MicroRNA‑466 inhibits the aggressive behaviors of hepatocellular carcinoma by directly targeting metadherin

Retraction in: /10.3892/or.2022.8357

  • Authors:
    • Chen Jia
    • Desheng Tang
    • Chen Sun
    • Lei Yao
    • Fujun Li
    • Yanhua Hu
    • Xinchen Zhang
    • Dequan Wu
  • View Affiliations

  • Published online on: October 3, 2018     https://doi.org/10.3892/or.2018.6763
  • Pages: 3890-3898
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Numerous microRNAs (miRNAs) have been demonstrated to be downregulated or upregulated in hepatocellular carcinoma (HCC) and play important roles in its occurrence and development. Therefore, the investigation of miRNAs and their functions implicated in the genesis and development of HCC may provide key clues for the identification of effective therapeutic approaches for patients with this disease. The aims of the present study were to detect miRNA‑466 (miR‑466) expression in HCC tissues and cell lines and to determine its effects on HCC cell proliferation, apoptosis and metastasis, as well as to explore the mechanisms underlying the tumor‑suppressing roles of miR‑466 in HCC. In the present study, reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) was performed to detect miR‑466 expression in HCC tissues and cell lines. The effects of miR‑466 upregulation on HCC cell proliferation, apoptosis, migration and invasion were determined using Cell Counting Kit‑8 assay, flow cytometry analysis and Transwell chamber assay, respectively. The potential target gene of miR‑466 was predicted using bioinformatic analysis, which was further confirmed by luciferase reporter assay, RT‑qPCR and western blot analysis. It was found that miR‑466 was obviously decreased in HCC tissues and cell lines. The results of functional experiments revealed that restoration of miR‑466 expression suppressed the proliferation, induced apoptosis, and reduced the metastasis of HCC cells. In addition, metadherin (MTDH) was identified as a direct target of miR‑466 in HCC cells. Furthermore, MTDH was upregulated in HCC tissues, which was inversely correlated with the miR‑466 level. Moreover, inhibition of MTDH displayed similar tumor‑suppressing roles as miR‑466 upregulation in HCC cells. In addition, MTDH reintroduction restored the tumor‑suppressor activity of miR‑466 overexpression in HCC cells. These findings suggest that miR‑466 is a potential therapeutic tool for HCC therapy.

Introduction

Hepatocellular carcinoma (HCC) ranks as the 5th most commonly diagnosed human malignancy and the 3rd leading cause of cancer-related mortalities globally (1). Recently, the morbidity and mortality from HCC are gradually increasing, particularly in China (2). It is estimated that there will be approximately 854,000 cases and 810,000 deaths yearly due to HCC worldwide (3). Owing to the asymptomatic nature of this disease, the majority of HCC patients are diagnosed late, at a time when curative treatments are infeasible (4). Despite the great development in the fight against HCC, the long-term survival of patients with HCC remains unsatisfactory with a 5-year survival rate of less than 30% (5). Multiple factors, such as hepatitis B or C viral infection, exposure to aflatoxin, and excessive drinking, have been implicated in the pathogenesis of HCC (6); however, the detailed mechanism remains unknown and warrants further study. In this regard, disclosing the molecular mechanisms underlying the formation and progression of HCC is desperately needed for identifying novel therapeutic strategies for patients with this aggressive malignant tumor.

MicroRNAs (miRNAs) are a group of highly conserved, single stranded, and short RNA molecules, playing significant roles in the regulation of gene expression (7). miRNAs regulate the expression of genes by complete or incomplete base-pairing with the 3′-untranslated regions (3′-UTRs) of their target genes, leading to translational suppression and/or mRNA degradation (8). To date, more than 2,000 miRNAs have been verified in the human genome which may modulate the expression of approximately 30% of all the protein-coding genes (9). The deregulation of miRNA expression has been validated in almost all human cancer types and was demonstrated to be closely related with carcinogenesis and cancer progression (1012). Numerous miRNAs are downregulated or upregulated in HCC, such as miR-21 (13), miR-493 (14), miR-873 (15) and miR-3662 (16). Upregulated miRNAs usually serve as oncogenes (17,18), whereas lowly expressed miRNAs display tumor-suppressor activity (19,20) in regards to HCC initiation and development. Therefore, inhibition or restoration of a specific miRNA might be an effective therapeutic technique for HCC treatment.

A variety of miRNAs have been identified to be deregulated in HCC; however, their functions in HCC formation and progression and the underlying mechanisms by which this occurs remain largely unknown. miR-466 was previously reported to be involved in the development of colorectal (21), prostate (22) and cervical (23) cancers. However, the expression level of miR-466 in HCC and its actual functions in the development of HCC, as well as the associated mechanisms, have remained elusive. Therefore, this study aimed to detect miR-466 expression in HCC tissues and cell lines and to determine its effects on HCC cell proliferation, apoptosis and metastasis, as well as to explore the mechanisms underlying the tumor-suppressive roles of miR-466 in HCC. The findings of the present study may provide further insight into the general mechanisms underlying HCC genesis and development and may promote the development of attractive therapeutic approaches for HCC.

