved in methanol. Image-Pro Plus version 6.0 was used to count the number of cells that adhered to the membrane of the inserts. A total of three replicates were performed.

Statistical analysis

All data were processed using the SPSS 19.0 statistical software program (IBS SPSS, Armonk, NY, USA). Comparisons between 2 groups were performed using an independent sample t-test and comparisons among >2 groups were performed using one-way analysis of variance. Correlation analyses were conducted using the Spearman's rank correlation coefficient analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

NDRG1 protein expression in HCC tissue

The present study analyzed the characteristics of NDRG1 protein expression in HCC tissue. The NDRG1 protein was primarily expressed in the cytoplasm, membrane and nucleus of cancerous cells and the corresponding adjacent degenerative cells. The NDRG1 protein immunoreactivities differed in various subcellular locations; cytoplasm immunoreactivity was the strongest, followed by that in the membrane, and was weakest within the nucleus. No significant difference was identified between NDRG1 expression in the cytoplasm or nucleus of cancerous cells compared with that of the corresponding adjacent degenerative cells. However, membrane expression in cancer cells was significantly higher compared with the expression of NDRG1 in adjacent degenerative cells (P<0.05; Fig. 1A-C). There was also a negative correlation between the NDRG1 cytoplasm staining in cancerous cells and tumor-node-metastasis (TNM) stage (P<0.01; rs=−0.333; Fig. 1D) (31). In other words, increased NDRG1 cytoplasm expression was observed in tumors with earlier TNM stages compared with tumors with later TNM stages. A positive correlation was observed between NDRG1 membrane expression and cytoplasmic expression in cancer cells (P<0.05; rs=0.173; Fig. 1E). A negative correlation was also identified between membrane expression in cancerous cells and cytoplasmic expression in adjacent degenerative cells (P<0.05; rs=−0.208; Fig. 1F). A positive correlation was also observed between NDRG1 nucleus expression in adjacent degenerative cells and cancer cells (P<0.05; rs=0.265; Fig. 1G).

NDRG1 expression levels in different cell culture models

The present study analyzed the expression of NDRG1 mRNA in different cell culture models using qPCR. The results revealed an increase in NDRG1 messenger (m)RNA expression in detached human HCC BEL7402 cells compared with that in attached BEL7402 cells (Fig. 2A). The NDRG1 mRNA expression levels were also higher in MHCC-97H cells compared with those in MHCC-97L cells (P<0.05; Fig. 2B). The NDRG1 mRNA expression levels in colon carcinoma SW620 cells were lower when compared with those in colon carcinoma SW480 cells (P<0.05; Fig. 2C).

NDRG1 expression is significantly downregulated in NDRG1-shRNA-transfected BEL7402 cells

To investigate NDRG1 expression following the knockdown of the NDRG1 gene with shRNA, the present study performed a western blot analysis and qPCR on transfected and control cells. The western blot analysis and qPCR demonstrated that NDRG1 expression was significantly decreased in the BEL7402 cells transfected with NDRG1-shRNA compared with that in the control cells (P<0.05; Fig. 3A-C).

NDRG1 inhibits invasion and metastasis in BEL7402 cells

Transwell migration assay demonstrated that NDRG1-shRNA BEL7402 cells were significantly more capable of motility compared with BEL7402 cells transfected with empty plasmid vector (P<0.05; Fig. 4A and B). The Matrigel invasion assay revealed that the BEL7402 cells had a significantly higher invasive ability with NDRG1 knockdown (P<0.05), indicating that NDRG1 may inhibit HCC cell invasion in vitro (Fig. 4C and D). The wound healing assay also showed that NDRG1-shRNA BEL7402 cells were significantly more capable of movement compared with BEL7402 cells transfected with empty plasmid vector (P<0.05; Fig. 4E and F). Therefore, NDRG1 had the ability to inhibit HCC cell migration and invasion in vitro.

NDRG1 inhibits BEL7402 cellular proliferation

The CCK-8 assay was performed to detect the proliferation of BEL7402 cells at a 48 h following transfection with NDRG1-shRNA. The results demonstrated that BEL7402 cellular proliferation was enhanced due to NDRG1 knockdown. Therefore, NDRG1 may inhibit BEL7402 cellular proliferation at a level that was statistically significant (P<0.05; Fig. 5).

