MicroRNA‑548b inhibits proliferation and invasion of hepatocellular carcinoma cells by directly targeting specificity protein 1

Retraction in: /10.3892/etm.2022.11491

  • Authors:
    • Haile Qiu
    • Gehong Zhang
    • Bin Song
    • Junmei Jia
  • View Affiliations

  • Published online on: July 25, 2019     https://doi.org/10.3892/etm.2019.7812
  • Pages: 2332-2340
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Emerging studies have revealed that microRNAs (miRNAs) are aberrantly expressed in hepatocellular carcinoma (HCC), and the dysregulation of miRNAs exerts crucial roles in the carcinogenesis and development of HCC. Therefore, elucidating the relationship between miRNAs and HCC progression is of great importance to develop novel therapeutic techniques and to improve the prognosis of patients with this malignancy. Recently, miR‑548b‑3p (miR‑548b) has been demonstrated to be a cancer‑associated miRNA in tongue squamous cell carcinoma and glioma. However, the expression and function of miR‑548b in HCC remain poorly understood. In the present study, it was found that miR‑548b is expressed at low levels in HCC tissues and cell lines. Decreased miR‑548b expression was found to be positively associated with the clinical features of HCC, including the TNM stage and lymph node metastasis. Functional experiments revealed that upregulation of miR‑548b expression decreased proliferation and invasion of HCC cells. Specificity protein 1 (SP1) was verified to be a direct target of miR‑548b in HCC cells; as Spearman's correlation analysis identified miR‑548b expression to be negatively correlated with that of SP1 expression in HCC tissue specimens. In addition, SP1 inhibition exhibited similar effects as miR‑548b overexpression in HCC cells. SP1 reintroduction significantly reversed the suppressive effects of miR‑548b upregulation on the proliferation and invasion of HCC cells. In conclusion, the results presented in the present study demonstrated that miR‑548b may serve as a tumor suppressive miRNA that inhibits the proliferation and invasion of HCC cells by directly targeting SP1. Consequently, miR‑548b can be exploited as a novel therapeutic target for treating patients with HCC in the future, but this needs to be investigated further.

Introduction

Hepatocellular carcinoma (HCC), or cancer of the liver, is one of the most prevalent human malignancies. Currently, HCC ranks fifth in incidence and is the third leading cause of cancer-associated death globally (1). It is estimated that there will be ~854,000 new cases and 810,000 deaths due to HCC annually worldwide (2). The rates of morbidity and mortality of HCC have been increasing in the past decade, particularly in China (3). Therapeutic strategies including surgical resection, radiofrequency ablation, chemoradiotherapy and liver transplantation, have been developed and are widely used (46). Unfortunately, the clinical outcomes of patients with HCC remain unsatisfactory due to recurrence as a result of frequent tumor invasion, intrahepatic spreading and extrahepatic metastasis (7,8). The carcinogenesis and pathogenesis of HCC are complex, and have been reported to involve a multitude of factors, including Hepatitis B or C viral infection, exposure to aflatoxin, excessive drinking, genetic and epigenetic alterations (911). However, the precise underlying mechanism for HCC pathogenesis remain poorly characterized. Therefore, it is necessary to uncover the molecular mechanisms involved in HCC initiation and progression to develop more effective therapeutic interventions, improving prognosis for patients with this disease.

MicroRNAs (miRNAs) are a group of evolutionarily conserved, noncoding and short RNA molecules that are typically 18–25 nucleotides in length (12). Mature miRNAs can post-transcriptionally regulate gene expression by directly targeting the 3′-untranslated regions (3′-UTRs) of target genes via complementary base pairing (13). This form of miRNA-3′UTR interaction results in the target messenger RNA (mRNA) being degraded and/or translationally suppressed (13). In this manner, miRNAs have been demonstrated to serve crucial roles in a wide range of physiological processes including cell proliferation, cell cycle progression, differentiation, metabolism, metastasis and tumorigenesis (14). In particular, accumulating evidence supports that miRNAs are aberrantly expressed in a substantial number of human disorders including cancer (1517); with changes in miRNA expression being reported in HCC (18), colorectal cancer (19), lung cancer and bladder cancer (20,21). In HCC alone, miRNAs have been implicated either as oncogenes or tumor suppressors during oncogenesis and development (2224). For instance, miR-1306-3p has been reported to promote growth and metastasis of HCC cells in vitro and in vivo by directly targeting F-box and leucine rich repeat protein 5 (25), whereas the upregulation of miR-506 has been implied to suppress the aggressive characteristics of HCC by targeting interleukin (IL)-8 both in vitro and in vivo (26).