Materials and methods

Tissue samples

This study was approved by the Ethics Committee of The Second Affiliated Hospital of Harbin Medical University and written informed consent was provided by all patients enrolled in the research. A total of 32 pairs of HCC tissues and pair-matched adjacent normal tissues were collected from The Second Affiliated Hospital of Harbin Medical University. The clinicopathological features of the 32 patients [mean age, 57; (range, 45–70 years); 12 female and 20 male patients)] recruited to the study are shown in Table I. The collection dates of these tissue specimens were from July 2015 to May 2017. None of the patients had received chemotherapy or radiotherapy before surgical resection. All tissues were stored in liquid nitrogen until further RNA isolation.

Table I.

Clinicopathological features of the HCC patients recruited to the study.

Table I.

Clinicopathological features of the HCC patients recruited to the study.

Patient no.SexAge (years)TNM stagePatient no.SexAge (years)TNM stage
1M63T1N0M017M61T3aN1M0
2F69T1N0M018M47T1N0M0
3F54T1N0M019M55T1N0M0
4M57T1N0M020M57T1N0M0
5M62T2N0M021M52T1N0M0
6F48T1N0M022M65T2N0M0
7F69T2N1M023F68T3aN1M0
8M55T1N0M024M50T2N1M0
9M58T3aN1M025M64T1N0M0
10F62T1N0M026F48T1N0M0
11M50T1N0M027M46T1N0M0
12F51T2N1M028M56T2N0M0
13M64T1N0M029M66T1N0M0
14F70T1N0M030M58T1N0M0
15F49T2N0M031F50T1N0M0
16M45T1N0M032F69T2N1M0

[i] HCC, hepatocellular carcinoma; TNM, Tumor, Node, Metastasis; M, male; F, female.

Cell culture

Two human HCC cell lines (Huh7 and Hep3B) and an immortalized normal human liver epithelial cell line (L-O2) were obtained from the Cell Bank of the Chinese Academy of Biological Science (Shanghai, China). Dulbecco's modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS), 100 U/ml of penicillin and 100 µg/ml of streptomycin (all from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) was used to culture all cell lines mentioned above. All cells were maintained at 37°C in a humidified atmosphere containing 5% CO2.

Cell transfection

miR-466 mimics and negative control miRNA mimics (miR-NC) were chemically produced by RiboBio Co., Ltd. (Guangzhou, China). Specific small interfering RNA (siRNA) targeting the MTDH gene (si-MTDH) and the negative control siRNA (si-NC) were constructed by Guangzhou GeneCopoeia Co., Ltd. (Guangzhou, China). MTDH overexpression plasmid (pcDNA3.1-MTDH) lacking the 3′-UTR and negative control plasmid (pcDNA3.1) were purchased from Genepharma Co., Ltd. (Shanghai, China). Cells were placed into 6-well plates, grown to approximately 70% confluence, and transfected with miRNA mimics, siRNA or plasmid using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). Transfected cells were incubated at 37°C under 5% CO2. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was applied to determine the transfection efficiency of the miR-466 mimics 48 h after transfection. At 72-h post-transfection, western blot analysis was performed to detect the efficiency of si-MTDH and pcDNA3.1-MTDH plasmid transfection.

RT-qPCR

TRIzol® reagent (Thermo Fisher Scientific, Inc.) was used to extract total RNA from the tissue specimens and cultured cells. The concentration of total RNA was detected using a Nanodrop 1000 spectrophotometer (Thermo Fisher Scientific, Inc.). To quantify miR-466 expression, reverse transcription was performed using a TaqMan® MicroRNA Reverse Transcription Kit (Applied Biosystems; Thermo Fisher Scientific, Inc.). Subsequently, the synthesized complementary DNA (cDNA) was subjected to quantitative polymerase chain reaction (qPCR) using a TaqMan MicroRNA Assay kit (Applied Biosystems; Thermo Fisher Scientific, Inc.). The cycling conditions for qPCR were as follows: 50°C for 2 min, 95°C for 10 min; 40 cycles of denaturation at 95°C for 15 sec; and annealing/extension at 60°C for 60 sec.