Downstream molecular candidates of NDRG1 inhibit tumor metastasis

The PCR array assay was used to determine the downstream gene candidates of NDRG1 that inhibited tumor metastasis. The results demonstrated that NDRG1 knockdown upregulated ITGB3, MMP10 and SERPINE1 and downregulated TNFSF10 in both BEL7402 and SMMC7721 cell lines (Fig. 6A). Furthermore, western blot analysis was used to determine ITGB3 protein expression. The results demonstrated that ITGB3 protein expression was increased in NDRG1-downregulated BEL7402 and SMMC7721 cells compared with that in the control cells, indicating that the NDRG1 gene may suppress tumor metastasis by regulating ITGB3 expression (P<0.05; Fig. 6B and C).

Discussion

Currently, the function of the NDRG1 gene is controversial (1719); therefore, the present study investigated the role of NDRG1 in HCC as well as its molecular mechanism. Firstly, NDRG1 expression was detected in HCC tissue and cells. The present results demonstrated that NDRG1 was expressed in cancerous liver cells and adjacent degenerative liver cells. A previous study observed that NDRG1 was expressed in 6% of cirrhosis and benign liver lesions subsequent to staining (10). These results indicated that NDRG1 may participate in the full progression of the occurrence and development of HCC, from liver cell degeneration to malignant changes. Although NDRG1 stained stronger on the membrane of liver cancerous cells compared with that in the adjacent degenerative liver cells, there was no staining difference in the cytoplasm or nucleus. As mentioned previously, the expression of the NDRG1 gene was always negative in normal liver cells (810,18). Therefore, the levels of NDRG1 expression in liver cell membrane may estimate the degree of injury in cells or the extent of cell cancerization.

A previous study demonstrated that high NDRG1 expression in patients with HCC was associated with a short overall survival rate and a poor prognosis. High NDRG1 expression was detected in poorly differentiated HCC and high TNM stages (10). However, when the present study considered NDRG1 expression in different subcellular localizations, it identified that there was a negative correlation between the cytoplasmic expression of NDRG1 in cancerous liver cells and the TNM stage, which indicates that higher expression levels were observed in smaller sized tumors. In our previous study, it was revealed that the cytoplasmic expression of NDRG1 may be associated with lymph node metastasis (27).

The expression of NDRG1 may therefore become a marker of the degree of invasiveness for local tumors, although the NDRG1 expression observed in the nucleus of the cancerous and degenerative liver cells in the present study may be coincidental. However, there was a positive correlation between the expression of NDRG1 in the nucleus of cancerous liver cells and that in the adjacent degenerative liver cells, indicating that the regulating factor of NDRG1 nuclear expression acts equally on cancerous and degenerative liver cells. However, WoLF PSORT (an advanced protein subcellular localization prediction tool) and amino acid sequence analysis demonstrated that the NDRG1 protein lacked the motifs used for localization in nucleus (32). Thus, NDRG1 nucleus localization may rely on protein phosphorylation. Several phosphorylation sites have been identified in the C-terminal site of the NDRG1 protein (33).

Reports have shown that serum- and glucocorticoid-induced protein kinase 1 could phosphorylate NDRG1 on the amino acids Thr328, Ser330, Thr346, Thr356 and Thr366 (34,35). Glycogen synthesis kinase 3β (GSK3β) was shown to be able to phosphorylate Ser342, Ser352 and Ser362 of NDRG1 (36). These results are valuable to understanding the role of NDRG1, since protein phosphorylation is reversible and affects NDRG1 subcellular localization in the cell (37). In addition, NDRG1 can localize in the nucleus subsequent to binding to the 70-kDa heat shock cognate protein (Hsc70) (38). Hsc70 is a molecular chaperone that mediates mast cell transport between the cytoplasm and nucleus (39).

