MicroRNA‑873 inhibits the proliferation and invasion of endometrial cancer cells by directly targeting hepatoma‑derived growth factor

Retraction in: /10.3892/etm.2022.11475

  • Authors:
    • Qin Wang
    • Weipei Zhu
  • View Affiliations

  • Published online on: June 26, 2019     https://doi.org/10.3892/etm.2019.7713
  • Pages: 1291-1298
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

An accumulation of evidence has demonstrated that abnormal microRNA (miRNA or miR) expression is associated with different types of cancer, including endometrial cancer (EC). The dysregulation of miRNAs may serve important roles in the development and progression of EC by regulating multiple aggressive biological behaviors, including cell proliferation, apoptosis, metastasis and angiogenesis. An in‑depth understanding of the miRNAs associated with EC initiation and progression may be crucial for identifying successful therapeutic techniques. miR‑873 has been demonstrated to be dysregulated in different types of cancer. However, the expression status and regulatory roles of miR‑873 are yet to be elucidated in EC. In the present study, reverse transcription‑quantitative PCR was carried out to detect miR‑873 expression in EC tissues and cell lines. Cell Counting Kit‑8 and in vitro invasion assays were utilized to determine the influence of miR‑873 on the proliferation and invasion of EC cells. miR‑873 expression was revealed to be downregulated in EC tissues and cell lines. Decreased miR‑873 expression was significantly associated with International Federation of Gynecology and Obstetrics stage and lymph node metastasis of patients with EC. Functional assays revealed that resumed miR‑873 expression suppressed the proliferation and invasion of EC cells. Additionally, hepatoma‑derived growth factor (HDGF) was indicated to be a direct target gene of miR‑873 in EC cells. HDGF was highly expressed in EC tissues and inversely correlated with miR‑873 expression. HDGF silencing also imitated the tumor‑suppressor activity of miR‑873 overexpression in EC cells. A series of rescue experiments identified that recovered HDGF expression hindered the anti‑proliferative and anti‑invasive roles of miR‑873 upregulation in EC cells. In conclusion, the present study indicated that miR‑873 serves an important role as a tumor suppressor in EC development by directly targeting HDGF. The results may provide a novel insight into clinical treatments, which can be used to prevent EC aggression.

Introduction

Endometrial cancer (EC) is one of the most common gynecologic malignancies, ranking as the sixth most common malignant tumor in females, with an estimated 226,000 new cases being diagnosed per year, worldwide (1,2). The current therapeutic approaches for patients with EC are surgical resection, brachytherapy, radiotherapy and chemotherapy (3). A variety of factors, including obesity, hypertension, postmenopausal status, infertility, a family history of EC and long-term use of estrogens have been demonstrated to serve a role in endometrial carcinogenesis and progression (4). However, the specific events associated with EC pathogenesis remain unidentified. Advancements have been made in EC diagnosis and therapy such that patient prognosis has improved, but therapeutic outcomes still exhibit low success, with a survival rate of only 5 years (5,6). Further elucidation of the mechanisms involving EC oncogenesis is therefore urgently required to identify effective therapeutic techniques.

MicroRNAs (miRNAs or miRs) are a large family of short, single stranded non-coding RNA molecules (7). miRNAs serve as specific sequence regulators in gene expression by directly pairing with the 3′-untranslated regions (UTRs) of their target genes, causing mRNA cleavage or translational inhibition (8). A single miRNA can regulate a variety of target genes and a single gene can be negatively modulated by multiple miRNAs (9). The abnormal expression of miRNA has been identified in the majority of human malignancies, including in EC (10), gastric cancer (11), breast cancer (12) and osteosarcoma (13). A variety of studies have demonstrated that miRNA expression is dysregulated in EC, which implicates EC formation and progression by influencing a wide range of biological processes (1416). An investigation into the roles of dysregulated miRNAs in EC may therefore be useful in the future development of therapeutic targets for patients with this disease.

miR-873 has been demonstrated to be dysregulated in different types of human cancer, serving a crucial role in tumorigenesis and tumor development (1719). However, the expression status and the regulatory role of miR-873 have yet to be elucidated in EC. The current study aimed to detect miR-873 expression in EC and elucidate the biological functions and mechanisms of miR-873 in EC progression.

