Knockdown of metadherin inhibits cell proliferation and migration in colorectal cancer

  • Authors:
    • Jian‑Wang Li
    • Chun‑Zhen Huang
    • Jian‑Hua Li
    • Jian‑Hua Yuan
    • Qiong‑Hui Chen
    • Wei‑Fang Zhang
    • Zhen‑Sheng Xu
    • Ying‑Ping Liu
    • Yong Li
    • Mei‑Xiao Zhan
    • Li‑Gong Lu
  • View Affiliations

  • Published online on: July 17, 2018     https://doi.org/10.3892/or.2018.6581
  • Pages: 2215-2223
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Metadherin (MTDH) is a multifunctional oncogene involved in tumor cell migration and metastasis through regulating a number of oncogenic signaling pathways in various human malignancies. Previous studies have demonstrated that MTDH is overexpressed in human colorectal cancer (CRC) and associated with cancer progression and a poor prognosis. However, the underlying mechanisms remain largely unknown. The present study investigated the expression and role of MTDH in CRC cells as well as the underlying mechanism of this. Western blot analysis and quantitative polymerase chain reaction were conducted to determine protein and mRNA expression of MTDH in three human CRC cell lines. A short hairpin RNA (shRNA) targeting MTDH was introduced into CRC HCT116 cells to stably inhibit MTDH expression. A Cell Counting Kit‑8 assay, colony formation assay, Transwell assay and flow cytometry were used to investigate the effect of MTDH‑knockdown on cell proliferation, migration, apoptosis and cell cycle arrest. Western blotting was performed to examine the protein expression levels of cell growth‑ and apoptosis‑associated genes. The results demonstrated that MTDH was commonly expressed in CRC cell lines. MTDH silencing significantly suppressed cell growth, colony forming ability and migration while inducing the apoptosis of HCT116 cells. In addition, MTDH depletion induced S phase cell cycle arrest in HCT116 cells. Mechanistically, knockdown of MTDH markedly downregulated the expression of phosphorylated protein kinase B, c‑Myc, proliferating cell nuclear antigen and B‑cell lymphoma 2 (Bcl‑2) protein in HCT116 cells, and the expression of p53 and Bcl‑2‑associated X protein was significantly increased compared with the negative control shRNA group (P<0.05), suggesting that MTDH may function through the expression of numerous types of apoptosis‑associated and signaling channel proteins in CRC cells. Taken together, these data indicated that MTDH may serve as a biomarker and candidate therapeutic target for CRC.

Introduction

Colorectal cancer (CRC) is the third most common type of cancer worldwide, accounting for 8% of all cancer-related mortality (1). There is an urgent requirement to identify potential therapeutic targets to effectively inhibit CRC cell growth, invasion and metastasis (2,3). It is well-established that cancer progression is associated with genetic and epigenetic alterations along with the constructional changes in the CRC microenvironment. The activities of several signaling pathways, including Ras, Wnt and Myc signaling, have been correlated with CRC carcinogenesis, which provides potential biomarkers for early diagnosis and prognosis of CRC as well as therapeutic targets for CRC (4,5).

Metadherin (MTDH), also known as astrocyte elevated gene 1 (AEG-1) and lysine-rich CEACAM1 co-isolated (LYRIC), is located on chromosome 8q22 and encodes a single-pass transmembrane protein (6). MTDH has been reported to be overexpressed in solid tumors and promotes tumor cell proliferation, migration and invasion (710). Clinical studies have also demonstrated that MTDH overexpression is associated with tumorigenesis, tumor development and short survival times in hepatocellular carcinoma (HCC), gastric and breast cancer, and CRC (1114). MTDH-mediated tumor progression is regulated by multiple signaling pathways, including nuclear factor-κB (NF-κB), phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt), mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) and Wnt (1518). The overexpression of MTDH in CRC is significantly correlated with the Union for International Cancer Control stages, Ki-67 expression, histological differentiation and shorter survival times (19). However, the expression and role of MTDH in CRC as well as the underlying mechanism remain largely unknown.

The present study determined the protein and mRNA expression level of MTDH in human CRC cell lines and investigated its role in CRC cell behavior, including proliferation, colony-forming and migratory abilities, cell cycle arrest, and apoptosis in vitro. The present study also investigated the underlying molecular mechanism of MTDH-regulated CRC growth by detecting the protein expression of important cell growth- and apoptosis-associated genes in MTDH-deficient CRC cells.

