Silencing the long noncoding RNA, TINCR, a molecular sponge of miR‑335, inhibits the malignant phenotype of epithelial ovarian cancer via FGF2 suppression

Retraction in: /10.3892/ijo.2022.5411

  • Authors:
    • Rui Li
    • Yue Wang
    • Yuexun Xu
    • Xiaoli He
    • Yali Li
  • View Affiliations

  • Published online on: September 10, 2019     https://doi.org/10.3892/ijo.2019.4875
  • Pages: 1110-1124
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Aberrant terminal differentiation‑induced noncoding RNA (TINCR) expression has been identified in multiple human cancer types and is functionally significant in cancer progression. However, to the best of our knowledge, no reported studies have investigated the expression pattern and precise role of TINCR in epithelial ovarian cancer (EOC). Here, TINCR expression levels in EOC tissues and cell lines were determined by reverse transcription‑quantitative polymerase chain reaction. Cell Counting Kit‑8 assays, flow cytometric analysis, Transwell migration and invasion assays, and in vivo xenograft experiments were performed to determine the influence of TINCR on the malignant phenotype of EOC cells in vitro and in vivo. The molecular mechanisms associated with the tumor‑promoting roles of TINCR during EOC progression were elucidated using a series of experiments. TINCR expression was higher in EOC tissues and cell lines compared with normal cells. An analysis of the association between TINCR expression and clinicopathological characteristics showed that increased TINCR expression was closely related to tumor size, FIGO stage, and lymphatic metastasis. In addition, the overall survival rates of EOC patients with high TINCR expression levels were lower than in those with low TINCR expression levels. Functional experiments showed that TINCR deficiency attenuated the proliferation, migration, and invasion of EOC cells in vitro and hindered EOC tumor growth in vivo. In addition, EOC cell apoptosis increased after TINCR knockdown. Mechanistically, TINCR was shown to function as a sponge of microRNA‑335 (miR‑335) in EOC cells, thereby regulating fibroblast growth factor 2 (FGF2) expression. miR‑335 inhibition partially counteracted the effect of TINCR knockdown on the aggressive behavior of EOC cells. This study showed, for the first time to the best of our knowledge, that silencing TINCR, which interacts with miR‑335, inhibited EOC progression in vitro and in vivo by decreasing FGF2 expression. Hence, this lncRNA could be a potential prognostic biomarker and effective target for therapeutic intervention in EOC.

Introduction

Ovarian cancer, the most lethal gynecological malignancy, ranks as the third leading cause of cancer-associated mortalities among women (1). Every year, ~220,000 females are diagnosed with ovarian cancer and 140,000 deaths are linked to ovarian cancer globally (2). Epithelial ovarian cancer (EOC), the most common type of ovarian cancer, accounts for ~90% of all ovarian cancer cases (3). Over the last few decades, there have been major advancements in therapeutic techniques, including surgical resection and chemotherapeutic and radiotherapeutic therapy. Unfortunately, the clinical outcomes of patients with EOC are still unsatisfactory, with a 5-year survival rate of <50% (4,5). Multiple risk factors have been shown to be responsible for the formation and progression of EOC, but the detailed molecular mechanisms underlying these phenomena remain largely unexplored, which is another major factor contributing to its unsatisfactory prognosis (6,7). Therefore, an in-depth understanding of the mechanisms underlying the aggressive behavior of EOC is urgently required for the development of novel clinical therapeutic methods.

An increasing number of studies have indicated that long noncoding RNAs (lncRNAs) serve important roles in tumorigenesis (8-10). LncRNAs, a group of endogenous non-protein-coding RNAs that are >200 nucleotides in length, were first identified from sequencing and microarray analyses of the whole genome and transcriptome (11). Accumulating evidence suggests that lncRNAs are dysregulated in nearly all types of human cancer and they significantly influence a variety pathophysiological processes, including innate immunity, metabolism, and carcinogenesis (12-14). Numerous lncRNAs dysregulated in EOC have been widely acknowledged in recent years (15-17). For instance, lncRNAs SNHG15 (18), JPX (19), and LINC01118 (20) are upregulated in EOC, and serve tumor-promoting roles during cancer progression. On the contrary, CASC2 (21), XIST (22) and CPS1-IT1 (23) are expressed at low levels in EOC, and inhibit the generation of malignant phenotypes.

lncRNAs have been implicated in the pathogenesis of EOC via interactions with proteins (24), microRNAs (miRNAs/miRs) (25-28), or mRNAs (29,30). Accordingly, therapies that target lncRNAs may be attractive strategies for treating patients with EOC.