miR-548b-3p (miR-548b) has been recently demonstrated to be a cancer-associated miRNA in tongue squamous cell carcinoma and glioma (27,28). However, the expression and function of miR-548b in HCC remain unclear. Therefore, it was hypothesized that miR-548b may exert a crucial role in the progression and development of HCC. The aim of the present study was to assess miR-548b expression in HCC and to determine its clinical significance in HCC patients. Notably, in-depth roles of miR-548b in the development of HCC with any potential molecular mechanisms involved were explored. Investigating the expression profile and detailed roles of miR-548b in HCC may be useful for identifying promising therapeutic techniques for use in patients with this malignancy. Results from the present study found that miR-548b was downregulated endogenously in HCC, the ectopic expression of which in the form of a miRNA mimic suppressed the aggressive phenotypes of HCC. This was mechanistically revealed to be due to the miR-548b-mediated targeting of the SP1 protein. Altogether, these observations implicate this miRNA to be a potential target for the therapeutic design of HCC treatment.

Materials and methods

Tissue samples and cell lines

A total of 51 pairs of HCC tissues and matched adjacent noncancerous tissues were collected from patients (17 females, 34 males; age range, 46–69 years) in the First Hospital of Shanxi Medical University (Shanixi, China) between May 2014 and September 2017. Tissue specimens were obtained from patients who were diagnosed with HCC and had not been treated with chemotherapy or radiotherapy prior to surgical resection. Fibrolamellar carcinoma is a rare form of HCC, and it was confirmed, by a pathologist, that no patients diagnosed with this type of HCC were recruited for this study. Patients that had received chemotherapy or radiotherapy were excluded from this research. Following resection, all tissues were rapidly frozen in liquid nitrogen and stored at −80°C until total RNA extraction. The protocol of this study was approved by the Ethics Committee of the First Hospital of Shanxi Medical University, and written informed consent was provided by all enrolled subjects.

Two human HCC cell lines (Huh7 and Hep3B) and an immortalized normal human liver epithelial cell (L-02) were purchased from Cell Bank of the Typical Culture Preservation Committee of the Chinese Academy of Sciences (Type Culture Collection of the Chinese Academy of Sciences). Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 U/ml streptomycin (all from Gibco; Thermo Fisher Scientific, Inc.) was used to maintain the cell lines. The cells were routinely cultured at 37°C in a humidified incubator containing 5% CO2.

Cell transfection with miRNA mimics, small interfering RNA (siRNA) and plasmid

miR-548b mimics and negative control miRNA mimics (miR-NC) were obtained from GenePharma (Shanghai GenePharma Co., Ltd.). siRNA against SP1 (SP1 siRNA) and negative control siRNA (NC siRNA) were chemically synthesized by Ribobio (Guangzhou RiboBio Co., Ltd.). The SP1 siRNA sequence was 5′-GCAACAUGGGAAUUAUGAATT-3′ and the NC siRNA sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. Specificity protein 1 (SP1) expression plasmid pcDNA3.1-SP1 (pc-SP1) and corresponding empty pcDNA3.1 plasmid were purchased from GeneCopoeia (GeneCopoeia, Inc.). Cells were plated into 6-well plates at a density of 5×105 cells/well. miRNA mimics (100 pmol), siRNA (100 pmol) or plasmid (4 µg) were subsequently introduced into the cells using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to manufacturer's protocols. Following a 48 h incubation, efficacy was determined using reverse transcription-quantitative PCR (RT-qPCR) or western blot analysis. Cell Counting kit-8 (CCK-8) and invasion assays were carried out at 24 and 48 h post-transfection, respectively.

RNA extraction and reverse transcription-quantitative PCR (RT-qPCR)

The isolation of total RNA form HCC tissues or cells was performed using the TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to manufacturer's protocol. Total RNA was reverse-transcribed into cDNA using the Taqman MicroRNA Reverse Transcription Kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) for miRNA analysis and PrimeScript RT Reagent kit (Takara Biotechnology Co., Ltd.) for mRNA analysis. The reverse transcription temperature protocol for miRNA was as follows: 16°C for 30 min, 42°C for 30 min and 85°C for 5 min. The reverse transcription temperature protocol for mRNA was as follows: 37°C for 15 min and 85°C for 5 sec. qPCR was performed using Taqman MicroRNA Assay kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) and SYBR Premix Ex Taq™ kit (Takara Biotechnology Co., Ltd.), respectively. The qPCR cycling conditions for miR-548b 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. The qPCR cycling conditions for SP1 mRNA 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 were used as normalization for miR-548b and SP1 mRNA, respectively. Relative gene expression was calculated using the 2−ΔΔCq method (29). The primers were designed as follows: miR-548b forward, 5′-ACACTCCAGCTGGGCAAAAATCTCAAT-3′ and reverse, 5′-CTCAACTGGTGTCGTGGAAACTGGTGTC-3′; U6 forward, 5′-CTCGCTTCGGCAGCACAGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGTCGCTTCACGAATT-3′; SP1 forward, 5′-TGGTGGGCAGTATGTTGT-3′ and reverse, 5′-GCTATTGGCATTGGTGAA-3′; and GAPDH forward, 5′-GGGCAGTATGTTGT-3′ and reverse, 5′-GCTATTGGCATTGGTGAA-3′.