Materials and methods

Cell lines

CRC SW480, HCT116 and LoVo cell lines (Type Culture Collection of the Chinese Academy of Sciences, Shanghai, China), and the normal colonic mucosa epithelial NCM460 cell line (American Type Culture Collection, Manassas, VA, USA) were cultured in Dulbecco's modified Eagle's medium (DMEM; Corning Incorporated, Corning, NY, USA), containing 10% fetal bovine serum (FBS; Shanghai VIAN-SAGA Biotech Ltd., Shanghai, China), streptomycin (100 µg/ml), and penicillin (١٠٠ IU/ml) at 37°C in a humidified atmosphere of 5% CO2.

Reverse transcription polymerase chain reaction (RT-PCR)

Extraction of total RNA from cells was performed using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). RNA was reverse transcribed to cDNA using a PrimeScript RT reagent kit (Takara Biotechnology Co., Ltd., Dalian, China). PCR amplification was conducted using the following primer sequences: MTDH forward, 5′-AAGCAGTGCAAAACAGTTCACG-3′ and reverse, 5′-GCACCTTATCACGTTTACGCT-3′; and GAPDH forward, 5′-TGACTTCAACAGCGACACCCA-3′ and reverse, 5′-CACCCTGTTGCTGTAGCCAAA-3′. The thermocycling conditions were 95°C for 15 sec, 45 cycles at 95°C for 5 sec, and 60°C for 30 sec.

Lentivirus-mediated short hairpin RNA (Lenti-shRNA) against MTDH

The Lenti-shRNA vector system (pGCSIL-GFP-puromycin) was constructed, packaged and purified by Shanghai GeneChem Co., Ltd. (Shanghai, China). Cells were seeded in 6-well plates at 3×105 cells/well using Lipofectamine 2000 transfection reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The following 3 specific MTDH short hairpin RNAs (shRNAs): shMTDH-1, 5′AGGAATAAAGGATTCTGAT3′; shMTDH-2, 5′-AAGTCAAATACCAAGCAAA-3′; and shMTDH-3, 5′-AACTTACAACCGCATCATT-3′, as well as a negative control shRNA (shNC), 5′-TTCTCCGAACGTGTCACGT-3′. The cells were cultured for the next 48 h and were then harvested for RT-PCR and simple western blot analysis or prepared for the following experiments.

RT-quantitative PCR (RT-qPCR)

Isolation of total RNA was conducted using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). The sequences of the primers (Shanghai GeneChem Co., Ltd.) used for qPCR were as follows: MTDH forward, 5′-AAGCAGTGCAAAACAGTTCACG-3′ and reverse, 5′-CACCCTGTTGCTGTAGCCAAA-3′; and GAPDH forward, 5′-AAGCAGTGCAAAACAGTTCACG-3′ and reverse, 5′-GCACCTTATCACGTTTACGCT-3′. qPCR was performed using a SYBR Master Mix (Takara Biotechnology Co., Ltd.) on a Stratagene MX3000p Real-time PCR thermocycler (Agilent Technologies, Inc., Santa Clara, CA, USA) with an initial denaturation step at 95°C for 10 min, followed by 45 cycles of 95°C for 3 sec and 60°C for 30 sec. The experiments were performed in triplicate. mRNA levels of MTDH were analyzed using the 2−ΔΔCq method (20).

Size-based simple and traditional western blot analyses

MTDH proteins following shRNA-mediated knockdown for simple western analysis of total receptor levels were diluted to a final protein concentration of 1X sample buffer, 1X fluorescent molecular weight markers and 40 mM DTT, prior to being heated at 95°C for 5 min in a Master mix (ProteinSimple, San Jose, CA, USA) and processed at room temperature in a Sally Sue Simple Western instrument (ProteinSimple). Proteins were subsequently incubated with anti-MTDH (cat. no., Ab45338; dilution, 1:20; Abcam, Cambridge, MA, USA) and anti-β-actin (cat no., SC-69879; dilution, 1:50; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) primary antibodies, Wes 12–230 kDa Master kit with Split Buffer (cat. nos., Ab45338 and 77960; ProteinSimple) and Wes 12–230 kDa Rabbit Master Kit (cat. nos., Ab45338 and 77961; ProteinSimple). Milk-free Antibody Diluent II (cat. no., ZLI-9030; dilution, 1:50; OriGene Technologies, Inc., Beijing, China) was used to dilute primary antibodies and as a blocking reagent at 4°C for 2 h. Proteins were then incubated with anti-rabbit (cat no., PS-MK١٤; dilution, 1:1,000; ProteinSimple) immunoglobulin G antibodies or anti-mouse immunoglobulin G antibodies (cat no., PS-MK15; dilution, 1:1,000; ProteinSimple) secondary antibodies. All antibodies were diluted in Immunobooster (Bioworld Technology, Inc., St. Louis Park, MN, USA) or antibody diluent (ProteinSimple) with a dilution of 1:50 or 1:100. All antibody incubations were performed at 4°C for 10–15 min with Immunobooster or 60–120 min with antibody diluents. Simple Western assay buffers, nano-volume capillaries and the prepared assay plate were placed in Simon (ProteinSimple), which performs all assay steps automatically. Next Luminol-S and peroxide (ProteinSimple) were added to produce chemiluminescence, which was captured by a CCD camera. Compass 2.5.11 Software (ProteinSimple) was used to analyze digital images. Of the three cell lines, the highest level of MTDH mRNA and protein expression was observed in the HCT116 cells and therefore, these cells were used to examine the function of MTDH in CRC cell behavior in the subsequent experiments.