NDRG1 was revealed to inhibit the proliferation and invasion of BEL7402 cells, which facilitated the hypothesis that NDRG1 expression levels may be lower in a cell line with a high metastatic potential compared with those in a cell line with a low metastatic potential. However, higher expression of NDRG1 mRNA was detected in MHCC-97H cells compared with that in MHCC-97L cells. In addition, higher expression of NDRG1 mRNA was detected in detached BEL7402 cells compared with that in attached BEL7402 cells. Thus, these findings were contrary to the expected results, and suggest that NDRG1 overexpression may be the compensatory mechanism. To clarify the expression trend of different metastatic potential cells lines, the present study detected the NDRG1 mRNA expression of colon carcinoma SW480 and SW620 cells. SW480 cells and SW620 cells were respectively derived from primary tumor and metastatic lymph nodes of the same person, which may make clear the expression of NDRG1 in primary and metastatic tissues. The result showed that NDRG1 expression was lower in SW620 compared with SW480 cells, which was contrary to the results of HCC cell lines. Maybe the NDRG1 expression was tissue specific. It has been reported that the level of RNA polymerase II bound to the NDRG1 promoter was lower in SW620 cells compared with SW480 cells, which reduced histone H4 acetylation, and enhanced histone H3 Ser10 phosphorylation (40). The unique histone modifications may be the possible mechanism for the different expression of NDRG1 in cell lines of different metastatic potential (40).

A previous study identified that NDRG1 inhibited the proliferation and invasion of cancer cells. In prostate cancer cells, the aberrant methylation of NDRG1 CpG islands caused downregulation of the NDRG1 promotor, leading to accelerated cellular proliferation and invasion of cancer cells (41). To illustrate the function of NDRG1 in HCC, the present study performed a series of in vitro experiments. Knockdown of the NDRG1 gene increased the proliferation of BEL7402 cells, indicating that NDRG1 inhibits the proliferation of hepatoma cells in vitro, which is consistent with the results of a previous study (17). However, a study in Hep3B and HepG2 cells revealed that the downregulation of NDRG1 decreased cell growth (19). A previous study indicated that NDRG1 directly interacted with GSK3β and Nur77 (also known as NR4A1, nuclear receptor subfamily 4 group A member 1) (42). This interaction could prevent the degradation of β-catenin, and consequently regulate β-catenin-relevant downstream signaling pathways to promote the growth of Hep3B and HepG2 cells (42). A previous study demonstrated that NDRG1 induced G0/G1-phase cell cycle arrest in HCC cell lines, and may incorporate cell cycle regulators such as p21 and cyclin-dependent kinase 4 in the NDRG1-induced cell cycle arrest (17). The suppression of NDRG1 inhibited tumor growth by inducing extensive cellular senescence in HCC cells (17,43).

To confirm the controversial function of NDRG1 in the metastasis of HCC, NDRG1 was knocked down using NDRG1-shRNA in BEL7402 cells in the present study, demonstrating that NDRG1 downregulation promotes HCC cell invasion and metastasis. This result indicates that NDRG1 inhibits the invasion and metastasis of HCC cells. A previous study demonstrated that NDRG1 overexpression maintained membrane E-cadherin and β-catenin levels while inhibiting transforming growth factor (TGF)-β-stimulated cellular migration and invasion (44), and this finding indirectly confirmed the results of the present study. A previous study demonstrated a potential mechanism by which NDRG1 effectively inhibited rat prostate cancer AT6.1 cells from metastasizing to the lungs (45). This mechanism may also regulate the cell structural protein actin. Therefore, NDRG1 could affect the formation and regulation of actin filaments by inhibiting the rho-associated protein kinase 1/phosphorylated myosin light chain 2 signaling pathway (46). A recent study has also demonstrated that NDRG1 could suppress tumor metastasis by inhibiting the focal adhesion kinase/paxillin signaling pathway (47).

To investigate the mechanism by which NDRG1 is involved in the proliferation and metastasis of HCC, the present study screened the downstream gene candidates regulated by NDRG1 that inhibited tumor metastasis. The results demonstrated that, in the BEL7402 and SMMC7721 cell lines, a decrease in NDRG1 expression led to an increase in ITGB3 expression. The expression levels of the ITGB3 protein were detected using western blot analysis, and were consistent with the PCR array results. A previous study has demonstrated that integrin αvβ3 could increase adhesion and invasion of HCC (48). ITGB3 could enhance the TGF-β1/H2O2/HOCl-inducing invasive ability of non-metastatic HCC cells via the TGF-β1 signaling pathway (49).