CCK-8 assay

CCK-8 assay (Dojindo Molecular Technologies, Inc.) was used to assess cellular proliferation, according to manufacturer's protocol. In detail, transfected cells were harvested at 24 h and seeded into 96-well plates at 3×103 cells/well. Cells were subsequently incubated at 37°C in a humidified incubator containing 5% CO2 for 0, 24, 48 and 72 h, before a total of 10 µl CCK-8 reagent was added into each well at every timepoint. Following incubation for an additional 2 h at 37°C, the optical density (450 nm) of each well was detected using an automated microplate reader (Bio-Rad Laboratories).

Invasion assay

In total, 5×104 of transfected cells suspended in 200 µl FBS-free DMEM medium were inoculated into the upper chamber of each Matrigel (BD Biosciences)-coated Transwell inserts (Corning Inc.), with 8 µm pore size in polycarbonate membranes. A total of 500 µl of DMEM supplemented with 20% FBS was plated in the lower chamber. Following 24 h of incubation at 37°C with 5% CO2, the non-invaded cells remaining on the upper surface of the polycarbonate membranes were carefully removed with a cotton swab. The invaded cells on the lower surface of the polycarbonate membranes were subsequently fixed with 4% paraformaldehyde at room temperature for 30 min, stained with 0.5% crystal violet at room temperature for 30 min and imaged with an IX73 inverted microscope (magnification, ×200; Olympus Corporation). Five random chosen fields of view were analyzed for each Transwell insert.

Bioinformatics analysis

TargetScan (Release 7.2; March 2018; www.targetscan.org), microRNA (August 2010 Release; http://www.microrna.org/microrna/home.do) and miRDB (Last modified: January 22, 2019; www.mirdb.org) were employed investigate potential targets of miR-548b.

Luciferase activity assay

The sequences of SP1 3′-UTR containing the putative wild-type (wt) or mutant (mut) miR-548b binding region were amplified by GenePharma (Shanghai GenePharma Co., Ltd.), and inserted into the pGL3 luciferase reporter vector (Promega Corporation). The generated luciferase plasmids were referred to as pGL3-SP1-3′-UTR wt and pGL3-SP1-3′-UTR mut respectively thereafter. Cells were seeded into 24-well plates with an initial density of 1.0×105 cells/well. The miR-548b mimic or miR-NC was subsequently co-transfected along with pGL3-SP1-3′-UTR wt or pGL3-SP1-3′-UTR mut into the cells using Lipofectamine® 2000, in accordance with the manufacturer's protocol. Following 48 h of transfection, the cells were collected and analyzed using a Dual Luciferase reporter system (Promega Corporation). Luciferase activity was normalized to Renilla activity.

Western blot analysis

Homogenized tissues or cells were first lysed using cold radio-immunoprecipitation lysis buffer (Santa Cruz Biotechnology, Inc.). Bicinchoninic acid assay protein assay kit II (Bio-Rad Laboratories, Inc.) was used to quantify the total protein concentration. Equal amounts of total protein (30 µg) were subsequently separated by electrophoresis using 10% sodium dodecyl sulfate (SDS)-polyacrylamide gels before transferal onto PVDF membranes (Beyotime Institute of Biotechnology). Non-specific binding sites were subsequently blocked by incubating the membranes in 5% fat-free milk dissolved in tris-buffered saline containing 0.1% Tween-20 (TBS-T) for 2 h at room temperature. Following blocking the membranes were incubated overnight at 4°C with primary antibodies as follows: Rabbit anti-human SP1 antibody (cat. no. ab124804; 1:1,000) and rabbit anti-human GAPDH antibody (cat. no. ab181602; 1:1,000; both from Abcam). Following extensive washing with TBS-T, horseradish peroxidase-conjugated goat anti-rabbit secondary antibody (cat. no. ab205718; 1:5,000; Abcam) was utilised to incubate the membranes at room temperature for 1 h. Finally, the protein signals were visualized using an enhanced chemiluminescence detection kit (EMD Millipore). Quantity One software version 4.62 (Bio-Rad Laboratories, Inc.) was used for densitometry.