Traditional western blot analysis was performed. Proteins were isolated using lysis buffer containing 1 mM EDTA, 50 mM Tris-HCl, 1% NP40, 0.1% SDS, 150 mM NaCl and protease inhibitor. Protein concentrations were measured using a bicinchoninic acid protein assay kit. A total of 5 µg protein/lane was loaded onto a 12% SDS-PAGE gel and separated, followed by transfer onto a polyvinylidene difluoride membrane. The membrane was then blocked with 2% dry skimmed milk in Tris-buffered saline with Tween-20 (TBST) for 1 h at room temperature, prior to being incubated with primary antibodies against C-Myc (cat no., 6341S; dilution, 1:1,000; Cell Signaling Technology, Inc., Danvers, MA, USA), GAPDH (cat. no., SC-32233, dilution, 1:4,000; Santa Cruz Biotechnology, Inc.), B-cell lymphoma 2 (Bcl-2; dilution, 1:1,000; Abcam), Bcl-2-associated X protein (BAX; cat. no., AB7977; dilution, 1:1,000; Abcam), phosphorylated protein kinase B (p-Akt; cat. no., 13038; dilution, 1:1,000; Cell Signaling Technology, Inc.), Akt (cat no., 9272, dilution, 1:1,000; Cell Signaling Technology, Inc.), p53 (cat no., 2527, dilution, 1:1,000; Cell Signaling Technology, Inc.) and proliferating cell nuclear antigen (PCNA; cat. no., 2586S; dilution, 1:1,000; Cell Signaling Technology, Inc.) overnight at 4°C. Following three rinses with TBST, the membrane was incubated with a horseradish peroxidase-conjugated rabbit IgG secondary antibody (cat no., sc-2004; dilution, 1:5,000; Santa Cruz Biotechnology, Inc.) and mouse IgG (cat no., sc-2005; dilution, 1:5,000; Santa Cruz Biotechnology, Inc.) for 1 h at room temperature. The protein bands were detected using an enhanced chemiluminescence kit (Thermo Fisher Scientific, Inc.) and analyzed using Bio-Rad 680 Quantity One software (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Cell proliferation assay

The proliferation rate of HCT116 cells was evaluated by a Cell Counting Kit-8 (CCK-8; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). A total of 5,000 stably-transfected cells were seeded into each well of a 96-well plate in 6 replicates and grown overnight. A total of 10 µl CCK-8 reagent was added into each well at different time points. Following incubation at 37°C for 2 h, absorbance was measured at 450 nm with a Bio-Rad plate reader (Bio-Rad Laboratories, Inc.).

Colony formation assay

A total of 2,000 stably-transfected HCT116 cells were plated into each well of a 6-well plate in triplicate. Following incubation at 37°C for 10 days, the cells were stained with crystal violet for 10 min at room temperature. Colonies containing >50 cells were counted under an Olympus IX71 inverted light microscope (magnification, ×100; Olympus Corporation, Tokyo, Japan). The images were captured using an Olympus digital camera (Olympus Corporation).

Cell apoptosis assay

Stably-transfected HCT116 cells were collected and resuspended in binding buffer (Invitrogen; Thermo Fisher Scientific, Inc.). On the seventh day after transfection, cell apoptosis was examined using the Annexin V-allophycocyanin (APC) apoptosis detection kit (cat. no., 88-8007-72; eBioscience; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. A FACS Calibur flow cytometer (cat. no., 557706; BD Biosciences, Franklin Lakes, NJ, USA) was used to analyze the percentage of apoptotic cells. The cells were stained with 100 µl cell suspension containing ٥ µl Annexin V-APC at room temperature in the dark for ١٠-15 min. All experiments were performed in triplicate. WinMDI 2.8 software was used for analysis.