Materials and methods

Patients and tissue samples

A total of 47 female patients with EC (age range, 45–73 years) who underwent surgery at The First People's Hospital of Kunshan (Suzhou, China) between August 2015 and September 2017 were included in the present study. All patients enrolled in the present study were divided into a miR-873-low (≤0.382) or miR-873-high (>0.382) expression group based on the median miR-873 expression value they exhibited. All patients included in the current study received no adjuvant treatment prior to surgical resection to avoid therapy-associated expression change. Patients that had been treated with preoperative brachytherapy, radiotherapy or chemotherapy were excluded from the current study. All EC tissues and adjacent normal endometrial tissues were frozen in liquid nitrogen and stored at −80°C until subsequent use. Adjacent normal tissues were obtained 2 cm away from EC tissues. All tissues were removed during surgery for the purposes of the present study. The present study was approved by the Ethics Committee of The First People's Hospital of Kunshan (Suzhou, China) and all participating patients or their families provided written informed consent.

Cell culture

A total of four human EC cell lines, including AN3CA, HEC-59, HEC-1B and KLE were obtained from the American Type Culture Collection. EC arises from the endometrium epithelial cells. Therefore, normal human endometrial epithelial primary cells were obtained from PriCells (cat. no. HUM-CELL-0111) and used as controls. Three passages of these cells were used in reverse transcription-quantitative (RT-q)PCR. All EC cell lines and normal human endometrial epithelial primary cells were cultured in Dulbecco's modified Eagle's medium (DMEM) containing 10% FBS and a 1% penicillin-streptomycin solution (all, Gibco; Thermo Fisher Scientific, Inc.). All cultures were maintained at 37°C in a 5% CO2 humidified atmosphere.

Transfection

miR-873 mimics, miRNA negative control mimics (miR-NC), small interfering RNA (siRNA) sequences targeting hepatoma-derived growth factor (HDGF siRNA) and a negative control siRNA (NC siRNA) were all produced by Shanghai Integrated Biotech Solutions Co., Ltd. The miR-873 mimics sequence was 5′-GCAGGAACUUGUGAGUCUCCU-3′, the miR-NC sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′ and the HDGF siRNA sequence was 5′-CCGGCAGAAGGAGUACAAATT-3′ and the NC siRNA was 5′-GCGACGAUCUGCCUAAGAUTT-3′. A HDGF overexpression plasmid (pcDNA3.1-HDGF) and an empty pcDNA3.1 plasmid were acquired from the Chinese Academy of Sciences. For transfection, cells were seeded into six-well plates at a density of 6×105 cells/well, one night prior to transfection. Cells were transfected with miR-873 mimics, miR-NC, HDGF siRNA or NC siRNA (all 100 pmol), and pcDNA3.1-HDGF or pcDNA3.1 (both 4 µg) using Lipofectamine 2000® (Invitrogen; Thermo Fisher Scientific, Inc.) in accordance with manufacturer's protocol. At 48 h post-transfection, RT-qPCR and in vitro invasion assays were performed. The Cell Counting Kit (CCK)-8 assay was carried out after 24 h of transfection. Western blot analysis was conducted to determine protein expression after 72 h of transfection.

RNA isolation and RT-qPCR

TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was used for total RNA isolation from EC tissues, adjacent normal endometrial tissues or transfected EC cells. cDNA was prepared from total RNA using a miScript Reverse Transcription kit (Qiagen GmbH). miR-873 expression was determined using a miScript SYBR Green PCR kit (Qiagen GmbH). U6 small nuclear RNA was used as an internal control. The temperature protocols for qPCR were as follows: 95°C for 2 min, and 40 cycles of 95°C for 10 sec, 55°C for 30 sec and 72°C for 30 sec.

For the detection of HDGF mRNA expression, cDNA production was performed using a PrimeScript RT Reagent kit (Takara Biotechnology Co., Ltd.). qPCR was subsequently performed using a SYBR Premix Ex Taq™ kit (Takara Biotechnology Co., Ltd.). GAPDH served as an endogenous control for HDGF mRNA expression. The temperature protocols for qPCR were as follows: 95°C for 5 min, followed by 40 cycles of 95°C for 30 sec and 65°C for 45 sec.