Statistical analysis

All data were presented as mean ± standard deviation. The differences between groups were examined using Student's t-test or one-way analysis of variance, followed by Student-Newman-Keuls test as a post hoc test. The changes in miR-548b and SP1 mRNA expression levels between HCC and adjacent noncancerous tissues were determined using paired Student's t-test. χ2 test was utilized to evaluate the association between miR-548b and the clinicopathological features of HCC patients. The association between miR-548b and SP1 mRNA levels in HCC tissues was assessed using Spearman's correlation analysis. All statistical analyses were carried out using the SPSS 20.0 software package (IBM Corp.). P<0.05 was considered to indicate a statistically significant difference.

Results

miR-548b is downregulated in HCC tissues and cell lines

To explore the possible involvement of miR-548b in HCC, miR-548b expression was first measured in 51 pairs of HCC tissues and matched adjacent noncancerous tissues by RT-qPCR. The data indicated that miR-548b expression was decreased in HCC tissues when compared with that in adjacent noncancerous tissues (Fig. 1A). The clinical value of miR-548b in HCC was subsequently evaluated. To achieve this, all enrolled HCC patients were divided into miR-548b low or high expression groups based on the median value of miR-548b. Reduced miR-548b expression was illustrated to be significantly associated with the TNM stage (P=0.032) and lymph node metastasis (P=0.029) of patients with HCC (Table I). To support this observation, the expression levels of miR-548b was determined in two human HCC cell lines (Huh7 and Hep3B) and an immortalized normal human liver epithelial cell line (L-02). miR-548b was revealed to be significantly downregulated in the two HCC cell lines compared with that in the L-02 cell line (Fig. 1B). These results suggest that the downregulation of miR-548b may be a frequent event in the development of HCC.

Table I.

Association between miR-548b expression and clinical features of HCC patients.

Table I.

Association between miR-548b expression and clinical features of HCC patients.

miR-548b expression

Clinical featuresLowHighχ2 valueP-value
Age (years) 0.4810.488
  <551512
  ≥551113
Sex 0.1570.692
  Female89
  Male1816
Tumor size (cm) 3.2960.069
  <5915
  ≥51710
Differentiation 0.1840.668
  Well and moderate1314
  Poor1311
TNM stage 4.5810.032
  I–II1118
  III–IV157
Lymph node metastasis 4.7630.029
  Negative1219
  Positive146
miR-548b attenuates HCC cell proliferation and invasion

To determine whether miR-548b exerts specific functions in HCC development, miR-548b was overexpressed in Huh7 and Hep3B cells by transfection using miR-548b mimics. Ectopic transfection with miR-548b mimics significantly increased the levels of miR-548b expression in Huh7 and Hep3B cells, suggesting that the transfection was successful (Fig. 2A). CCK-8 assay was subsequently applied to investigate the effect of miR-548b transfection on the proliferation of HCC cells. Ectopic miR-548b expression was revealed to significantly inhibit cellular proliferative ability of Huh7 and Hep3B cells (Fig. 2B).

In addition to proliferation, invasion assay was conducted to assess the regulatory role of miR-548b in HCC cell invasion. Huh7 and Hep3B cells transfected with miR-548b mimics exhibited significantly reduced capacities of cell invasion compared with those transfected with miR-NC (Fig. 2C). In summary, these findings implied that miR-548b exerts antitumor effects in HCC by inhibiting cell proliferation and invasion.

SP1 is a direct target gene of miR-548b in HCC cells

To explore the potential mechanism underlying the tumor-suppressive properties of miR-548b in HCC, bioinformatics analysis was performed to predict the potential target of miR-548b. SP1 was uncovered as a potential target of miR-548b in databases including TargetScan, miRDB, and microRNA (Fig. 3A). SP1 was previously reported to be implicated in the regulation of HCC progression (3034), and was therefore chosen for further investigation. To examine whether miR-548b can directly bind to the 3′-UTR of SP1, luciferase reporter plasmids were chemically synthesized, and subsequently co-transfected with either miR-548b mimics or miR-NC in Huh7 and Hep3B cells. In Huh7 and Hep3B cells transfected with the miR-548b mimic, luciferase activity was demonstrated to be significantly decreased in cells co-transfected with the reporter plasmid encoding wt SP1 3′-UTR, but not in those transfected with the plasmid encoding mut SP1 3′-UTR (Fig. 3B).