Cell cycle analysis

Stably-transfected HCT116 cells were collected, rinsed with PBS and fixed with 70% ethanol overnight at 4°C. Following a 30 min incubation with 100 µg/ml RNase A, the cells were stained with ٤٠ mg/ml PI (cat. no., P4170; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and 100 µg/ml RNase A (cat. no., EN0531; Fermentas; Thermo Fisher Scientific, Inc.) in the dark for an additional 30 min at room temperature. The results were analyzed using a FACSCalibur flow cytometer (cat. no., 557706; BD Biosciences). The results were analyzed using Multicycle software (version 300; Phoenix Flow System, San Diego, CA, USA).

Transwell migration assay

Transwell assays were conducted using Transwell chambers (Corning Incorporated, Corning, NY, USA) in a 24-well format. A total of 2×105 HCT116 cells in 0.1% FBS-containing serum-free RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.) were loaded into the upper chamber and 30% FBS-containing medium was added into the lower chamber. Cells were incubated for 48 h at 37°C. The migrating cells stained with crystal violet at 37°C for ١ h were counted in random microscopic fields under an Olympus IX71 inverted light microscope (magnification, ×100; Olympus Corporation). The experiments were performed in triplicate.

Statistical analysis

All experiments were independently repeated at least three times. Data are expressed as the mean ± standard deviation. Statistical significance was evaluated using Student's t-test or one-way analysis of variance with Prism 4.0 (GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

MTDH mRNA is highly expressed in human CRC cells

The expression pattern of MTDH in human CRC SW480, HCT116 and LoVo cell lines, and normal colonic mucosa epithelial NCM460 cell line was examined by RT-qPCR and western blot analysis. The results demonstrated that the MTDH mRNA and protein expression levels were significantly higher in the human CRC cells than in the NCM460 cells (Fig. 1A and B). In addition, a higher level of MTDH mRNA was observed in HCT116 cells compared with that in the SW480 or LoVo cells. Therefore, HCT116 cells were used to examine the function of MTDH in CRC cell behavior in the subsequent experiments.

MTDH is essential for HCT116 cell proliferation in vitro

To investigate the role of MTDH in CRC cell proliferation, lentiviral vectors expressing shMTDH or shNC were introduced into HCT116 cells to stably silence MTDH expression. Fluorescent staining, simple western and qPCR assays were used to assess the knockdown efficacy of shMTDH. As demonstrated in Fig. 2A, cells were infected with a lentivirus containing MTDH shRNA (shMTDH) or an empty vector (shNC). The protein expression of MTDH was significantly inhibited by shMTDH in HCT116 cells compared with the negative control (Fig. 2B). Additionally, the mRNA expression of MTDH was also significantly decreased in shMTDH-transfected cells compared with that in shNC cells (Fig. 2C). These data suggested that MTDH expression was efficiently inhibited by shMTDH. The infection efficiency was 94.2% in the shMTDH-2 group relative to shNC. Since shMTDH-2 exhibited improved inhibition of MTDH expression in HCT116 cells, it was selected for subsequent experiments. To determine whether MTDH is essential for CRC cell growth, cell viability and colony formation assays were performed in a MTDH-deficient HCT116 cell line. As demonstrated in Fig. 3A, knockdown of MTDH notably inhibited HCT116 cell proliferation compared with the negative control. In addition, a colony formation assay revealed that shMTDH markedly suppressed the colony-forming ability of HCT116 cells compared with shNC (Fig. 3B). Taken together, these data indicated that MTDH is essential for HCT116 cell growth in vitro and may serve a promotive role in CRC tumorigenesis.

Knockdown of MTDH significantly induces HCT116 cell apoptosis

Cell apoptosis is critically important in cell growth suppression (21). To investigate the mechanism underlying MTDH-mediated HCT116 cell growth, cell apoptosis analysis was performed using PI-APC-Annexin in HCT116 cells. As demonstrated in Fig. 3C, flow cytometry revealed that the percentage of apoptosis was significantly increased to 14.84% (SD=0.22%, P<0.01) in the MTDH-shRNA HCT116 group, from 3.05% (SD=0.05%) in the shNC group, suggesting that knockdown of MTDH may induce cell apoptosis, resulting in an inhibition of cell proliferation in CRC cells.