Relative gene expression was analyzed using the 2−ΔΔCq method (20). The primers were designed as follows: miR-873, 5′-GCAGGAACUUGUGAGUCUCCU-3′ (forward) and 5′-AGGAGACUCACAAGUUCCUGC-3′ (reverse); U6, 5′-GCTTCGGCAGCACATATACTAAAAT-3′ (forward) and 5′-CGCTTCACGAATTTGCGTGTCAT-3′ (reverse); HDGF, 5′-ATCAACAGCCAACAAATACC-3′ (forward) and 5′-TTCTTATCACCGTCACCCT-3′ (reverse); and GAPDH, 5′-CGGAGTCAACGGATTTGGTCGTAT-3′ (forward) and 5′-AGCCTTCTCCATGGTGGTGAAGAC-3′ (reverse).

Cell Counting Kit (CCK)-8 assay

At 24 h after transfection, HEC-59 and HEC-1B cells were collected, suspended in DMEM containing 10% FBS and inoculated at a density of 3,000 cells/well into 96-well plates. Cells were then incubated at 37°C in 5% CO2 for 0, 24, 48 or 72 h. A CCK-8 assay was performed at these indicated time points to evaluate cell proliferation. A total of 10 µl CCK-8 solution (Beyotime Institute of Biotechnology) was added to each well and cells were incubated for a further 2 h at 37°C with 5% CO2. The optical density of each well was detected at a wavelength of 450 nm using a microplate reader (Bio-Rad Laboratories, Inc.).

In vitro invasion assay

HEC-59 and HEC-1B transfected cells were incubated at 37°C for 48 h, collected and resuspended in FBS-free DMEM. A total of 200 µl cell suspension containing 5×104 cells was inoculated into the upper compartment of transwell inserts (Corning Inc.) that were precoated with Matrigel (BD Biosciences). A total of 500 µl DMEM medium supplemented with 10% FBS was added into the lower compartments. Following 24 h of incubation at 37°C with 5% CO2, non-invaded cells on top of the transwell inserts were gently removed by scraping. Invaded cells were fixed with 100% methanol at 37°C for 30 min, stained with 0.5% crystal violet at 37°C for 30 min and washed with PBS (Gibco; Thermo Fisher Scientific, Inc.). Cell invasion was evaluated by counting the number of invaded cells in five individual fields/inserts using an Olympus IX51 inverted microscope (magnification, ×200; Olympus Optical Co., Ltd.).

Bioinformatics analysis

Bioinformatics analysis was performed using TargetScan (www.targetscan.org) and miRDB (http://www.mirdb.org/). miR-873 was revealed to target sites 32–38 of the HDGF 3′-UTR.

Luciferase reporter assay

HDGF 3′-UTR fragments containing the wild-type (wt) and mutant (mut) seed miR-873 regions were amplified by Shanghai Genepharma Co., Ltd. and inserted into the pmirGLO luciferase reporter vector (Promega Corporation). The chemically synthesized luciferase plasmids were named pmirGLO-HDGF-3′-UTR wt and pmirGLO-HDGF-3′-UTR mut, respectively.

HEC-59 and HEC-1B cells were inoculated into 24-well plates at 60–70% confluence one day prior to transfection. Cells were co-transfected with the reporter plasmid and miR-873 mimics or miR-NCs using Lipofectamine 2000®, according to manufacturer's protocol.

Subsequent to a 48-h incubation at 37°C, luciferase activity was determined using a Dual-Luciferase Reporter Assay System (Promega Corporation) according to manufacturer's protocol. Renilla luciferase activity was normalized to that of firefly luciferase activity.

Western blot analysis

Homogenized tissue samples, and transfected HEC-59 and HEC-1B cells were washed with PBS and protein was isolated using a total protein extraction kit (Nanjing KeyGen Biotech Co., Ltd.). Isolated total protein was then subjected to concentration detection using a Bicinchoninic Acid Assay kit (Pierce; Thermo Fisher Scientific, Inc.). After separation by SDS-PAGE on 10% gels, equal quantities of protein (30 µg) were transferred to PVDF membranes (EMD Millipore) and blocked with 5% fat-free milk at room temperature for 2 h that was diluted in Tris-buffered saline and 0.05% Tween-20. Membranes were subsequently incubated overnight at 4°C with mouse anti-human HDGF (1:1,000; cat. no. sc-271344; Santa Cruz Biotechnology, Inc.) or mouse anti-human GAPDH antibodies (1:1,000; cat. no. sc-47724; Santa Cruz Biotechnology, Inc.). Samples were then incubated with horseradish peroxidase-conjugated goat anti-mouse secondary antibodies (1:5,000; cat. no. sc-516102; Santa Cruz Biotechnology, Inc.) at room temperature for 2 h. Protein signals were then visualized using an ECL Protein Detection kit (Pierce; Thermo Fisher Scientific, Inc.). GAPDH was used as a loading control. Quantity One software 4.62 (Bio-Rad Laboratories, Inc.) was utilized for densitometry.