To analyze the MTDH mRNA level, cDNA was produced from total RNA using a PrimeScript RT Reagent kit (Takara Biotechnology, Co., Ltd., Dalian, China). Afterwards, qPCR was carried out using a SYBR Premix Ex Taq™ kit (Takara Biotechnology, Co., Ltd.). The cycling conditions for qPCR were as follows: 5 min at 95°C, followed by 40 cycles of 95°C for 30 sec and 65°C for 45 sec. U6 small nuclear RNA and GAPDH served as internal references for miR-466 and MTDH mRNA, respectively. The primers were designed as follows: miR-466, 5′-ATGGTTCGTGGGATACACATACACGCA-3′ (forward) and 5′-GCAGGGTCCGAGGTATTC-3′ (reverse); U6, 5′-GCTTCGGCAGCACATATACTAAAAT-3′ (forward) and 5′-CGCTTCACGAATTTGCGTGTCAT-3′ (reverse); MTDH, 5′-TGCTCTCTCACAGACAA-3′ (forward) and 5′-TCGCTCTGCAGATGAGATAG-3′ (reverse); and GAPDH, 5′-CGGAGTCAACGGATTTGGTCGTAT-3′ (forward) and 5′-AGCCTTCTCCATGGTGGTGAAGAC-3′ (reverse). The 2−ΔΔCq method (24) was used to calculate gene expression.

Cell counting Kit-8 (CCK-8) assay

Cell proliferation was assessed using the CCK-8 assay in accordance with the supplied protocol and instructions. In detail, transfected cells were harvested and inoculated into 96-well plates at a density of 3×103 cells/well. Cells were maintained at 37°C in a humidified atmosphere containing 5% CO2. A total of 10 µl of CCK-8 solution (Dojindo Molecular Technologies, Inc., Kumamoto, Japan) was added into every well at four time points (0, 24, 48 and 72 h after inoculation). After incubation at 37°C for another 2 h, the optical density (OD) values were measured using a microplate reader (Bio-Rad Laboratories, Hercules, CA, USA) at a wavelength of 490 nm.

Flow cytometry analysis

Cells were collected at 48 h after being transfected, and the apoptosis rate was determined using an Annexin V fluorescein isothiocyanate (FITC) Apoptosis Detection kit (Biolegend, San Diego, CA, USA) according to the manufacturer's instruction. Briefly, transfected cells were washed twice with cold phosphate-buffered saline, suspended in 100 µl of binding buffer, and incubated with 5 µl Annexin V-FITC and 5 µl propidium iodide (PI). The cells were then incubated in the dark at room temperature for 30 min. The rate of apoptosis was assessed by FACScan flow cytometry (BD Biosciences, Franklin Lakes, NJ, USA).

Transwell chamber assay

The invasive and migratory properties of HCC cells were examined using 24-well Transwell plate cell culture inserts (Corning, New York, NY, USA) covered with or without Matrigel (BD Biosciences, San Jose, CA, USA), respectively. After 48 h of transfection, cells were harvested and suspended with FBS-free DMEM. A total of 5×104 transfected cells were plated in the upper compartments, and the lower compartments were maintained with 500 µl of DMEM that was supplemented with 10% FBS. The transfected cells were then cultured for 24 h at 37°C with 5% CO2. Cells remaining on the upper surface of the membranes were gently removed by swabbing the top layer. The migrated or invaded cells, which were attached to the lower surface of the membranes, were fixed with 100% methanol and stained with 0.05% crystal violet. The number of migrated or invaded cells was counted under an inverted microscope (×200 magnification; Olympus Corp., Tokyo, Japan). Five randomly selected fields were analyzed for each insert.

Bioinformatic prediction and luciferase reporter assay

microRNA.org (www.microrna.org/microrna/) and TargetScan (www.targetscan.org) were adopted to search for the putative targets of miR-466. MTDH was predicted as a candidate for miR-466. To verify this, luciferase reporter plasmids, including psi-CHECK-MTDH-3′-UTR wild-type (Wt) and psi-CHECK-MTDH-3′-UTR mutant (Mut), were constructed by GenePharma Co., Ltd., and were cotransfected with miR-466 mimics or miR-NC into the cells using Lipofectamine 2000. The cotransfection was performed according to the manufacturer's instructions. After approximately 48 h of post-transfection, luciferase activities were measured using the Dual-Luciferase Reporter Assay system (Promega Corps, Madison, WI, USA) following the manufacturer's protocol. The firefly luciferase activities were normalized to the Renilla luciferase activities.