Knockdown of MTDH induces cell cycle arrest at the S phase

To further investigate the mechanism underlying MTDH-mediated growth and apoptosis of HCT116 cells, a cell cycle analysis was performed. As demonstrated in Fig. 4, compared with the negative control, MTDH deficiency in HCT116 cells induced a significant increase in the cell population in the S phase (48.97 vs. 26.61%, P<0.01), indicating that MTDH may serve an essential role in S phase arrest in CRC cells.

MTDH is required for CRC cell migration

To further investigate whether MTDH has an effect on CRC cell migration, a Transwell migration assay was performed to test the migratory ability of MTDH-deficient HCT116 cells. As demonstrated in Fig. 5, MTDH deficiency significantly suppressed the migratory ability of HCT116 cells compared with the negative control, suggesting that MTDH may promote CRC metastasis.

Knockdown of MTDH inhibits p-Akt and c-Myc, and increases apoptosis-related protein expression

To further investigate the underlying molecular mechanism of MTDH-mediated CRC cell growth and migration, the present study examined the activity of Akt/c-Myc signaling and apoptosis-related protein in shMTDH-transfected HCT116 cells using western blot analysis. As demonstrated in Fig. 6, knockdown of MTDH markedly downregulated the expression of p-Akt, c-Myc and Bcl-2 protein in HCT116 cells, and the expression of p53 and Bax protein was significantly increased compared with the shNC group (P<0.05). Indicating that MTDH may function through the expression of multiple types of apoptosis-related and signaling channel protein in CRC cells.

Knockdown of MTDH downregulates the expression of PCNA

To determine whether the depletion of MTDH can regulate PCNA expression in CRC cells, the protein expression of PCNA in MTDH-deficient HCT116 cells was analyzed. As demonstrated in Fig. 6, the protein expression of PCNA in shMTDH-transfected cells was significantly downregulated compared with that in shNC-transfected cells, suggesting that PCNA may serve as a downstream effector of MTDH to regulate CRC cell growth, apoptosis, cell cycle and migration.

Discussion

The present study investigated the expression and role of MTDH in CRC cells in vitro and revealed that MTDH was highly expressed in CRC cell lines. In addition, shRNA-mediated knockdown of MTDH significantly inhibited CRC cell proliferative, colony-forming and migratory abilities while inducing cell apoptosis and S phase cell cycle arrest. Mechanistically, knockdown of MTDH markedly downregulated the protein expression of p-Akt, c-Myc, Bcl-2 and PCNA, while upregulating the protein of p53 and Bax expression of in HCT116 cells. These results suggested that MTDH-knockdown induces apoptosis of HCT116 cells, and its mechanism may be associated with upregulation of Bax protein expression and downregulation of Bcl-2 protein expression. MTDH is essential for CRC cell growth and migration in vitro possibly through the Akt/c-Myc signaling pathway.

MTDH can be induced by human immunodeficiency virus (HIV)-1 infection or recombinant HIV-1 envelope glycoprotein (gp120) treatment in primary human fetal astrocytes (22). Previous studies have reported that MTDH expression is positively correlated with high risks of cancer in humans (23,24). A recent study demonstrated that the knockdown of MTDH can inhibit ovarian cancer cell growth and invasion while inducing apoptosis and cell cycle arrest at the G0/G1 phase (25). In addition, MTDH was revealed to be highly abundant in HCC cells and tissues, and knockdown of MTDH can suppress HCC cell growth and colony formation in vitro, as well as inhibiting xenografted tumor growth in vivo (26). Furthermore, the present study also demonstrated that MTDH depletion significantly inhibits cell proliferation and colony formation while inducing apoptosis and cell cycle arrest at the S phase in CRC cells. Based on these findings, we hypothesized that MTDH may function as an oncogene and serve a critical role in CRC development and progression.

Metastasis is one of the most critical hallmarks of cancer. The upregulation of MTDH has been demonstrated to promote invasive and metastatic abilities of non-small cell lung cancer cells (23). MTDH overexpression in patients with primary CRC may be considered as a biomarker for lung-specific metastases (27). Consistent with these findings, the present study demonstrated that MTDH depletion can suppress CRC cell migration (Fig. 5), indicating that MTDH is possibly involved in CRC metastasis and may serve as a novel biomarker for CRC metastasis. Targeting MTDH may be a potential therapeutic strategy against CRC.