Statistical analysis

All results were presented as the mean ± standard deviation from three independent experiments. The association between miR-873 expression and EC patient clinicopathological variables was evaluated using a χ2 test. Spearman's correlation analysis was used to investigate the association between miR-873 and HDGF mRNA levels in EC tissues. A Student's t-test and one-way ANOVA followed by a Newman Keuls post-hoc test were performed to compare data between groups. All statistical analysis was performed using SPSS software (version 18.0; SPSS, Inc.). P<0.05 was considered to indicate a statistically significant result.

Results

miR-873 is downregulated in EC tissues and cell lines

To assess the effect of miR-873 in EC progression, EC tissues and adjacent normal endometrial tissues were obtained from 47 patients diagnosed with EC. RT-qPCR was performed to detect miR-873 expression, the results of which revealed that miR-873 expression in EC tissues was significantly lower when compared with normal adjacent endometrial tissues (P<0.05; Fig. 1A). The association of miR-873 and the clinicopathological features of patients with EC was also assessed. Patients were divided into a miR-873-low or miR-873-high expression group. As presented in Table I, miR-873 expression was significantly associated with International Federation of Gynecology and Obstetrics (FIGO) stage (P=0.001) and lymph node metastasis (P=0.014) in patients with EC. However, there was no significant association with age (P=0.679), pathology classification (P=0.609) or the depth of myometrial infiltration (P=0.191) in patients with EC. Furthermore, miR-873 expression was determined in four human EC cell lines, including AN3CA, HEC-59, HEC-1B and KLE. Human normal endometrial epithelial cells were used as a control. The results of the current study demonstrated that the expression of miR-873 was lower in all four EC cell lines when compared with normal endometrial epithelial cells (P<0.05; Fig. 1B). These results indicate that miR-873 may be closely associated with EC development.

Table I.

The association between miR-873 expression and clinicopathological features of patients with endometrial cancer.

Table I.

The association between miR-873 expression and clinicopathological features of patients with endometrial cancer.

miR-873 expression

Clinicopathological featuresLowHighP-value
Age (years) 0.679
  <5589
  ≥551614
Pathology classification 0.609
  Well + Moderate1516
  Poor97
Depth of myometrial infiltration 0.191
  <1/2913
  ≥1/21510
FIGO stages 0.001a
  I–II1020
  III–IV143
Lymph node metastasis 0.014a
  Negative1320
  Positive113

a P<0.05. miR, microRNA; FIGO, Federation of Gynecology and Obstetrics.

miR-873 inhibits the proliferation and invasion of EC cells in vitro

HEC-59 and HEC-1B cell lines exhibited a significantly lower expression of miR-873 and were therefore selected for use in subsequent experiments (Fig. 1). To assess the role of miR-873 in EC cells, miR-873 mimics or a miR-NC was transfected into HEC-59 and HEC-1B cells. Transfection with miR-873 mimics significantly increased miR-873 expression in HEC-59 and HEC-1B cells compared with miR-NC transfected cells (P<0.05; Fig. 2A). A CCK-8 assay was subsequently performed to assess the effect of miR-873 in EC cell proliferation. As presented in Fig. 2B, miR-873 upregulation significantly attenuated the proliferation of HEC-59 and HEC-1B cells at both 48 and 72 h (P<0.05). In addition, an in vitro invasion assay revealed that cells transfected with miR-873 mimics were markedly less invasive when compared with the miR-NC group in both cell lines (P<0.05; Fig. 2C). The results indicate that miR-873 may serve anti-proliferative and anti-invasive roles in EC progression.

HDGF is a direct target gene of miR-873 in EC cells

Two algorithms (TargetScan and miRDB) were used to predict potential targets of miR-873. HDGF contains a miR-873 binding site in its 3′-UTR (Fig. 3A) and was therefore selected for further investigation due to this gene also being associated with EC genesis and development (21). A luciferase reporter assay was then performed to validate the potential miR-873 binding site. miR-873 overexpression downregulated the luciferase activity of the plasmid containing the wt miR-873 binding site (P<0.05) but did not effect the plasmid containing the mut site in HEC-59 and HEC-1B cells (Fig. 3B).