In addition, reducing the levels of integrin αvβ3 suppressed the cathepsin B-induced proliferation of HCC (50). These previous studies confirmed the findings of the present study. Integrin signaling induced cytoskeleton organization change and contraction, which consequently promoted cellular migration. A potential mechanism by which this happens could be that activated integrin αvβ3 was not effective for the primary tumor growth, but it could collaborate with platelets in order to promote tumor cell metastasis from the bloodstream (51). To the best of our knowledge, the association between NDRG1 and ITGB3 has not been studied to date. The role of NDRG1 on the inhibition of tumor metastasis and cellular proliferation was considered to occur via the regulation of ITGB3. The microarray results from colon cancer tissue demonstrated a negative correlation between NDRG1 expression in the nucleus of cancerous cells and ITGB3 stromal expression in adjacent degenerative cells. A negative correlation was also revealed between NDRG1 expression in the nucleus of cancerous cells and ITGB3 expression in cancer tissue. These results suggest that NDRG1 is involved in regulating ITGB3 expression at the transcriptional level (data not shown).

In conclusion, as a key signaling molecule regulating tumor proliferation and metastasis, NDRG1 is likely to become the target of anti-tumor metastasis or to be a biomarker observing the prognosis and metastasis of patients with tumors. In order to be considered as a drug target, additional studies are required on NDRG1 in order to identify the molecular mechanism behind its anti-tumor activity. The present study identified that NDRG1 inhibited the proliferation and metastasis of HCC by regulating ITGB3 expression at the transcriptional level. However, the association between NDRG1 and ITGB3 requires additional investigation through in vitro and in vivo experiments.

Acknowledgements

The present study was supported by grants from the Natural Science Foundation of China (grant no. 30873025), the Natural Science Foundation of Shandong Province (grant nos. ZR2010HM033 and ZR2014HM016) and the Science and Technology Development of Shandong Province (grant no. 2016GSF201118).

Glossary

Abbreviations

Abbreviations:

NDRG1

N-myc downstream-regulated gene 1

HCC

hepatocellular carcinoma

ITGB3

integrin β3

CCK-8

Cell Counting Kit-8

MMP10

matrix metalloprotease 10

SERPINE1

serine protease inhibitor clade E member 1

TNFSF10

tumor necrosis factor receptor superfamily member 10

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Bruix J and Llovet JM: Prognostic prediction and treatment strategy in hepatocellular carcinoma. Hepatology. 35:519–524. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Aravalli RN, Steer CJ and Cressman EN: Molecular mechanisms of hepatocellular carcinoma. Hepatology. 48:2047–2063. 2008. View Article : Google Scholar : PubMed/NCBI

4 

van Belzen N, Dinjens WN, Eussen BH and Bosman FT: Expression of differentiation-related genes in colorectal cancer: Possible implications for prognosis. Histol Histopathol. 13:1233–1242. 1998.PubMed/NCBI

5 

Thierry-Mieg D and Thierry-Mieg J: AceView: A comprehensive cDNA-supported gene and transcripts annotation. Genome Biol. 7:(Suppl 1). S12.1–14. 2006. View Article : Google Scholar

6 

van Belzen N, Dinjens WN, Diesveld MP, Groen NA, van der Made AC, Nozawa Y, Vlietstra R, Trapman J and Bosman FT: A novel gene which is up-regulated during colon epithelial cell differentiation and down-regulated in colorectal neoplasms. Lab Invest. 77:85–92. 1997.PubMed/NCBI

7 

Zhou D, Salnikow K and Costa M: Cap43, a novel gene specifically induced by Ni2+ compounds. Cancer Res. 58:2182–2189. 1998.PubMed/NCBI

8 

Sibold S, Roh V, Keogh A, Studer P, Tiffon C, Angst E, Vorburger SA, Weimann R, Candinas D and Stroka D: Hypoxia increases cytoplasmic expression of NDRG1, but is insufficient for its membrane localization in human hepatocellular carcinoma. FEBS Lett. 581:989–994. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Akiba J, Ogasawara S, Kawahara A, Nishida N, Sanada S, Moriya F, Kuwano M, Nakashima O and Yano H: N-myc downstream regulated gene 1 (NDRG1)/Cap43 enhances portal vein invasion and intrahepatic metastasis in human hepatocellular carcinoma. Oncol Rep. 20:1329–1335. 2008.PubMed/NCBI

10 

Chua MS, Sun H, Cheung ST, Mason V, Higgins J, Ross DT, Fan ST and So S: Overexpression of NDRG1 is an indicator of poor prognosis in hepatocellular carcinoma. Mod Pathol. 20:76–83. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Kurdistani SK, Arizti P, Reimer CL, Sugrue MM, Aaronson SA and Lee SW: Inhibition of tumor cell growth by RTP/rit42 and its responsiveness to p53 and DNA damage. Cancer Res. 58:4439–4444. 1998.PubMed/NCBI