A previous study has demonstrated that MTDH regulates malignancy development through various cellular signaling cascades, including the MAPK, Ha-ras, NF-κB, PI3K/Akt and Wnt pathways (28). Akt signaling is important for regulating tumor cell proliferation, invasion, apoptosis, cell cycling and survival. Previous studies have demonstrated that the PI3K/Akt signaling pathway is aberrantly activated in human cancer (29,30). Although the overexpression of Akt frequently occurs during CRC carcinogenesis, this is not the case in CRC with microsatellite instability (31). c-Myc is a protooncogene (32) that regulates cell growth and proliferation through regulating a number of downstream target genes, serving an oncogenic role in multiple types of human cancer (33). The upregulation of c-Myc significantly enhances the invasive and metastatic abilities of cancer cells (34). Another previous study demonstrated that MTDH expression can be induced by Ha-ras via the PI3K-Akt signaling pathway (16). MTDH also affects Akt phosphorylation, which is involved in c-Myc-suppressed apoptosis (35). Therefore, as one of the downstream effectors of c-Myc and Ha-ras signaling, MTDH may serve a key role in tumor development and progression (16). The present study consistently indicated that knockdown of MTDH leads to a markedly reduced expression of c-Myc, p-Akt, Bcl-2 and PCNA. MTDH shRNA also upregulated the protein expression of p53 and Bax in HCT116 cells, suggesting that MTDH silencing inhibits CRC cell proliferation and migration. MTDH-knockdown induces apoptosis possibly by upregulation of Bax protein expression and downregulation of Bcl-2 protein expression, and the Akt and c-Myc signal channel may be involved in the changes in protein expression. Akt activation may inhibit its downstream target glycogen synthase kinase-3, which phosphorylates c-Myc at 58 asinine residue, resulting in the inhibition of c-Myc protein degradation (36,37). Therefore, MTDH-activated Akt signaling may cause an upregulation of the protein level of c-Myc, which further promotes CRC development.

In summary, the results of the present study demonstrated that MTDH expression is commonly expressed in CRC cell lines and shRNA-mediated knockdown of MTDH inhibits HCT116 cell proliferation, colony formation and migration while inducing cell cycle arrest at the S phase and apoptosis, which is not only associated with the downregulation of p-Akt, c-Myc, Bcl-2 and PCNA expression, but also associated with the upregulation of p53 and Bax protein. Further studies are required to elucidate the mechanisms underlying the regulatory activity of MTDH in CRC. Therefore, targeting MTDH may be a potential therapeutic strategy in CRC treatment. Further in vivo investigation is required to develop MTDH inhibitors for CRC therapy.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Foundation of Haikou Science and Technology Bureau, Haikou, Hainan, China (grant no, 2015-035).

Availability of data and materials

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

Authors' contributions

JWL and CZH performed the research, the data acquisition and drafted the manuscript; JHL, JHY, YPL, WFZ, MXZ, YL and QHC provided assistance for data acquisition, data analysis and statistical analysis; LGL designed the research, edited and reviewed the manuscript. ZSX provided assistance on the data acquisition, data analysis and statistical analysis. All authors have read and approved the content of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

All authors declare that they have no competing interests.