HDGF expression was subsequently detected in EC tissues and the association between HDGF expression and miR-873 was assessed. RT-qPCR analysis revealed EC tissues exhibited significantly upregulated HDGF mRNA levels when compared with adjacent normal endometrial tissues (P<0.05; Fig. 3C). Spearman's correlation analysis identified an inverse correlation between miR-873 and HDGF mRNA levels in EC tissues (r=0.5367; P=0.0001; Fig. 3D). Furthermore, transfection of the miR-873 mimics resulted in a significant reduction of HDGF expression in HEC-59 and HEC-1B cells compared with cells transfected with miR-NC at the mRNA (P<0.05; Fig. 3E) and protein (P<0.05; Fig. 3F) level, as determined by RT-qPCR and western blot analysis, respectively. The results indicate that HDGF is a direct target gene of miR-873 in EC cells.

Knockdown of HDGF imitates the tumor-suppressive role of miR-873 in EC cells

Loss-of-function assays were performed to elucidate HDGF functional roles in EC cells. HDGF siRNA and an NC siRNA were chemically synthesized and transfected into HEC-59 and HEC-1B cells. Western blot analysis demonstrated that HDGF protein levels were knocked-down in HEC-59 and HEC-1B cells after HDGF siRNA transfection (P<0.05; Fig. 4A). Functional assays revealed that the inhibition of HDGF led to a significant decrease in the proliferative at both 48 and 72 h (P<0.05; Fig. 4B) and invasive (P<0.05; Fig. 4C) capacities of HEC-59 and HEC-1B cells. The loss-of-function assays demonstrated that HDGF knockdown may imitate the tumor suppressive roles of miR-873 overexpression in EC cells, indicating that HDGF may be a direct downstream target of miR-873 in EC.

HDGF reintroduction inhibits the anti-proliferative and anti-invasive role of miR-873 upregulation in EC cells

Rescue experiments were performed to confirm that the miR-873-induced inhibition of EC cell proliferation and invasion is mediated by HDGF inhibition. HEC-59 and HEC-1B cells were treated with the HDGF overexpression plasmid (pcDNA3.1-HDGF) or an empty pcDNA3.1 plasmid. The results of Western blot analysis indicated that miR-873 expression significantly decreased HDGF protein levels in HEC-59 and HEC-1B cells and that this outcome could be rescued following co-transfection with pcDNA3.1-HDGF (P<0.05; Fig. 5A). CCK-8 and in vitro invasion assays also demonstrated that the restoration of HDGF expression abrogated the suppression of HEC-59 and HEC-1B cell proliferation (P<0.05; Fig. 5B) and invasion (P<0.05; Fig. 5C) caused by miR-873 overexpression at both 48 and 72 h. The results demonstrate that HDGF may serve as a direct target gene of miR-873 and is implicated in miR-873-mediated EC phenotype malignancy.

Discussion

An accumulation of evidence has indicated that abnormal miRNA expression is an indication of different types of cancer, including EC (11,22,23). The dysregulation of miRNAs may serve an important role in the development and progression of EC by regulating a variety of processes, including cell proliferation, apoptosis, cycle, differentiation, metastasis and sensitivity of radiotherapy and chemotherapy (2426). An understanding of the miRNAs associated with EC initiation and progression is crucial for the development of improved therapeutic techniques. Previous studies have demonstrated that numerous miRNAs are associated with EC (2729). In the current study, to the best of our knowledge, expression status, functions and governing molecular mechanisms of miR-873 in EC were detected for the first time.

miR-873 is upregulated in hepatocellular carcinoma and lung cancer (18,19). A high miR-873 level is associated with α-fetoprotein, tumor size, vascular invasion and Edmondson grade in hepatocellular carcinoma (17). A decreased expression of miR-873 has also been demonstrated in colorectal cancer and is strongly associated with tumor stage (30). Patients with colorectal cancer and a low miR-873 expression exhibit a shorter overall survival than patients with a high miR-873 expression (30). The downregulation of miR-873 has also been indicated in breast cancer (31), glioblastoma (32), gastric cancer (33), esophageal cancer (34) and ovarian cancer (35). However, the expression pattern of miR-873 in EC has not yet been elucidated. In the present study, RT-qPCR was performed to assess miR-873 expression in EC tissues and cell lines, the results of which revealed a decreased expression in both. Furthermore, the decreased miR-873 expression was closely associated with FIGO stage and lymph node metastasis of patients with EC. The results demonstrated that miR-873 may be an effective biomarker for the diagnosis of the patients with EC.