Aberrant terminal differentiation-induced noncoding RNA (TINCR) expression has been identified in multiple human cancer types, and its aberrant expression has been shown to have effects on cancer progression (31-36). However, to the best of our knowledge, no reported studies have investigated the expression patterns and precise role of TINCR in EOC. Therefore, in this study, we analyzed TINCR expression in EOC and evaluated the prognostic value of TINCR in patients with EOC. In addition, the biological functions of TINCR with regards to the malignant phenotypes of EOC and the underlying mechanisms, were explored in detail.

Materials and methods

Patients and tissue specimens

In total, 53 pairs of EOC tissues and their adjacent normal tissues were collected from patients (age range, 42-71 years) at The People's Hospital of Zhengzhou University between June 2011 and February 2013. Immediately after surgical resection, all tissue specimens were snap frozen in liquid nitrogen and then stored at -80°C until further use. EOC patients were followed-up for ≤60 months. EOC patients who had been treated with chemotherapy or radiotherapy prior to surgical resection were excluded from the study. The International Federation of Gynecology and Obstetrics classification (25) was used to analyze the stage of disease. The current study was approved by the Ethics Committee of The People's Hospital of Zhengzhou University and was carried out in accordance with the Declaration of Helsinki. Written informed consent was provided by all the enrolled patients before their participation in the study.

Cell culture

The human EOC cell lines, ES-2, CAOV-3, OVCAR3 and SKOV3, were purchased from the Cell Bank of Type Culture Collection, Chinese Academy of Science. A normal human ovarian epithelial cell line, (NOEC), was obtained from the ScienCell Research Laboratories (cat. no. 7310). All cells were cultured in Dulbecco's Modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc.) containing 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin (Sigma-Aldrich; Merck KGaA), and 100 mg/ml streptomycin (Sigma-Aldrich; Merck KGaA). Cell cultures were maintained at 37°C in a humidified atmosphere under 5% CO2.

Transfection assays

Small interfering RNAs (siRNA) against TINCR (si-TINCR) and a nontargeting control siRNA (si-NC) were chemically synthesized by Shanghai GenePharma Co., Ltd. The si-TINCR sequence was 5′-AAT ACC TGC TAC TTC ATG C-3′ and the si-NC sequence was 5′-UUC UCC GAA CGU GUC ACG UTT-3′. miR-335 mimics, negative control (NC) miRNA mimics (miR-NC), an miR-335 inhibitor, and an NC inhibitor were obtained from Guangzhou Ribobio Co., Ltd. The miR-335 mimics sequence was 5′-UCA AGA GCA AUA ACG AAA AAU GU-3′ and the miR-NC sequence was 5′-UUG UAC UAC ACA AAA GUA CUG-3′. The miR-335 inhibitor sequence was 5′-AGU UCU CGU UAU UGC UUU UUA CA-3′ and the NC inhibitor sequence was 5′-ACU ACU GAG UGA CAG UAG A-3′. Overexpression of fibroblast growth factor (FGF2) was achieved using the FGF2 overexpression plasmid, pcDNA3.1-FGF2 (pc-FGF2; GeneCopoeia Inc.). The empty pcDNA3.1 plasmid was used as a control for pc-FGF2 transfection. Cells were plated into 6-well plates at a density of 5×105 cells per well. Cell transfection was performed with Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Approximately 6 h after transfection, the culture medium was replaced with fresh DMEM supplemented with 10% FBS.