References

1 

Theodoratou E, Farrington SM, Tenesa A, McNeill G, Cetnarskyj R, Korakakis E, Din FV, Porteous ME, Dunlop MG and Campbell H: Associations between dietary and lifestyle risk factors and colorectal cancer in the Scottish population. Eur J Cancer Prev. 23:8–17. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Akagi Y, Kinugasa T, Adachi Y and Shirouzu K: Prognostic significance of isolated tumor cells in patients with colorectal cancer in recent 10-year studies. Mol Clin Oncol. 1:582–592. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Guo Y, Xu F, Lu T, Duan Z and Zhang Z: Interleukin-6 signaling pathway in targeted therapy for cancer. Cancer Treat Rev. 38:904–910. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Lièvre A, Blons H and Laurent-Puig P: Oncogenic mutations as predictive factors in colorectal cancer. Oncogene. 29:3033–3043. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Grady WM and Pritchard CC: Molecular alterations and biomarkers in colorectal cancer. Toxicol Pathol. 42:124–139. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Kang DC, Su ZZ, Sarkar D, Emdad L, Volsky DJ and Fisher PB: Cloning and characterization of HIV-1-inducible astrocyte elevated gene-1, AEG-1. Gene. 353:8–15. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Hu G, Wei Y and Kang Y: The multifaceted role of MTDH/AEG-1 in cancer progression. Clin Cancer Res. 15:5615–5620. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Li M, Dai Y, Wang L and Li L: Astrocyte elevated gene-1 promotes the proliferation and invasion of breast cancer cells by activating the Wnt/β-catenin signaling pathway. Oncol Lett. 13:2385–2390. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Yao Y, Gu X, Liu H, Wu G, Yuan D, Yang X and Song Y: Metadherin regulates proliferation and metastasis via actin cytoskeletal remodelling in non-small cell lung cancer. Br J Cancer. 111:355–364. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Zhu HD, Liao JZ, He XX and Li PY: The emerging role of astrocyte elevated gene in hepatocellular carcinoma (Review). Oncol Rep. 34:539–546. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Jung HI, Ahn T, Bae SH, Chung JC, Kim H, Chin S, Jeong D, Cho HD, Lee MS, Kim HC, et al: Astrocyte elevated gene-1 overexpression in hepatocellular carcinoma: An independent prognostic factor. Ann Surg Treat Res. 88:77–85. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Jian-bo X, Hui W, Yu-long H, Chang-hua Z, Long-juan Z, Shirong C and Wen-hua Z: Astrocyte-elevated gene-1 overexpression is associated with poor prognosis in gastric cancer. Med Oncol. 28:455–462. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Tokunaga E, Nakashima Y, Yamashita N, Hisamatsu Y, Okada S, Akiyoshi S, Aishima S, Kitao H, Morita M and Maehara Y: Overexpression of metadherin/MTDH is associated with an aggressive phenotype and a poor prognosis in invasive breast cancer. Breast Cancer. 21:341–349. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Gnosa S, Shen YM, Wang CJ, Zhang H, Stratmann J, Arbman G and Sun XF: Expression of AEG-1 mRNA and protein in colorectal cancer patients and colon cancer cell lines. J Transl Med. 10:1092012. View Article : Google Scholar : PubMed/NCBI

15 

Emdad L, Sarkar D, Su ZZ, Randolph A, Boukerche H, Valerie K and Fisher PB: Activation of the nuclear factor kappaB pathway by astrocyte elevated gene-1: Implications for tumor progression and metastasis. Cancer Res. 66:1509–1516. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Lee SG, Su ZZ, Emdad L, Sarkar D and Fisher PB: Astrocyte elevated gene-1 (AEG-1) is a target gene of oncogenic Ha-ras requiring phosphatidylinositol 3-kinase and c-Myc. Proc Natl Acad Sci USA. 103:17390–17395. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Wei J, Li Z, Chen W, Ma C, Zhan F, Wu W and Peng Y: AEG-1 participates in TGF-beta1-induced EMT through p38 MAPK activation. Cell Biol Int. 37:1016–1021. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Li PP, Feng LL, Chen N, Ge XL, Lv X, Lu K, Ding M, Yuan D and Wang X: Metadherin contributes to the pathogenesis of chronic lymphocytic leukemia partially through Wnt/β-catenin pathway. Med Oncol. 32:212015. View Article : Google Scholar

19 

Song H, Li C, Li R and Geng J: Prognostic significance of AEG-1 expression in colorectal carcinoma. Int J Colorectal Dis. 25:1201–1209. 2010. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

Wang F, Wang H, Sun X and Li M: Apoptosis-induction is a novel therapeutic strategy for gastrointestinal and liver cancers. Curr Gene Ther. 15:193–200. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Su ZZ, Kang DC, Chen Y, Pekarskaya O, Chao W, Volsky DJ and Fisher PB: Identification and cloning of human astrocyte genes displaying elevated expression after infection with HIV-1 or exposure to HIV-1 envelope glycoprotein by rapid subtraction hybridization, RaSH. Oncogene. 21:3592–3602. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Sun S, Ke Z, Wang F, Li S, Chen W, Han A, Wang Z, Shi H, Wang LT and Chen X: Overexpression of astrocyte-elevated gene-1 is closely correlated with poor prognosis in human non-small cell lung cancer and mediates its metastasis through up-regulation of matrix metalloproteinase-9 expression. Hum Pathol. 43:1051–1060. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Yu JQ, Zhou Q, Zhu H, Zheng FY and Chen ZW: Overexpression of astrocyte elevated gene-1 (AEG-1) in cervical cancer and its correlation with angiogenesis. Asian Pac J Cancer Prev. 16:2277–2281. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Wang J, Chen X and Tong M: Knockdown of astrocyte elevated gene-1 inhibited cell growth and induced apoptosis and suppressed invasion in ovarian cancer cells. Gene. 616:8–15. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Li WF, Ou Q, Dai H and Liu CA: Lentiviral-mediated short hairpin RNA knockdown of MTDH inhibits cell growth and induces apoptosis by regulating the PTEN/AKT pathway in hepatocellular carcinoma. Int J Mol Sci. 6:19419–19432. 2015. View Article : Google Scholar