Western blot analysis

Tissue specimens and cells were lysed with radioimmunoprecipitation assay buffer (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) containing 0.1 mg/ml phenylmethylsulfonyl fluoride, 1 mM sodium orthovanadate and 1 mg/ml aprotinin. A BCA Protein Assay kit (Beyotime Institute of Biotechnology, Shanghai, China) was then utilized to determine the concentration of total protein. An equal amount of protein (30 µg) was separated on a 10% sodium dodecyl sulfate-polyacrylamide gel and transferred into polyvinylidene difluoride membranes (Bio-Rad Laboratories). After that, the membranes were blocked in Tris-buffered saline-Tween (TBST) containing 5% non-fat milk, followed by incubation with the primary antibodies at 4°C overnight. Subsequent to washes with TBST thrice, the membranes were probed with appropriate horseradish peroxidase-conjugated secondary antibodies (1:5,000 dilution; cat. no. sc-516102; Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA). The protein blots were detected by BM Chemiluminescence Western blotting kit (Sigma-Aldrich; Merck KGaA). The primary antibodies used in this study included mouse anti-human monoclonal MTDH (1:1,000 dilution; cat. no. sc-517220; Santa Cruz Biotechnology, Inc.) and mouse anti-human monoclonal GAPDH (1:1,000 dilution; cat. no. sc-32233; Santa Cruz Biotechnology, Inc.). GAPDH was used as the internal reference. Protein expression was quantified using Quantity One software version 4.62 (Bio-Rad Laboratories, Inc).

Statistical analysis

All data are shown as the mean ± standard deviation and were subjected to SPSS software version 18 (SPSS, Inc., Chicago, IL, USA) for statistical analysis. Differences between groups were analyzed using Student's t-test, paired Student's t-test or one-way ANOVA, followed by the Student-Newman-Keuls (SNK) multiple comparison test. Spearman's correlation analysis was performed between miR-466 and the mRNA levels of MTDH in HCC tissues. Differences were defined as statistically significant if P<0.05.

Results

miR-466 expression is lower in HCC tissues and cell lines

miR-466 has been reported to be aberrantly expressed in several types of human malignancies (2123). However, the expression pattern of miR-466 in HCC remains unknown. First, we detected miR-466 expression in 32 pairs of HCC tissues and pair-matched adjacent normal tissues. The data of RT-qPCR showed that miR-466 was lowly expressed in HCC tissues in contrast to that noted in the pair-matched adjacent normal tissues (Fig. 1A, P<0.05). In addition, RT-qPCR was performed to assess the miR-466 expression in HCC cell lines. Compared with the miR-466 expression in an immortalized normal human liver epithelial cell line (L-O2), miR-466 was downregulated in the two HCC cell lines, including Huh7 and Hep3B (Fig. 1B, P<0.05).

miR-466 overexpression inhibits proliferation, facilitates apoptosis and decreases the metastasis potential of Huh7 and Hep3B cells

Given the downregulation of miR-466 in HCC, we hypothesized that miR-466 may play tumor suppressive roles in the progression of HCC. To confirm this, we transfected miR-466 mimics or miR-NC into Huh7 and Hep3B cells which possessed much lower miR-466 levels among the two HCC cell lines (data not shown). The expression of miR-466 was successfully overexpressed in Huh7 and Hep3B cells by the transfection of miR-466 mimics (Fig. 2A, P<0.05). CCK-8 assay was conducted to assess the effect of miR-466 restoration on cell proliferation in HCC. The results revealed that miR-466 upregulation significantly suppressed the proliferation of Huh7 and Hep3B cells (Fig. 2B, P<0.05). Alterations in cell proliferation are mostly related with the change in cell apoptosis; therefore, flow cytometric analysis was performed to detect the apoptosis rate of Huh7 and Hep3B cells that were transfected with the miR-466 mimics or miR-NC. It was demonstrated that the apoptosis rate of Huh7 and Hep3B cells was higher in the miR-466 overexpression group than that in the miR-NC group (Fig. 2C, P<0.05). We then examined the possible regulatory role of miR-466 in the migratory and invasive abilities of HCC cells. Transwell chamber assay indicated that ectopic miR-466 expression led to a significant suppression of Huh7 and Hep3B cell migration (Fig. 2D, P<0.05) and invasion (Fig. 2E, P<0.05). These results suggest that miR-466 may play a suppressive role in the proliferation and metastatic activity of HCC cells.

MTDH is a direct target gene of miR-466 in HCC cells

To identify the mechanisms underlying how miR-466 exerts its tumor-suppressor activity in HCC cells, we used bioinformatic analysis to search for the putative targets of miR-466. Complementary sequences were observed between miR-466 and the 3′-UTR of MTDH (Fig. 3A). MTDH was previously reported to be implicated in the oncogenesis and development of HCC (2532) and was therefore selected for further identification. Luciferase reporter assay was applied to confirm whether MTDH is a bona fide target gene of miR-466 in HCC cells. Fig. 3B illustrates that the restoration of miR-466 expression was able to suppress the luciferase activity of the wild-type (Wt) construct of MTDH 3′-UTR (P<0.05), while modulating the miR-466 level exhibited unaltered luciferase activity of mutant (Mut) construct of MTDH 3′-UTR in Huh7 and Hep3B cells. Next, we detected the MTDH expression in HCC tissues and explored its possible relationship with miR-466. RT-qPCR analysis indicated that the mRNA level of MTDH was higher in HCC tissues than that in pair-matched adjacent normal tissues (Fig. 3C, P<0.05). Meanwhile, an inverse correlation between miR-466 and MTDH mRNA expression was validated in HCC tissues using Spearman's correlation analysis (Fig. 3D; r=−0.6714, P=0.0002). Furthermore, results of RT-qPCR and western blot analysis revealed that miR-466 re-expression decreased the mRNA (Fig. 3E, P<0.05) and protein (Fig. 3F, P<0.05) levels of MTDH in Huh7 and Hep3B cells. Collectively, our data suggest that MTDH is a direct target of miR-466 in HCC cells.