RNA extraction and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted using a high-purity total RNA extraction kit (BioTeke Corporation) and then reverse transcribed using a miScript Reverse Transcription kit (Qiagen GmbH), according to the manufacturers' protocols. cDNA samples were then used for measuring miR-335 expression using a miScript SYBR Green PCR kit (Qiagen GmbH). The thermocycling conditions for qPCR were as follows: 95°C for 2 min, 95°C for 10 sec, 55°C for 30 sec and 72°C for 30 sec, for 40 cycles. To measure TINCR and FGF2 mRNA expression, cDNA was synthesized using a PrimeScript first-strand cDNA synthesis kit (Takara Bio, Inc.) and was then subjected to qPCR using a SYBR Premix ExTaq kit (Takara Biotechnology Co.). The thermocycling 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. The expression of miR-335 was normalized to small nuclear U6 RNA expression, while glyceraldehyde phosphate dehydrogenase (GAPDH) was used as the internal control for TINCR and FGF2 mRNA expression. All reactions were performed on the Applied Biosystems 7500 real-time PCR system (Thermo Fisher Scientific, Inc.). Relative gene expression was calculated using the 2−ΔΔCq method (37).

The primers were designed as follows: miR-335, 5′-AGC CGT CAA GAG CAA TAA CGA A-3′ (forward) and 5′-GTG CAG GGT CCG AGG T-3′ (reverse); U6, 5′-GCT TCG GCA GCA CAT ATA CTA AAA T-3′ (forward) and 5′-CGC TTC ACG AAT TTG CGT GTC AT-3′ (reverse); TINCR, 5′-TGT GGC CCA AAC TCA GGG ATA CAT-3′ (forward) and 5′-AGA TGA CAG TGG CTG GAG TTG TCA-3′ (reverse); FGF2, 5′-AGA AGA GCG ACC CTC ACA TCA-3′ (forward) and 5′-CGG TTA GCA CAC ACT CCT TTG-3′ (reverse); and GAPDH, 5′-CAT GTT CGT CAT GGG TGT GAA CCA-3′ (forward) and 5′-AGT GAT GGC ATG GAC TGT GGT CAT-3′ (reverse).

Cell Counting Kit-8 (CCK-8) assays

Transfected cells were collected after 24 h of incubation and suspended in complete culture medium. A total of 100 µl of each suspension containing 2,000 cells was seeded into 96-well plates. Cell proliferation was evaluated at four time points (0, 24, 48 and 72 h after incubation) using the CCK-8 assay (Dojindo Molecular Technologies, Inc.). For this assay, 10 µl of CCK-8 solution was added to the cells, which were then incubated at 37°C for an additional 2 h. The absorbance of the samples at 450 nm was measured using a VarioskanTM LUX microplate reader (Thermo Fisher Scientific, Inc.).

Analysis of apoptosis by flow cytometry

The rate of apoptosis was determined using an Annexin V-fluorescein isothiocyanate apoptosis detection kit (BioLegend, Inc.), according to the manufacturer's protocols. After 48 h of culture, transfected cells were collected and washed three times with ice-cold phosphate buffer solution (PBS; Gibco; Thermo Fisher Scientific, Inc.). Cells were then double-stained with 5 µl of Annexin V and 5 µl of propidium iodide, diluted in 100 µl of binding buffer include in the kit. Following incubation for 30 min in the dark, flow cytometry (FACScan; BD Biosciences) was performed to determine the apoptotic state of cells.

Transwell migration and invasion assays

At 48 h post-transfection, cells were washed three times with PBS and suspended in FBS-free DMEM. In total, 200 µl of cell suspension containing 5×104 transfected cells was plated into the upper compartments of Transwell inserts (8 µM pore size; Costar; Corning Inc.) that were coated with Matrigel (BD Biosciences). The bottom compartments were covered with 500 µl of DMEM containing 20% FBS (Gibco; Thermo Fisher Scientific, Inc) as the chemoattractant. Following incubation for 24 h at 37°C, the non-invading cells in the upper compartment were gently removed with a cotton swab, whereas the invading cells were fixed in 4% paraformaldehyde at room temperature for 30 min and stained with 0.5% crystal violet at room temperature for 30 min. Invasiveness was assessed by counting the average number of invading cells in six randomly selected fields of each insert under an IX83 inverted microscope (×200 magnification; Olympus Corporation). Experimental steps of the Transwell migration assay were similar to those of the invasion assay, except that the Transwell inserts were not coated with Matrigel.