12 

Lee JC, Chung LC, Chen YJ, Feng TH and Juang HH: N-myc downstream-regulated gene 1 downregulates cell proliferation, invasiveness, and tumorigenesis in human oral squamous cell carcinoma. Cancer Lett. 355:242–252. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Kim-Fuchs C, Winterhalder S, Winter A, Malinka T, Born D, Schäfer S, Stroka D, Gloor B, Candinas D and Angst E: The silencing of N-myc downstream-regulated gene-1 in an orthotopic pancreatic cancer model leads to more aggressive tumor growth and metastases. Dig Surg. 31:135–142. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Hu ZY, Xie WB, Yang F, Xiao LW, Wang XY, Chen SY and Li ZG: NDRG1 attenuates epithelial-mesenchymal transition of nasopharyngeal cancer cells via blocking Smad2 signaling. Biochim Biophys Acta. 1852:1876–1886. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Chang X, Xu X, Ma J, Xue X, Li Z, Deng P, Zhang S, Zhi Y, Chen J and Dai D: NDRG1 expression is related to the progression and prognosis of gastric cancer patients through modulating proliferation, invasion and cell cycle of gastric cancer cells. Mol Biol Rep. 41:6215–6223. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Hosoya N, Sakumoto M, Nakamura Y, Narisawa T, Bilim V, Motoyama T, Tomita Y and Kondo T: Proteomics identified nuclear N-myc downstream-regulated gene 1 as a prognostic tissue biomarker candidate in renal cell carcinoma. Biochim Biophys Acta. 1834:2630–2639. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Akiba J, Murakami Y, Noda M, Watari K, Ogasawara S, Yoshida T, Kawahara A, Sanada S, Yasumoto M, Yamaguchi R, et al: N-myc downstream regulated gene1/Cap43 overexpression suppresses tumor growth by hepatic cancer cells through cell cycle arrest at the G0/G1 phase. Cancer Lett. 310:25–34. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Cheng J, Xie HY, Xu X, Wu J, Wei X, Su R, Zhang W, Lv Z, Zheng S and Zhou L: NDRG1 as a biomarker for metastasis, recurrence and of poor prognosis in hepatocellular carcinoma. Cancer Lett. 310:35–45. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Yan X, Chua MS, Sun H and So S: N-Myc down-regulated gene 1 mediates proliferation, invasion, and apoptosis of hepatocellular carcinoma cells. Cancer Lett. 262:133–142. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Desgrosellier JS and Cheresh DA: Integrins in cancer: Biological implications and therapeutic opportunities. Nat Rev Cancer. 10:9–22. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Tadokoro S, Tomiyama Y, Honda S, Kashiwagi H, Kosugi S, Shiraga M, Kiyoi T, Kurata Y and Matsuzawa Y: Missense mutations in the beta(3) subunit have a different impact on the expression and function between alpha(IIb)beta(3) and alpha(v)beta(3). Blood. 99:931–938. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Varner JA and Cheresh DA: Tumor angiogenesis and the role of vascular cell integrin alphavbeta3. Important Adv Oncol. 69–87. 1996.PubMed/NCBI

23 

Brooks PC, Strömblad S, Klemke R, Visscher D, Sarkar FH and Cheresh DA: Antiintegrin alpha v beta 3 blocks human breast cancer growth and angiogenesis in human skin. J Clin Invest. 96:1815–1822. 1995. View Article : Google Scholar : PubMed/NCBI

24 

Petitclerc E, Strömblad S, von Schalscha TL, Mitjans F, Piulats J, Montgomery AM, Cheresh DA and Brooks PC: Integrin alpha(v)beta3 promotes M21 melanoma growth in human skin by regulating tumor cell survival. Cancer Res. 59:2724–2730. 1999.PubMed/NCBI