27 

Casimiro S, Fernandes A, Oliveira AG, Franco M, Pires R, Peres M, Matias M, Tato-Costa J, Guerra N, Ramos M, et al: Metadherin expression and lung relapse in patients with colorectal carcinoma. Clin Exp Metastasis. 31:689–696. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Shi X and Wang X: The role of MTDH/AEG-1 in the progression of cancer. Int J Clin Exp Med. 8:4795–4807. 2015.PubMed/NCBI

29 

Millis SZ, Ikeda S, Reddy S, Gatalica Z and Kurzrock R: Landscape of phosphatedy- linositol-3-kinase pathway alterations across 19 784 diverse solid tumors. JAMA Oncol. 2:1565–1573. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Brown JS and Banerji U: Maximising the potential of AKT inhibitors as anti-cancer treatments. Pharmacol Ther. 172:101–105. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Roy HK, Olusola BF, Clemens DL, Karolski WJ, Ratashak A, Lynch HT and Smyrk TC: AKT proto-oncogene over-expression is an early event during sporadic colon carcinogenesis. Carcinogenesis. 23:201–205. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Nesbit CE, Tersak JM and Prochownik EV: MYC oncogenes and human neoplastic disease. Oncogene. 18:3004–3016. 1999. View Article : Google Scholar : PubMed/NCBI

33 

Kalkat M, De Melo J, Hickman KA, Lourenco C, Redel C, Resetca D, Tamachi A, Tu WB and Penn LZ: MYC deregulation in primary human cancers. Genes. 8:E1512017. View Article : Google Scholar : PubMed/NCBI

34 

Liu Z, He Q, Ding X, Zhao T, Zhao L and Wang A: SOD2 is a C-myc target gene that promotes the migration and invasion of tongue squamous cell carcinoma involving cancer stem-like cells. Int J Biochem Cell Biol. 60:139–146. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Lee SG, Su ZZ, Emdad L, Sarkar D, Franke TF and Fisher PB: Astrocyte elevated gene-1 activates cell survival pathways through PI3K-Akt signaling. Oncogene. 27:1114–1121. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Gregory MA, Qi Y and Hann SR: Phosphorylation by glycogen synthase kinase-3 controls c-Myc proteolysis and subnuclear localization. J Biol Chem. 278:51606–51612. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Lepique AP, Moraes MS, Rocha KM, Eichler CB, Hajj GN, Schwindt TT and Armelin HA: c-Myc protein is stabilized by fibroblast growth factor 2 and destabilized by ACTH to control cell cycle in mouse Y1 adrenocortical cells. J Mol Endocrinol. 33:623–638. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2018
Volume 40 Issue 4

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
Li JW, Huang CZ, Li JH, Yuan JH, Chen QH, Zhang WF, Xu ZS, Liu YP, Li Y, Zhan MX, Zhan MX, et al: Knockdown of metadherin inhibits cell proliferation and migration in colorectal cancer. Oncol Rep 40: 2215-2223, 2018
APA
Li, J., Huang, C., Li, J., Yuan, J., Chen, Q., Zhang, W. ... Lu, L. (2018). Knockdown of metadherin inhibits cell proliferation and migration in colorectal cancer. Oncology Reports, 40, 2215-2223. https://doi.org/10.3892/or.2018.6581
MLA
Li, J., Huang, C., Li, J., Yuan, J., Chen, Q., Zhang, W., Xu, Z., Liu, Y., Li, Y., Zhan, M., Lu, L."Knockdown of metadherin inhibits cell proliferation and migration in colorectal cancer". Oncology Reports 40.4 (2018): 2215-2223.
Chicago
Li, J., Huang, C., Li, J., Yuan, J., Chen, Q., Zhang, W., Xu, Z., Liu, Y., Li, Y., Zhan, M., Lu, L."Knockdown of metadherin inhibits cell proliferation and migration in colorectal cancer". Oncology Reports 40, no. 4 (2018): 2215-2223. https://doi.org/10.3892/or.2018.6581