Inhibition of MTDH displays similar tumor-suppressing roles as the miR-466 upregulation in Huh7 and Hep3B cells

MTDH was identified as a direct target gene of miR-466 in HCC cells; hence, our subsequent analyses were focused on the functional roles of MTDH in HCC cells. To this end, siRNA against the expression of MTDH (si-MTDH) was employed to knock down endogenous MTDH expression in Huh7 and Hep3B cells. MTDH protein expression was obviously downregulated by si-MTDH transfection relative to its expression in the si-NC group (Fig. 4A, P<0.05). Analysis of cell proliferation and apoptosis using CCK-8 assay and flow cytometric analysis, respectively, showed that MTDH knockdown restricted the cell proliferation (Fig. 4B, P<0.05) and promoted the apoptosis (Fig. 4C, P<0.05) of Huh7 and Hep3B cells compared with these parameters in the si-NC group. Transwell chamber assays revealed that transfection of si-MTDH in Huh7 and Hep3B cells caused significant suppression of migration (Fig. 4D, P<0.05) and invasiveness (Fig. 4E, P<0.05) compared with these abilities in the cells transfected with si-NC. Taken together, these observations demonstrated that inhibition of MTDH exhibits similar tumor suppressive roles as miR-466 upregulation in HCC cells, suggesting that MTDH is a downstream target of miR-466 in HCC.

Overexpression of MTDH reverses the tumor-suppressing roles of miR-466 in Huh7 and Hep3B cells

To further determine whether MTDH downregulation is essential for the tumor-suppressing roles of miR-466 in HCC, rescue experiments were performed in Huh7 and Hep3B cells by co-transfection with miR-466 mimics and empty pcDNA3.1 or pcDNA3.1-MTDH plasmid lacking the 3′-UTR. The downregulation of MTDH protein induced by miR-466 overexpression was restored in Huh7 and Hep3B cells after co-transfection with pcDNA3.1-MTDH (Fig. 5A, P<0.05). MTDH overexpression blunted the tumor-suppressor activity of miR-466 in regards to Huh7 and Hep3B cell proliferation (Fig. 5B, P<0.05), apoptosis (Fig. 5C, P<0.05), migration (Fig. 5D, P<0.05) and invasion (Fig. 5E, P<0.05) in vitro. In summary, these results suggest that the antitumor effects of miR-466 overexpression in HCC cells were, at least partly, mediated by inhibition of MTDH.

Discussion

Accumulating evidence has demonstrated that numerous miRNAs are either downregulated or upregulated in HCC and play important roles in its occurrence and development (3335). Notably, in-depth understanding of miRNAs could expand the current knowledge regarding the mechanisms underlying hepatocarcinogenesis (36). Therefore, investigation of miRNAs and their functions implicated in the genesis and development of HCC would provide key clues for the identification of effective therapeutic approaches for patients with this disease. In the present study, we found that miR-466 is downregulated in HCC tissues and cell lines. Ectopic miR-466 expression impeded the proliferation, induced apoptosis, and restricted the metastasis ability of HCC cells. Mechanistically, this study confirmed that MTDH is a direct target gene of miR-466 in HCC cells. These observations suggest the that miR-466/MTDH axis may potentially serve as attractive and effective therapeutic targets in HCC.

miR-466 was previously reported to be lowly expressed in colorectal (21) and prostate (22) cancers. Low miR-466 expression was significantly associated with tumor size, TNM stage, lymph node metastasis, and distant metastasis of colorectal cancer patients. In addition, colorectal cancer patients with a reduced miR-466 level had a reduced overall survival period than patients with a high miR-466 level. Furthermore, multivariate analysis validated miR-466 expression as a prognostic marker for colorectal cancer patients (21). Functional experiments revealed that miR-466 serves as a tumor suppressor in colorectal (21) and prostate (22) cancers by regulating diverse biological behaviors. Nevertheless, miR-466 was found to be overexpressed in cervical cancer tissues and cell lines. High miR-466 expression was correlated with lymph node metastasis of cervical cancer patients (23). These conflicting studies suggest that miR-466 displays tissue specificity in regards to its expression status and biological roles in human malignancies. Hence, miR-466 may represent a valuable target for the diagnosis and therapy of patients with these specific cancer types.