To support this observation made in the HCC cell lines, SP1 expression was also measured in HCC tissues and matched adjacent noncancerous tissues. RT-qPCR analysis appeared to verify this finding, as the expression level of SP1 mRNA was notably increased in HCC tissues compared with that in matched adjacent noncancerous tissue (Fig. 3C). Spearman's correlation analysis indicated that miR-548b expression was negatively correlated with SP1 mRNA levels in HCC tissues (r=−0.5955; Fig. 3D). Furthermore, RT-qPCR and western blot analysis showed that following ectopic miR-548b mimic expression, SP1 mRNA and protein levels were significantly reduced in Huh7 and Hep3B cells (Fig. 3E and F). Overall, SP1 appeared to be a direct target of miR-548b in HCC cells.

SP1 inhibition is able to mimic the inhibitory effects of miR-548b upregulation in HCC cells

A loss-of-function study was performed to explore the roles of SP1 in HCC cells. SP1 siRNA was applied to knockdown endogenous SP1 expression in Huh7 and Hep3B cells. The protein level of SP1 was efficiently suppressed in Huh7 and Hep3B cells by SP1 siRNA transfection, as determined by western blot analysis (Fig. 4A). Subsequently, results from CCK-8 and invasion assays revealed that SP1 knockdown resulted in significantly reduced proliferation and invasion in Huh7 and Hep3B cells (Fig. 4B and C), indicating that SP1 inhibition may mimic the effects of miR-548b upregulation. These findings further support the notion that SP1 is a direct target gene of miR-548b in HCC cells.

Antitumor roles of miR-548b in HCC cells are exerted through downregulation of SP1

To clarify if miR-548b-dependent inhibition of HCC cell proliferation and invasion was mediated directly by SP1, rescue experiments were performed on Huh7 and Hep3B cell lines. Plasmids expressing either pcDNA3.1-SP1 (pc-SP1) or their corresponding empty pcDNA3.1 were transfected into Huh7 and Hep3B cells along with the miR-548b mimics. First, RT-qPCR analysis was used to identify the upregulation of SP1 mRNA expression in Huh7 and Hep3B cells that were transfected with pc-SP1 (Fig. 5A; P<0.05). Western blot analysis demonstrated that co-transfecting miR-548b mimics with the pc-SP1 plasmid significantly recovered SP1 protein levels in Huh7 and Hep3B cells (Fig. 5B). Reintroduction of SP1 using this approach also rescued the suppressive effects of ectopic miR-548b expression on Huh7 and Hep3B cell proliferation and invasion significantly (Fig. 5C and D). In conclusion, miR-548b was demonstrated as a tumor suppressor in HCC progression, at least partly, through downregulating SP1 expression.

Discussion

A number of studies have emerged revealing that miRNAs are aberrantly expressed in HCC (3537). Changes in miRNA expression have been continuously reported to serve as the main drivers of HCC carcinogenesis and development by regulating a variety of cancer-associated biological processes (22). Notably, miRNAs have been proposed as diagnostic and prognostic biomarkers, as well as effective therapeutic targets for anticancer treatment (38). Therefore, elucidating the relationship between miRNAs and HCC progression is of great importance to develop novel therapeutic techniques and to improve the prognosis of patients with this malignancy. To the best of our knowledge, the present study is the first to analyze miR-548b expression in HCC, clarify the clinical value of miR-548b in HCC, and to investigate the specific functions of miR-548b in the development of HCC. Importantly, the mechanism underlying the tumor-suppressing properties of miR-548b in HCC was also explored.

miR-548b is found to be upregulated in tongue squamous cell carcinoma (27). Multivariate analyses validated miR-548b as an independent biomarker for predicting the prognosis of patients with this malignancy (27). In contrast, levels of miR-548b expression is low in glioma tissues and cell lines (28). The inconsistent observations among miR-548b expression implicate tissue specificity of miR-548b expression in human malignancies. However, the expression pattern of miR-548b in HCC remains unclear. Therefore, in the present study RT-qPCR was performed to measure miR-548b expression in HCC tissues and cell lines. miR-548b expression was revealed to be noticeably decreased in both HCC tissues and cell lines. Reduced miR-548b expression was observed to strongly correlate with the TNM stage and lymph node metastasis of HCC patients. These findings suggest that miR-548b may be a potential diagnostic biomarker for patients with HCC, but this needs to be investigated further.

Functionally, miR-548b upregulation has been revealed to restrict the proliferative, colony formation and invasive abilities of glioma cells in vitro, whereas miR-548b overexpression suppressed the tumor growth of glioma cells in vivo (28). Nevertheless, the detailed role of miR-548b in the progression and development of HCC remain largely elusive. As a result, a series of functional experiments were performed in the present study to determine the regulatory effects of miR-548b overexpression in HCC cells. The results demonstrated that ectopic miR-548b expression attenuated the proliferative and invasive capacities of HCC cells. These findings suggest that miR-548b might be a promising molecular target for the therapy of patients with HCC.