Previous studies have demonstrated that miR-873 serves as an oncogene in hepatocellular carcinoma (1820). miR-873 overexpression has been shown to promote hepatocellular carcinoma cell growth and metastasis, and inhibit G1 phase arrest in vitro by directly targeting tumor suppressor in lung cancer 1 gene (17). miR-873 has also been revealed to serve an oncogenic role in the progression of non-small cell lung cancer by directly targeting SRC kinase signaling inhibitor 1 (18) and glioma-associated oncogene homolog 1 (19). In contrast, miR-873 upregulation inhibits the proliferation of colorectal cancer cells by blocking tumor necrosis factor receptor-associated factor 5 and TGF-β activated kinase 1 binding protein 1 (31). In esophageal cancer, miR-873 expression suppresses cell growth and metastasis in vitro through the negative regulation of differentiated embryonic chondrocyte expressed gene 2 (34). miR-873 has also been identified as a tumor suppressor in breast cancer (31), glioblastoma (32), gastric cancer (33) and ovarian cancer (35). However, the detailed functions of miR-873 in EC progression remain elusive. In the present study, results indicated that miR-873 expression attenuated cell proliferation and invasion in vitro. The results demonstrated that miR-873 may be developed as a potential therapeutic target for the patients with EC, but further elucidation is required.

The validation of miR-873 as a direct target gene in EC may provide an improved understanding of miR-873 regulatory mechanisms. In the current study, HDGF was demonstrated to be a direct and functional downstream target of miR-873 in EC. HDGF, which is located on chromosome 1 at the q21-q23 region, is overexpressed in different types of cancer, including colorectal cancer (36), hepatocellular carcinoma (37), cervical cancer (38) and ovarian cancer (39). HDGF is also highly expressed in EC (21). A high HDGF expression is positively associated with FIGO stage (21). Patients with EC and a high HDGF expression exhibit lower overall survival rates when compared with patients exhibiting a low HDGF expression (21). In the current study, the results indicated that the inhibition of HDGF prevents EC cell proliferation and invasion in vitro. The results revealed that miR-873 targeted HDGF and blocked EC progression and development. Silencing HDGF expression using miR-873 mediated targeting may therefore be a valuable therapeutic strategy for the management of patients with EC. However, future studies are required to assess this.

In summary, miR-873 was downregulated in EC tissues and cell lines. A low miR-873 expression was positivity associated with FIGO stage and lymph node metastasis. The ectopic expression of miR-873 inhibited EC cell proliferation and invasion by directly targeting HDGF. However, further research is required to assess the anti-cancer role of miR-873 in EC to provide novel therapeutic techniques for patients with this disease. The current study did not assess the effect of miR-873 in the apoptosis or autophagy of EC cells. Therefore, future studies should be performed to determine this.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Jiangsu Maternal and Child Health Care Research Project (grant no. F201709).

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

QW and WZ designed the research and performed all functional experiments. WZ wrote the manuscript. Both authors read and approved the final draft.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of The First People's Hospital of Kunshan and was performed in accordance with the Declaration of Helsinki and the guidelines of the Ethics Committee of The First People's Hospital of Kunshan. Written informed consent was provided by all patients or their families.

Patient consent for publication

Written informed consent was obtained from patients or the families all patients involved in the present study.

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 

Dizon DS: Treatment options for advanced endometrial carcinoma. Gynecol Oncol. 117:373–381. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Bendifallah S, Ballester M and Darai E: Endometrial cancer: Predictive models and clinical impact. Bull Cancer. 104:1022–1031. 2017.(In French). View Article : Google Scholar : PubMed/NCBI

4 

Fong P and Meng LR: Effect of mTOR inhibitors in nude mice with endometrial carcinoma and variable PTEN expression status. Med Sci Monit Basic Res. 20:146–152. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Vale CL, Tierney J, Bull SJ and Symonds PR: Chemotherapy for advanced, recurrent or metastatic endometrial carcinoma. Cochrane Database Syst Rev. CD0039152012.PubMed/NCBI