The mechanisms underlying the tumor-suppressive effects of miR-466 in HCC cells were explored in the present study. First, bioinformatic analysis predicted that MTDH is a putative target of miR-466. Second, luciferase reporter assays revealed that miR-466 could recognize and directly bind to the 3′-UTR of MTDH in HCC cells. Third, MTDH expression was upregulated in HCC tissues, and upregulation of MTDH was inversely related with the miR-466 level. Fourth, miR-466 restoration decreased MTDH mRNA and protein levels in HCC cells. Finally, inhibition of MTDH was able to simulate the effects of miR-466 overexpression in HCC cells. MTDH reintroduction blunted the tumor-suppressor activity of miR-466 in malignant phenotypes of ccRCC cells. These results provide sufficient evidence to validate that MTDH is a direct target gene of miR-466 in HCC cells.

MTDH, located on chromosome 8q22, was first discovered in human foetal astrocytes in 2002 (37). MTDH was reported to be highly expressed in several types of human cancer, such as breast (38), colorectal (39), thyroid (40) and bladder (41) cancers. MTDH is a multifunctional oncogene implicated in the regulation of various processes (4244). MTDH was found to be upregulated in HCC, and its high expression exhibited a significant association with age, Edmondson grade, microvascular invasion, TNM stage, histological differentiation, American Joint Committee on Cancer T stage, and α-fetoprotein level (2528). HCC patients with high MTDH expression had a shorter survival period compared with patients with low MTDH expression (27). In addition, multivariate analysis identified high MTDH expression as an independent predictor of shorter disease-free survival (28). Dysregulation of MTDH was found to be closely related to the aggressive behaviors of HCC by regulating several pathological processes, including cell growth, viability, the cell cycle, cell survival, apoptosis, transformation, metastasis, epithelial-to-mesenchymal transition, chemoresistance, angiogenesis and senescence (26,2932). In the present study, we demonstrated that miR-466 targeted MTDH to inhibit the progression of HCC. Thus, MTDH inhibition using miR-466-based targeted therapy could be a potential therapeutic technique for elimination of HCC in the future.

The present study, to the best of our knowledge, presents initial evidence that miR-466 expression is downregulated in both HCC tissues and cell lines. miR-466 restoration impeded the development of HCC by directly targeting MTDH. Understanding the expression pattern and biological roles of miR-466 in hepatocarcinogenesis and progression would facilitate the development of miR-466 as a potential therapeutic tool for HCC patients. However, in this study, we did not show the localization of miR-466 and MTDH in clinical pathological tissues. In addition, the association between miR-466 and clinicopathological features as well as the prognosis of HCC patients was not examined. These are limitations of the present study which will be resolved in further experiments.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

DW and CJ designed the present research. CJ, DT and CS performed RT-qPCR, cell transfection and CCK-8 assay. LY, FL and YH conducted the flow cytometric analysis, the Transwell chamber assay and the luciferase reporter assay. Western blot analysis and statistical analysis was carried out by XZ. All authors participated in the writing and read and approved the final draft. All authors agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of The Second Affiliated Hospital of Harbin Medical University, and was performed in accordance with the Declaration of Helsinki and the guidelines of the Ethics Committee of The Second Affiliated Hospital of Harbin Medical University. Written informed consent was obtained from all patients for the use of their clinical tissues.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Global Burden of Disease Cancer Collaboration, . Fitzmaurice C, Allen C, Barber RM, Barregard L, Bhutta ZA, Brenner H, Dicker DJ, Chimed-Orchir O, Dandona R, et al: Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 32 cancer groups, 1990 to 2015: A systematic analysis for the global burden of disease study. JAMA Oncol. 3:524–548. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Galun D, Basaric D, Zuvela M, Bulajic P, Bogdanovic A, Bidzic N and Milicevic M: Hepatocellular carcinoma: From clinical practice to evidence-based treatment protocols. World J Hepatol. 7:2274–2291. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Ercolani G, Grazi GL, Ravaioli M, Del Gaudio M, Gardini A, Cescon M, Varotti G, Cetta F and Cavallari A: Liver resection for hepatocellular carcinoma on cirrhosis: Univariate and multivariate analysis of risk factors for intrahepatic recurrence. Ann Surg. 237:536–543. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Nguyen VT, Law MG and Dore GJ: Hepatitis B-related hepatocellular carcinoma: Epidemiological characteristics and disease burden. J Viral Hepat. 16:453–463. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Yates LA, Norbury CJ and Gilbert RJ: The long and short of microRNA. Cell. 153:516–519. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Hammond SM: An overview of microRNAs. Adv Drug Deliv Rev. 87:3–14. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Xie B, Ding Q, Han H and Wu D: miRCancer: A microRNA-cancer association database constructed by text mining on literature. Bioinformatics. 29:638–644. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Bishop KS, Xu H and Marlow G: Epigenetic regulation of gene expression induced by butyrate in colorectal cancer: Involvement of MicroRNA. Genet Epigenet. 9:1179237X177299002017. View Article : Google Scholar : PubMed/NCBI