25 

Landen CN, Kim TJ, Lin YG, Merritt WM, Kamat AA, Han LY, Spannuth WA, Nick AM, Jennnings NB, Kinch MS, et al: Tumor-selective response to antibody-mediated targeting of alphavbeta3 integrin in ovarian cancer. Neoplasia. 10:1259–1267. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Kong Q, Wu G, Han L, Zhang Z, Du J, Sun W and Cao L: A transfection method of PS-asODNs targeting ANGPTL4 in multicellular structures of hepatocarcinoma cell line. Cancer Gene Ther. 22:285–290. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Song Y, Lv L, Du J, Yue L and Cao L: Correlation of N-myc downstream-regulated gene 1 subcellular localization and lymph node metastases of colorectal neoplasms. Biochem Biophys Res Commun. 439:241–246. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Homo sapiens N-myc downstream regulated 1 (NDRG1), transcript variant 3, mRNA. NCBI Reference Sequence: NM_001258432.1. https://www.ncbi.nlm.nih.gov/nuccore/386643031

29 

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

30 

Cao L, Han L, Zhang Z, Li J, Qu Z, Du J, Liang X, Liu Y, Liu H, Shi Y, et al: Involvement of anoikis-resistance in the metastasis of hepatoma cells. Exp Cell Res. 315:1148–1156. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Edge SB and Compton CC: The American joint committee on cancer: The 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol. 17:1471–1474. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Horton P, Park KJ, Obayashi T, Fujita N, Harada H, Adams-Collier CJ and Nakai K: WoLF PSORT: Protein localization predictor. Nucleic Acids Res. 35:(Web Server issue). W585–W587. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Bandyopadhyay S, Wang Y, Zhan R, Pai SK, Watabe M, Iiizumi M, Furuta E, Mohinta S, Liu W, Hirota S, et al: The tumor metastasis suppressor gene Drg-1 down-regulates the expression of activating transcription factor 3 in prostate cancer. Cancer Res. 66:11983–11990. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Inglis SK, Gallacher M, Brown SG, McTavish N, Getty J, Husband EM, Murray JT and Wilson SM: SGK1 activity in Na+ absorbing airway epithelial cells monitored by assaying NDRG1-Thr346/356/366 phosphorylation. Pflugers Arch. 457:1287–1301. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Hoang B, Frost P, Shi Y, Belanger E, Benavides A, Pezeshkpour G, Cappia S, Guglielmelli T, Gera J and Lichtenstein A: Targeting TORC2 in multiple myeloma with a new mTOR kinase inhibitor. Blood. 116:4560–4568. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Murray JT, Campbell DG, Morrice N, Auld GC, Shpiro N, Marquez R, Peggie M, Bain J, Bloomberg GB, Grahammer F, et al: Exploitation of KESTREL to identify NDRG family members as physiological substrates for SGK1 and GSK3. Biochem J. 384:477–488. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Segawa T, Nau ME, Xu LL, Chilukuri RN, Makarem M, Zhang W, Petrovics G, Sesterhenn IA, McLeod DG, Moul JW, et al: Androgen-induced expression of endoplasmic reticulum (ER) stress response genes in prostate cancer cells. Oncogene. 21:8749–8758. 2002. View Article : Google Scholar : PubMed/NCBI

38 

Lachat P, Shaw P, Gebhard S, van Belzen N, Chaubert P and Bosman FT: Expression of NDRG1, a differentiation-related gene, in human tissues. Histochem Cell Biol. 118:399–408. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Sugiki T, Taketomi Y, Kikuchi-Yanoshita R, Murakami M and Kudo I: Association of N-myc downregulated gene 1 with heat-shock cognate protein 70 in mast cells. Biol Pharm Bull. 27:628–633. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Li Q and Chen H: Transcriptional silencing of N-Myc downstream-regulated gene 1 (NDRG1) in metastatic colon cancer cell line SW620. Clin Exp Metastasis. 28:127–135. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Ma W, Na M, Tang C, Wang H and Lin Z: Overexpression of N-myc downstream-regulated gene 1 inhibits human glioma proliferation and invasion via phosphoinositide 3-kinase/AKT pathways. Mol Med Rep. 12:1050–1058. 2015.PubMed/NCBI

42 

Lu WJ, Chua MS, Wei W and So SK: NDRG1 promotes growth of hepatocellular carcinoma cells by directly interacting with GSK-3β and Nur77 to prevent β-catenin degradation. Oncotarget. 6:29847–29859. 2015.PubMed/NCBI