6 

Boll D, Verhoeven RH, van der Aa MA, Pauwels P, Karim-Kos HE, Coebergh JW and van Doorn HC: Incidence and survival trends of uncommon corpus uteri malignancies in the Netherlands, 1989–2008. Int J Gynecol Cancer. 22:599–606. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Calin GA and Croce CM: MicroRNA signatures in human cancers. Nat Rev Cancer. 6:857–866. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Lai EC: Micro RNAs are complementary to 3′UTR sequence motifs that mediate negative post-transcriptional regulation. Nat Genet. 30:363–364. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Filipowicz W, Bhattacharyya SN and Sonenberg N: Mechanisms of post-transcriptional regulation by microRNAs: Are the answers in sight? Nat Rev Genet. 9:102–114. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Chen S, Sun KX, Liu BL, Zong ZH and Zhao Y: MicroRNA-505 functions as a tumor suppressor in endometrial cancer by targeting TGF-α. Mol Cancer. 15:112016. View Article : Google Scholar : PubMed/NCBI

11 

Link A and Kupcinskas J: MicroRNAs as non-invasive diagnostic biomarkers for gastric cancer: Current insights and future perspectives. World J Gastroenterol. 24:3313–3329. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Piasecka D, Braun M, Kordek R, Sadej R and Romanska H: MicroRNAs in regulation of triple-negative breast cancer progression. J Cancer Res Clin Oncol. 144:1401–1411. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Kushlinskii NE, Fridman MV and Braga EA: Molecular mechanisms and microRNAs in osteosarcoma pathogenesis. Biochemistry (Mosc). 81:315–328. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Rarani FZ, Borhani F and Rashidi B: Endometrial pinopode biomarkers: Molecules and microRNAs. J Cell Physiol. 233:9145–9158. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Srivastava SK, Ahmad A, Zubair H, Miree O, Singh S, Rocconi RP, Scalici J and Singh AP: MicroRNAs in gynecological cancers: Small molecules with big implications. Cancer Lett. 407:123–138. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Kanekura K, Nishi H, Isaka K and Kuroda M: MicroRNA and gynecologic cancers. J Obstet Gynaecol Res. 42:612–617. 2016. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Gao Y, Xue Q, Wang D, Du M, Zhang Y and Gao S: miR-873 induces lung adenocarcinoma cell proliferation and migration by targeting SRCIN1. Am J Transl Res. 7:2519–2526. 2015.PubMed/NCBI

19 

Jin S, He J, Li J, Guo R, Shu Y and Liu P: MiR-873 inhibition enhances gefitinib resistance in non-small cell lung cancer cells by targeting glioma-associated oncogene homolog 1. Thorac Cancer. 9:1262–1270. 2018. 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(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Wang L, Jiang Q, Hua S, Zhao M, Wu Q, Fu Q, Fang W and Guo S: High nuclear expression of HDGF correlates with disease progression and poor prognosis in human endometrial carcinoma. Dis Markers. 2014:2987952014. View Article : Google Scholar : PubMed/NCBI

22 

Yanokura M, Banno K, Iida M, Irie H, Umene K, Masuda K, Kobayashi Y, Tominaga E and Aoki D: MicroRNAS in endometrial cancer: Recent advances and potential clinical applications. EXCLI J. 14:190–198. 2015.PubMed/NCBI

23 

Iqbal MA, Arora S, Prakasam G, Calin GA and Syed MA: MicroRNA in lung cancer: Role, mechanisms, pathways and therapeutic relevance. Mol Aspects Med. 2018.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

24 

Xiong H, Chen R, Liu S, Lin Q, Chen H and Jiang Q: MicroRNA-183 induces epithelial-mesenchymal transition and promotes endometrial cancer cell migration and invasion in by targeting CPEB1. J Cell Biochem. 119:8123–8137. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Kontomanolis EN and Koukourakis MI: MicroRNA: The potential regulator of endometrial carcinogenesis. Microrna. 4:18–25. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Zhuo Z and Yu H: miR-205 inhibits cell growth by targeting AKT-mTOR signaling in progesterone-resistant endometrial cancer Ishikawa cells. Oncotarget. 8:28042–28051. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Zhang W, Chen JH, Shan T, Aguilera-Barrantes I, Wang LS, Huang TH, Rader JS, Sheng X and Huang YW: miR-137 is a tumor suppressor in endometrial cancer and is repressed by DNA hypermethylation. Lab Invest. 98:1397–1407. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Liu J, Li C, Jiang Y, Wan Y, Zhou S and Cheng W: Tumor-suppressor role of miR-139-5p in endometrial cancer. Cancer Cell Int. 18:512018. View Article : Google Scholar : PubMed/NCBI