In vivo xenograft experiments

All animal experiments were approved by the Animal Care and Use Committee of The People's Hospital of Zhengzhou University. CAOV-3 cells transfected with si-TINCR or si-NC were subcutaneously injected into nude mice (Shanghai SLAC Laboratory Animal Co. Ltd., Shanghai, China). The width and length of the tumor xenografts were recorded every 4 days for 4 weeks. Tumor volume was measured using the formula: Tumor volume=Length × (width)2/2. At the end of the experiment, all nude mice were sacrificed and tumor xenografts were resected and analyzed.

RNA immunoprecipitation (RIP) assay

RIP assays were performed to examine the interaction between TINCR and miR-335, using a Magna RIP RNA-Binding Protein Immunoprecipitation Kit (EMD Millipore), according to the manufacturer's instructions. Cell lysates were prepared (300 × g; 4°C; 5 min) and incubated with RIP immunoprecipitation buffer containing magnetic beads conjugated with human anti-Argonaute 2 (Ago2) antibodies and normal immunoglobulin G (IgG; cat. no. 03-110; 1:5,000; EMD Millipore). Precipitated RNA was extracted and subjected to RT-qPCR analysis as aforementioned to determine the expression levels of TINCR and miR-335. Antibodies against Ago2 and IgG were purchased from Abcam.

Bioinformatics analysis and luciferase reporter assays

StarBase v3.0 (http://starbase.sysu.edu.cn/) was used to predict binding sites between TINCR and miR-335. The potential target genes of miR-335 were predicted using TargetScan (Release 7.2: March 2018; http://www.targetscan.org) and microRNA.org (August 2010 Release Last Update: 2010-11-01; http://www.microrna.org). FGF2 was found to be a putative target of miR-335.

Fragments of TINCR containing the predicted wild-type (wt) and mutant (mut) miR-335-binding sites were amplified by Shanghai GenePharma Co., Ltd. and cloned into pmirGLO reporter vectors (Promega Corporation) to generate the TINCR-wt and TINCR-mut plasmids, respectively. FGF2-wt and FGF2-mut reporter plasmids were constructed using the same approach. For reporter assays, cells were seeded into 24-well plates at a density of 8×105 cells per well, 1 day before transfection. The generated luciferase reporter plasmids, along with the miR-335 mimics or miR-NC, were transfected into cells using Lipofectamine 2000. Transfected cells were collected after 48 h of transfection and subjected to a dual luciferase reporter assay (Promega Corporation) to measure luciferase activity. Firefly luciferase activity was normalized to Renilla luciferase activity.

Western blotting

Proteins were isolated from tissues or cells using radioimmunoprecipitation assay (RIPA) lysis buffer (Beyotime Institute of Biotechnology). A Pierce Bicinchoninic Acid Protein Assay Kit (Thermo Fisher Scientific, Inc.) was used to measure total protein concentration. Equal amounts of protein (30 µg) were loaded and separated by 10% SDS-PAGE. The protein bands were then transferred onto polyvinylidene difluoride membranes (Beyotime Institute of Biotechnology). Membranes were then blocked with 10% skim milk, diluted in Tris-buffered saline with Tween (TBST), at room temperature for 2 h, followed by an overnight incubation with primary antibodies against FGF2 (ab208687; 1:1,000 dilution; Abcam) or GAPDH (ab181603; 1:1,000 dilution; Abcam). Membranes were then washed three times with TBST and incubated for 2 h at room temperature with goat anti-rabbit horseradish peroxidase-conjugated secondary antibodies (ab6721; 1:5,000 dilution; Abcam). Finally, protein signals were visualized using Pierce™ ECL Western Blotting Substrate (Pierce; Thermo Fisher Scientific, Inc.), and analyzed with Quantity One software version 4.62 (Bio-Rad Laboratories, Inc.).