Phosphatase and tensin homolog deleted on chromosome ten (PTEN) (27) and metastasis tumor-associated protein-2 (MTA2) have been identified as direct targets of miR-548b (28). To uncover the molecular mechanism responsible for the cellular response to miR-548b, the present study attempted to explore whether SP1 may be a novel target of miR-548b in HCC cells. Firstly, bioinformatics analysis indicated that SP1 contains a putative binding site for miR-548b in its 3′-UTR. Secondly, luciferase reporter assay demonstrated that miR-548b could directly interact with the 3′-UTR of SP1 gene in HCC cells. Thirdly, RT-qPCR and western blot analysis revealed that ectopic miR-548b expression significantly reduced SP1 expression in HCC cells at both mRNA and protein level. SP1 was upregulated in HCC tissues, and its upregulation was inversely correlated with miR-548b expression levels. In addition, SP1 knockdown was able to mimic the tumor suppressive roles of miR-548b overexpression in HCC cells. Finally, ectopic SP1 expression abolished the inhibitory effects in HCC cell proliferation and invasion caused by miR-548b upregulation. These results sufficiently attest that SP1 is a direct and functional downstream target of miR-548b in HCC cells, and that SP1 inhibition is required for the tumor suppressor activity of miR-548b. However, the correlation between SP1 and PTEN as well as MTA2 in HCC was not examined in the present study, which serves as a limitation and will be studied in further investigations.

SP1, located at 12q13.1, is a sequence-specific DNA-binding protein (39). It possesses the ability to directly bind GC/GT-rich promoter elements via its C2H2-type zinc fingers at the C-terminal domain, and therefore regulate the activity of target gene promoters (40). Previous studies reported that SP1 is expressed at high levels in a variety of human malignant tumors including colorectal cancer (41), osteosarcoma (42), lung cancer (43), and pancreatic cancer (44). Notably, SP1 was also found to be upregulated in HCC tissues and cell lines (30,31). Multivariate Cox regression analysis validated SP1 to be an independent predictor for the death rate of patients with HCC (31). In addition, HCC patients with high SP1 expression have been demonstrated to be associated with poorer clinical outcomes compared with those with low expression (31). In addition, SP1 has been implicated in the occurrence and development of HCC through regulation of a number of biological processes (3234). Observations from this study demonstrated that miR-548b directly targets SP1 to suppress the aggressive phenotypes of HCC cells. Therefore, silencing SP1 expression by miR-548b restoration may be a potential therapeutic approach for HCC patients.

In summary, the present study illustrated that miR-548b was downregulated in HCC, which is in turn associated with malignant clinical features in HCC patients. Ectopic miR-548b expression inhibited the proliferative and invasive ability of HCC cells by directly targeting SP1, suggesting that aberrant suppression of miR-548b expression may be an important driver for HCC formation and progression. Notably, the miR-548b/SP1 pathway may be an effective therapeutic target for the management of patients with HCC. However, further investigations are required to address this issue.

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

JJ designed this research study. JJ and HQ performed the RT-qPCR, western blot analyses and luciferase reporter assays. CCK-8 and invasion assays were carried out by GZ and BS. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of First Hospital of Shanxi Medical University (Shanxi, China), and was performed in accordance with the Declaration of Helsinki and the guidelines of the Ethics Committee of First Hospital of Shanxi Medical University. Written informed consent was provided by all enrolled subjects.

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 

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

3 

Peng W, Chen Y, Jiang Q and Zheng Y: Spatial analysis of hepatocellular carcinoma and socioeconomic status in China from a population-based cancer registry. Cancer Epidemiol. 34:29–33. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Chawla A and Ferrone C: Hepatocellular carcinoma surgical therapy: Perspectives on the current limits to resection. Chin Clin Oncol. 7:482018. View Article : Google Scholar : PubMed/NCBI

5 

Kulik L and El-Serag HB: Epidemiology and management of hepatocellular carcinoma. Gastroenterology. 156:477–491.e1. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Mak LY, Cruz-Ramón V, Chinchilla-López P, Torres HA, LoConte NK, Rice JP, Foxhall LE, Sturgis EM, Merrill JK, Bailey HH, et al: Global epidemiology, prevention, and management of hepatocellular carcinoma. Am Soc Clin Oncol Educ Book. 38:262–279. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Bosetti C, Turati F and La Vecchia C: Hepatocellular carcinoma epidemiology. Best Pract Res Clin Gastroenterol. 28:753–770. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein KD, Alteri R and Jemal A: Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 66:271–289. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Jeong SW, Jang JY and Chung RT: Hepatitis C virus and hepatocarcinogenesis. Clin Mol Hepatol. 18:347–356. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Nishida N and Goel A: Genetic and epigenetic signatures in human hepatocellular carcinoma: A systematic review. Curr Genomics. 12:130–137. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Zhang YJ: Interactions of chemical carcinogens and genetic variation in hepatocellular carcinoma. World J Hepatol. 2:94–102. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Agarwal V, Bell GW, Nam JW and Bartel DP: Predicting effective microRNA target sites in mammalian mRNAs. ELife. 4:2015.doi: 10.7554/eLife.05005. View Article : Google Scholar