29 

Shi W, Wang X, Ruan L, Fu J, Liu F and Qu J: MiR-200a promotes epithelial-mesenchymal transition of endometrial cancer cells by negatively regulating FOXA2 expression. Pharmazie. 72:694–699. 2017.PubMed/NCBI

30 

Gong H, Fang L, Li Y, Du J, Zhou B, Wang X, Zhou H, Gao L, Wang K and Zhang J: miR873 inhibits colorectal cancer cell proliferation by targeting TRAF5 and TAB1. Oncol Rep. 39:1090–1098. 2018.PubMed/NCBI

31 

Cui J, Yang Y, Li H, Leng Y, Qian K, Huang Q, Zhang C, Lu Z, Chen J, Sun T, et al: MiR-873 regulates ERalpha transcriptional activity and tamoxifen resistance via targeting CDK3 in breast cancer cells. Oncogene. 34:40182015. View Article : Google Scholar : PubMed/NCBI

32 

Wang RJ, Li JW, Bao BH, Wu HC, Du ZH, Su JL, Zhang MH and Liang HQ: MicroRNA-873 (miRNA-873) inhibits glioblastoma tumorigenesis and metastasis by suppressing the expression of IGF2BP1. J Biol Chem. 290:8938–8948. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Cao D, Yu T and Ou X: MiR-873-5P controls gastric cancer progression by targeting hedgehog-GLI signaling. Pharmazie. 71:603–606. 2016.PubMed/NCBI

34 

Liang Y, Zhang P, Li S, Li H, Song S and Lu B: MicroRNA-873 acts as a tumor suppressor in esophageal cancer by inhibiting differentiated embryonic chondrocyte expressed gene 2. Biomed Pharmacother. 105:582–589. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Wu DD, Li XS, Meng XN, Yan J and Zong ZH: MicroRNA-873 mediates multidrug resistance in ovarian cancer cells by targeting ABCB1. Tumour Biol. 37:10499–10506. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Liao F, Liu M, Lv L and Dong W: Hepatoma-derived growth factor promotes the resistance to anti-tumor effects of nordihydroguaiaretic acid in colorectal cancer cells. Eur J Pharmacol. 645:55–62. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Chen SC, Hu TH, Huang CC, Kung ML, Chu TH, Yi LN, Huang ST, Chan HH, Chuang JH and Liu LF: Hepatoma-derived growth factor/nucleolin axis as a novel oncogenic pathway in liver carcinogenesis. Oncotarget. 6:16253–16270. 2015.PubMed/NCBI

38 

Tsai CC, Huang SC, Tai MH, Chien CC, Huang CC and Hsu YC: Hepatoma-derived growth factor upregulation is correlated with prognostic factors of early-stage cervical adenocarcinoma. Int J Mol Sci. 15:21492–21504. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Liu XJ, Liu WL, Yang FM, Yang XQ and Lu XF: Hepatoma-derived growth factor predicts unfavorable prognosis of epithelial ovarian cancer. Onco Targets Ther. 8:2101–2109. 2015.PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 18 Issue 2

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
Wang Q and Wang Q: MicroRNA‑873 inhibits the proliferation and invasion of endometrial cancer cells by directly targeting hepatoma‑derived growth factor Retraction in /10.3892/etm.2022.11475. Exp Ther Med 18: 1291-1298, 2019
APA
Wang, Q., & Wang, Q. (2019). MicroRNA‑873 inhibits the proliferation and invasion of endometrial cancer cells by directly targeting hepatoma‑derived growth factor Retraction in /10.3892/etm.2022.11475. Experimental and Therapeutic Medicine, 18, 1291-1298. https://doi.org/10.3892/etm.2019.7713
MLA
Wang, Q., Zhu, W."MicroRNA‑873 inhibits the proliferation and invasion of endometrial cancer cells by directly targeting hepatoma‑derived growth factor Retraction in /10.3892/etm.2022.11475". Experimental and Therapeutic Medicine 18.2 (2019): 1291-1298.
Chicago
Wang, Q., Zhu, W."MicroRNA‑873 inhibits the proliferation and invasion of endometrial cancer cells by directly targeting hepatoma‑derived growth factor Retraction in /10.3892/etm.2022.11475". Experimental and Therapeutic Medicine 18, no. 2 (2019): 1291-1298. https://doi.org/10.3892/etm.2019.7713