43 

Lu WJ, Chua MS and So SK: Suppressing N-Myc downstream regulated gene 1 reactivates senescence signaling and inhibits tumor growth in hepatocellular carcinoma. Carcinogenesis. 35:915–922. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Chen Z, Zhang D, Yue F, Zheng M, Kovacevic Z and Richardson DR: The iron chelators Dp44mT and DFO inhibit TGF-β-induced epithelial-mesenchymal transition via up-regulation of N-Myc downstream-regulated gene 1 (NDRG1). J Biol Chem. 287:17016–17028. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Bandyopadhyay S, Pai SK, Gross SC, Hirota S, Hosobe S, Miura K, Saito K, Commes T, Hayashi S, Watabe M and Watabe K: The Drg-1 gene suppresses tumor metastasis in prostate cancer. Cancer Res. 63:1731–1736. 2003.PubMed/NCBI

46 

Sun J, Zhang D, Zheng Y, Zhao Q, Zheng M, Kovacevic Z and Richardson DR: Targeting the metastasis suppressor, NDRG1, using novel iron chelators: Regulation of stress fiber-mediated tumor cell migration via modulation of the ROCK1/pMLC2 signaling pathway. Mol Pharmacol. 83:454–469. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Wangpu X, Lu J, Xi R, Yue F, Sahni S, Park KC, Menezes S, Huang ML, Zheng M, Kovacevic Z and Richardson DR: Targeting the metastasis suppressor, N-Myc downstream regulated gene-1, with novel Di-2-pyridylketone thiosemicarbazones: Suppression of tumor cell migration and cell-collagen adhesion by inhibiting focal adhesion kinase/paxillin signaling. Mol Pharmacol. 89:521–540. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Tang NH, Chen YL, Wang XQ, Li XJ, Wu Y, Zou QL and Chen YZ: N-terminal and C-terminal heparin-binding domain polypeptides derived from fibronectin reduce adhesion and invasion of liver cancer cells. BMC Cancer. 10:5522010. View Article : Google Scholar : PubMed/NCBI

49 

Feng XX, Liu M, Yan W, Zhou ZZ, Xia YJ, Tu W, Li PY and Tian DA: β3 integrin promotes TGF-β1/H2O2/HOCl-mediated induction of metastatic phenotype of hepatocellular carcinoma cells by enhancing TGF-β1 signaling. PLoS One. 8:e798572013. View Article : Google Scholar : PubMed/NCBI

50 

Xu ZZ, Xiu P, Lv JW, Wang FH, Dong XF, Liu F, Li T and Li J: Integrin alphavbeta3 is required for cathepsin B-induced hepatocellular carcinoma progression. Mol Med Rep. 11:3499–3504. 2015.PubMed/NCBI

51 

Weber MR, Zuka M, Lorger M, Tschan M, Torbett BE, Zijlstra A, Quigley JP, Staflin K, Eliceiri BP, Krueger JS, et al: Activated tumor cell integrin αβ3 cooperates with platelets to promote extravasation and metastasis from the blood stream. Thromb Res. 140:(Suppl 1). S27–S36. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 13 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song Y, Wu G, Zhang M, Kong Q, Du J, Zheng Y, Yue L and Cao L: N‑myc downstream‑regulated gene 1 inhibits the proliferation and invasion of hepatocellular carcinoma cells via the regulation of integrin β3. Oncol Lett 13: 3599-3607, 2017
APA
Song, Y., Wu, G., Zhang, M., Kong, Q., Du, J., Zheng, Y. ... Cao, L. (2017). N‑myc downstream‑regulated gene 1 inhibits the proliferation and invasion of hepatocellular carcinoma cells via the regulation of integrin β3. Oncology Letters, 13, 3599-3607. https://doi.org/10.3892/ol.2017.5924
MLA
Song, Y., Wu, G., Zhang, M., Kong, Q., Du, J., Zheng, Y., Yue, L., Cao, L."N‑myc downstream‑regulated gene 1 inhibits the proliferation and invasion of hepatocellular carcinoma cells via the regulation of integrin β3". Oncology Letters 13.5 (2017): 3599-3607.
Chicago
Song, Y., Wu, G., Zhang, M., Kong, Q., Du, J., Zheng, Y., Yue, L., Cao, L."N‑myc downstream‑regulated gene 1 inhibits the proliferation and invasion of hepatocellular carcinoma cells via the regulation of integrin β3". Oncology Letters 13, no. 5 (2017): 3599-3607. https://doi.org/10.3892/ol.2017.5924