14 

Hwang HW and Mendell JT: MicroRNAs in cell proliferation, cell death, and tumorigenesis. Br J Cancer. 96 (Suppl):R40–R44. 2007.PubMed/NCBI

15 

Garzon R, Calin GA and Croce CM: MicroRNAs in cancer. Annu Rev Med. 60:167–179. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Garzon R and Croce CM: MicroRNAs and cancer: Introduction. Semin Oncol. 38:721–723. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Garzon R and Marcucci G: Potential of microRNAs for cancer diagnostics, prognostication and therapy. Curr Opin Oncol. 24:655–659. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Vasuri F, Visani M, Acquaviva G, Brand T, Fiorentino M, Pession A, Tallini G, D'Errico A and de Biase D: Role of microRNAs in the main molecular pathways of hepatocellular carcinoma. World J Gastroenterol. 24:2647–2660. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Yang S, Sun Z, Zhou Q, Wang W, Wang G, Song J, Li Z, Zhang Z, Chang Y, Xia K, et al: MicroRNAs, long noncoding RNAs, and circular RNAs: Potential tumor biomarkers and targets for colorectal cancer. Cancer Manag Res. 10:2249–2257. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Iqbal MA, Arora S, Prakasam G, Calin GA and Syed MA: MicroRNA in lung cancer: Role, mechanisms, pathways and therapeutic relevance. Mol Aspects Ned. Aug 17–2018.(Epub ahead of print). doi: 10.1016/j.mam.2018.07.003. View Article : Google Scholar

21 

Homami A and Ghazi F: MicroRNAs as biomarkers associated with bladder cancer. Med J Islam Repub Iran. 30:4752016.PubMed/NCBI

22 

Xu J, Li J, Zheng TH, Bai L and Liu ZJ: MicroRNAs in the occurrence and development of primary hepatocellular carcinoma. Adv Clin Exp Med. 25:971–975. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Jin K, Li T, Sanchez-Duffhues G, Zhou F and Zhang L: Involvement of inflammation and its related microRNAs in hepatocellular carcinoma. Oncotarget. 8:22145–22165. 2017.PubMed/NCBI

24 

Hayes CN and Chayama K: MicroRNAs as biomarkers for liver disease and hepatocellular carcinoma. Int J Mol Sci. 17:2802016. View Article : Google Scholar : PubMed/NCBI

25 

He ZJ, Li W, Chen H, Wen J, Gao YF and Liu YJ: miR-1306-3p targets FBXL5 to promote metastasis of hepatocellular carcinoma through suppressing snail degradation. Biochem Biophys Res Commun. 504:820–826. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Wang Z, Si M, Yang N, Zhang H, Fu Y, Yan K, Zong Y, Zhu N and Wei Y: MicroRNA-506 suppresses invasiveness and metastasis of human hepatocellular carcinoma cells by targeting IL8. Am J Cancer Res. 8:1586–1594. 2018.PubMed/NCBI

27 

Berania I, Cardin GB, Clement I, Guertin L, Ayad T, Bissada E, Nguyen-Tan PF, Filion E, Guilmette J, Gologan O, et al: Four PTEN-targeting co-expressed miRNAs and ACTN4-targeting miR-548b are independent prognostic biomarkers in human squamous cell carcinoma of the oral tongue. Int J Cancer. 141:2318–2328. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Pan Y, Liang W, Zhao X, Liu L, Qing Y and Li Y: miR-548b inhibits the proliferation and invasion of malignant gliomas by targeting metastasis tumor-associated protein-2. Neuroreport. 27:1266–1273. 2016. View Article : Google Scholar : PubMed/NCBI

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 

Liu C, Zhu J, Liu F, Wang Y and Zhu M: MicroRNA-138 targets SP1 to inhibit the proliferation, migration and invasion of hepatocellular carcinoma cells. Oncol Lett. 15:1279–1286. 2018.PubMed/NCBI