11 

Wang X, Ivan M and Hawkins SM: The role of MicroRNA molecules and MicroRNA-regulating machinery in the pathogenesis and progression of epithelial ovarian cancer. Gynecol Oncol. 147:481–487. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Zhou K, Liu M and Cao Y: New Insight into microRNA functions in cancer: Oncogene-microRNA-tumor suppressor gene network. Front Mol Biosci. 4:462017. View Article : Google Scholar : PubMed/NCBI

13 

Yi PS and Li JS: High expression of miR-21 is not a predictor of poor prognosis in all patients with hepatocellular carcinoma. Mol Clin Oncol. 8:733–739. 2018.PubMed/NCBI

14 

Ding W, Tan H, Li X, Zhang Y, Fang F, Tian Y, Li J and Pan X: MicroRNA-493 suppresses cell proliferation and invasion by targeting ZFX in human hepatocellular carcinoma. Cancer Biomark. 22:427–434. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Han G, Zhang L, Ni X, Chen Z, Pan X, Zhu Q, Li S, Wu J, Huang X and Wang X: MicroRNA-873 promotes cell proliferation, migration, and invasion by directly targeting TSLC1 in hepatocellular carcinoma. Cell Physiol Biochem. 46:2261–2270. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Chen Z, Zuo X, Zhang Y, Han G, Zhang L, Wu J and Wang X: miR-3662 suppresses hepatocellular carcinoma growth through inhibition of HIF-1α-mediated Warburg effect. Cell Death Dis. 9:5492018. View Article : Google Scholar : PubMed/NCBI

17 

Yu Z, Lin X, Tian M and Chang W: microRNA196b promotes cell migration and invasion by targeting FOXP2 in hepatocellular carcinoma. Oncol Rep. 39:731–738. 2018.PubMed/NCBI

18 

Sun X, Wang M, Liu H and Wang J: MicroRNA-423 enhances the invasiveness of hepatocellular carcinoma via regulation of BRMS1. Am J Transl Res. 9:5576–5584. 2017.PubMed/NCBI

19 

Zhu W, Qian J, Ma L, Ma P, Yang F and Shu Y: miR-346 suppresses cell proliferation through SMYD3 dependent approach in hepatocellular carcinoma. Oncotarget. 8:65218–65229. 2017.PubMed/NCBI

20 

Jiang D, Cho W, Li Z, Xu X, Qu Y, Jiang Z, Guo L and Xu G: miR-758-3p suppresses proliferation, migration and invasion of hepatocellular carcinoma cells via targeting MDM2 and mTOR. Biomed Pharmacother. 96:535–544. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Tong F, Ying Y, Pan H, Zhao W, Li H and Zhan X: MicroRNA-466 (miR-466) functions as a tumor suppressor and prognostic factor in colorectal cancer (CRC). Bosn J Basic Med Sci. 18:252–259. 2018.PubMed/NCBI

22 

Colden M, Dar AA, Saini S, Dahiya PV, Shahryari V, Yamamura S, Tanaka Y, Stein G, Dahiya R and Majid S: MicroRNA-466 inhibits tumor growth and bone metastasis in prostate cancer by direct regulation of osteogenic transcription factor RUNX2. Cell Death Dis. 8:e25722017. View Article : Google Scholar : PubMed/NCBI

23 

Zhou LL, Shen Y, Gong JM, Sun P and Sheng JH: MicroRNA-466 with tumor markers for cervical cancer screening. Oncotarget. 8:70821–70827. 2017.PubMed/NCBI

24 

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

25 

Zheng J, Li C, Wu X, Yang Y, Hao M, Sheng S, Sun Y, Zhang H, Long J and Hu C: Astrocyte elevated gene-1 is a novel biomarker of epithelial-mesenchymal transition and progression of hepatocellular carcinoma in two China regions. Tumour Biol. 35:2265–2269. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Zhu K, Dai Z, Pan Q, Wang Z, Yang GH, Yu L, Ding ZB, Shi GM, Ke AW, Yang XR, et al: Metadherin promotes hepatocellular carcinoma metastasis through induction of epithelial-mesenchymal transition. Clin Cancer Res. 17:7294–7302. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Gong Z, Liu W, You N, Wang T, Wang X, Lu P, Zhao G, Yang P, Wang D and Dou K: Prognostic significance of metadherin overexpression in hepatitis B virus-related hepatocellular carcinoma. Oncol Rep. 27:2073–2079. 2012.PubMed/NCBI