31 

Liu L, Ji P, Qu N, Pu WL, Jiang DW, Liu WY, Li YQ and Shi RL: The impact of high co-expression of Sp1 and HIF1a on prognosis of patients with hepatocellular cancer. Oncol Lett. 12:504–512. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Yin P, Zhao C, Li Z, Mei C, Yao W, Liu Y, Li N, Qi J, Wang L, Shi Y, et al: Sp1 is involved in regulation of cystathionine gamma-lyase gene expression and biological function by PI3K/Akt pathway in human hepatocellular carcinoma cell lines. Cell Signal. 24:1229–1240. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Zhao N, Li S, Wang R, Xiao M, Meng Y, Zeng C, Fang JH, Yang J and Zhuang SM: Expression of microRNA-195 is transactivated by Sp1 but inhibited by histone deacetylase 3 in hepatocellular carcinoma cells. Biochim Biophys Acta. 1859:933–942. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Yen WH, Ke WS, Hung JJ, Chen TM, Chen JS and Sun HS: Sp1-mediated ectopic expression of T-cell lymphoma invasion and metastasis 2 in hepatocellular carcinoma. Cancer Med. 5:465–477. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Klingenberg M, Matsuda A, Diederichs S and Patel T: Non-coding RNA in hepatocellular carcinoma: Mechanisms, biomarkers and therapeutic targets. J Hepatol. 67:603–618. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Shen S, Lin Y, Yuan X, Shen L, Chen J, Chen L, Qin L and Shen B: Biomarker MicroRNAs for diagnosis, prognosis and treatment of hepatocellular carcinoma: A functional survey and comparison. Sci Rep. 6:383112016. View Article : Google Scholar : PubMed/NCBI

37 

Szabo G and Bala S: MicroRNAs in liver disease. Nat Rev Gastroenterol Hepatol. 10:542–552. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Hu Y, Wang H, Chen E, Xu Z, Chen B and Lu G: Candidate microRNAs as biomarkers of thyroid carcinoma: A systematic review, meta-analysis, and experimental validation. Cancer Med. 5:2602–2614. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Chang WC and Hung JJ: Functional role of post-translational modifications of Sp1 in tumorigenesis. J Biomed Sci. 19:942012. View Article : Google Scholar : PubMed/NCBI

40 

Davie JR, He S, Li L, Sekhavat A, Espino P, Drobic B, Dunn KL, Sun JM, Chen HY, Yu J, et al: Nuclear organization and chromatin dynamics-Sp1, Sp3 and histone deacetylases. Adv Enzyme Regul. 48:189–208. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Bajpai R and Nagaraju GP: Specificity protein 1: Its role in colorectal cancer progression and metastasis. Crit Rev Oncol Hematol. 113:1–7. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Qian M, Gong H, Yang X, Zhao J, Yan W, Lou Y, Peng D, Li Z and Xiao J: MicroRNA-493 inhibits the proliferation and invasion of osteosarcoma cells through directly targeting specificity protein 1. Oncol Lett. 15:8149–8156. 2018.PubMed/NCBI

43 

Deacon K, Onion D, Kumari R, Watson SA and Knox AJ: Elevated SP-1 transcription factor expression and activity drives basal and hypoxia-induced vascular endothelial growth factor (VEGF) expression in non-small cell lung cancer. J Biol Chem. 287:39967–39981. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Black AR, Black JD and Azizkhan-Clifford J: Sp1 and kruppel-like factor family of transcription factors in cell growth regulation and cancer. J Cell Physiol. 188:143–160. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2019
Volume 18 Issue 3

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Qiu H, Zhang G, Song B and Jia J: MicroRNA‑548b inhibits proliferation and invasion of hepatocellular carcinoma cells by directly targeting specificity protein 1 Retraction in /10.3892/etm.2022.11491. Exp Ther Med 18: 2332-2340, 2019
APA
Qiu, H., Zhang, G., Song, B., & Jia, J. (2019). MicroRNA‑548b inhibits proliferation and invasion of hepatocellular carcinoma cells by directly targeting specificity protein 1 Retraction in /10.3892/etm.2022.11491. Experimental and Therapeutic Medicine, 18, 2332-2340. https://doi.org/10.3892/etm.2019.7812
MLA
Qiu, H., Zhang, G., Song, B., Jia, J."MicroRNA‑548b inhibits proliferation and invasion of hepatocellular carcinoma cells by directly targeting specificity protein 1 Retraction in /10.3892/etm.2022.11491". Experimental and Therapeutic Medicine 18.3 (2019): 2332-2340.
Chicago
Qiu, H., Zhang, G., Song, B., Jia, J."MicroRNA‑548b inhibits proliferation and invasion of hepatocellular carcinoma cells by directly targeting specificity protein 1 Retraction in /10.3892/etm.2022.11491". Experimental and Therapeutic Medicine 18, no. 3 (2019): 2332-2340. https://doi.org/10.3892/etm.2019.7812