28 

Ahn S, Hyeon J and Park CK: Metadherin is a prognostic predictor of hepatocellular carcinoma after curative hepatectomy. Gut Liver. 7:206–212. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Yoo BK, Emdad L, Su ZZ, Villanueva A, Chiang DY, Mukhopadhyay ND, Mills AS, Waxman S, Fisher RA, Llovet JM, et al: Astrocyte elevated gene-1 regulates hepatocellular carcinoma development and progression. J Clin Invest. 119:465–477. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Srivastava J, Siddiq A, Emdad L, Santhekadur PK, Chen D, Gredler R, Shen XN, Robertson CL, Dumur CI, Hylemon PB, et al: Astrocyte elevated gene-1 promotes hepatocarcinogenesis: Novel insights from a mouse model. Hepatology. 56:1782–1791. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Sarkar D: AEG-1/MTDH/LYRIC in liver cancer. Adv Cancer Res. 120:193–221. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Zhu HD, Liao JZ, He XX and Li PY: The emerging role of astrocyte-elevated gene-1 in hepatocellular carcinoma (Review). Oncol Rep. 34:539–546. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Mao B and Wang G: MicroRNAs involved with hepatocellular carcinoma (Review). Oncol Rep. 34:2811–2820. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Yao M, Wang L, Yao Y, Gu HB and Yao DF: Biomarker-based MicroRNA therapeutic strategies for hepatocellular carcinoma. J Clin Transl Hepatol. 2:253–258. 2014.PubMed/NCBI

35 

Lyra-González I, Flores-Fong LE, González-Garcia I, Medina-Preciado D and Armendáriz-Borunda J: MicroRNAs dysregulation in hepatocellular carcinoma: Insights in genomic medicine. World J Hepatol. 7:1530–1540. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Shi SQ, Ke JJ, Xu QS, Wu WQ and Wan YY: Integrated network analysis to identify the key genes, transcription factors, and microRNAs involved in hepatocellular carcinoma. Neoplasma. 65:66–74. 2018. View Article : Google Scholar : PubMed/NCBI

37 

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

38 

Li J, Zhang N, Song LB, Liao WT, Jiang LL, Gong LY, Wu J, Yuan J, Zhang HZ, Zeng MS and Li M: Astrocyte elevated gene-1 is a novel prognostic marker for breast cancer progression and overall patient survival. Clin Cancer Res. 14:3319–3326. 2008. View Article : Google Scholar : PubMed/NCBI

39 

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

40 

Li WF, Wang G, Zhao ZB and Liu CA: High expression of metadherin correlates with malignant pathological features and poor prognostic significance in papillary thyroid carcinoma. Clin Endocrinol (Oxf). 83:572–580. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Yang G, Zhang L, Lin S, Li L, Liu M, Chen H, Cao M, Liu D, Huang YR and Bo J: AEG-1 is associated with tumor progression in nonmuscle-invasive bladder cancer. Med Oncol. 31:9862014. View Article : Google Scholar : PubMed/NCBI

42 

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

43 

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

44 

Zhao Y, Moran MS, Yang Q, Liu Q, Yuan C, Hong S and Kong B: Metadherin regulates radioresistance in cervical cancer cells. Oncol Rep. 27:1520–1526. 2012.PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 40 Issue 6

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
Jia C, Tang D, Sun C, Yao L, Li F, Hu Y, Zhang X and Wu D: MicroRNA‑466 inhibits the aggressive behaviors of hepatocellular carcinoma by directly targeting metadherin Retraction in /10.3892/or.2022.8357. Oncol Rep 40: 3890-3898, 2018
APA
Jia, C., Tang, D., Sun, C., Yao, L., Li, F., Hu, Y. ... Wu, D. (2018). MicroRNA‑466 inhibits the aggressive behaviors of hepatocellular carcinoma by directly targeting metadherin Retraction in /10.3892/or.2022.8357. Oncology Reports, 40, 3890-3898. https://doi.org/10.3892/or.2018.6763
MLA
Jia, C., Tang, D., Sun, C., Yao, L., Li, F., Hu, Y., Zhang, X., Wu, D."MicroRNA‑466 inhibits the aggressive behaviors of hepatocellular carcinoma by directly targeting metadherin Retraction in /10.3892/or.2022.8357". Oncology Reports 40.6 (2018): 3890-3898.
Chicago
Jia, C., Tang, D., Sun, C., Yao, L., Li, F., Hu, Y., Zhang, X., Wu, D."MicroRNA‑466 inhibits the aggressive behaviors of hepatocellular carcinoma by directly targeting metadherin Retraction in /10.3892/or.2022.8357". Oncology Reports 40, no. 6 (2018): 3890-3898. https://doi.org/10.3892/or.2018.6763