Statistical analysis

All results are expressed as the mean ± standard deviation from at least three independent experiments. SPSS 13.0 software (SPSS, Inc.) was used for all statistical analyses. The association between TINCR expression and the clinicopathological characteristics of patients with EOC was evaluated by χ2 test. Comparisons between two groups were examined using a two-tailed Student's t-test, while one-way analysis of variance followed by a Dunnett's post-hoc test was used to determine differences among multiple groups. All patients with EOC were divided into either the TINCR-low (n=27) or TINCR-high (n=26) groups according to the median value of TINCR expression in EOC tissues. Overall survival rates were calculated using the Kaplan-Meier method and were analyzed with a log-rank test. The overall survival rates were analyzed during the time period between June 2011 and February 2018. In total, 9 and 13 deaths occurred in the TINCR-low and TINCR-high groups, respectively. The correlation between TINCR, miR-335, and FGF2 mRNA expression in the same EOC tissues was evaluated by Spearman's correlation analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

TINCR is upregulated in EOC tissues and cell lines

To explore the potential role of TINCR in the development of EOC, its expression pattern was investigated in 53 pairs of EOC tissues and adjacent normal tissues. Interestingly, RT-qPCR data revealed that TINCR was overexpressed in EOC tissues, compared with in adjacent normal tissues (P<0.05; Fig. 1A). In addition, further analysis of TINCR expression was performed in the human EOC cell lines, ES-2, CAOV-3, OVCAR3 and SKOV3. The normal human ovarian epithelial cell line, NOEC, served as a control. TINCR expression levels were upregulated in all examined EOC cell lines, compared with in the control cell line, NOEC (P<0.05; Fig. 1B). These results suggested that the upregulation of TINCR may be associated with the malignancy of EOC.

TINCR upregulation is closely associated with poor prognosis in EOC patients

Next, we determined the clinical value of TINCR in patients with EOC. According to the median level of TINCR expression in EOC tissues, all patients with EOC were divided into either the TINCR-low (n=27) or TINCR-high (n=26) groups. As presented in Table I, high levels of TINCR expression exhibited a significant association with tumor size (P=0.040), FIGO stage (P=0.037), and lymphatic metastasis (P=0.016). In addition, EOC patients with high TINCR expression levels exhibited shorter overall survival times than patients with low TINCR expression levels (P=0.0063; Fig. 1C). Thus, these results suggested that increased TINCR expression indicated a poor prognosis of EOC patients.

Table I

Association of TINCR expression with clinicopathological parameters in EOC patients.

Table I

Association of TINCR expression with clinicopathological parameters in EOC patients.

ParameterTINCR expression
P-value
High (n=27)Low (n=26)
Age (years)0.339
 <601114
 ≥601612
Tumor size (cm)0.040a
 <2916
 ≥21810
Differentiated degree0.477
 G11310
 G2 + G31416
FIGO stage0.037a
 I-II1118
 III-IV168
Lymphatic metastasis0.016a
 No1220
 Yes156

a P<0.05. G1, well differentiated; G2, moderately differentiated; G3, poorly differentiated; TINCR, terminal differentiation-induced noncoding RNA.

Silencing TINCR expression inhibited EOC cell proliferation, migration, and invasion, but promoted EOC cell apoptosis in vitro

To explore the specific roles of TINCR in the progression of EOC, the CAOV-3 and SKOV3 cell lines, which exhibited relatively high TINCR expression levels among the four EOC cell lines tested, were selected for functional experiments and transfected with si-TINCR or si-NC. RT-qPCR analysis confirmed efficient TINCR silencing in CAOV-3 and SKOV3 cells after transfection with si-TINCR (P<0.05; Fig. 2A).

A CCK-8 assay was performed to evaluate the influence of TINCR knockdown on EOC cell proliferation. Absorbance values were significantly lower in si-TINCR-transfected CAOV-3 and SKOV3 cells, compared with cells transfected with si-NC (P<0.05; Fig. 2B), suggesting that TINCR silencing decreased the proliferation of EOC cells. Furthermore, flow cytometric analysis showed that the knockdown of TINCR significantly promoted the apoptosis of CAOV-3 and SKOV3 cells compared with the control (P<0.05; Fig. 2C). Furthermore, the migration and invasion of CAOV-3 and SKOV3 cells after si-TINCR or si-NC transfection was measured using Transwell migration and invasion assays. Knockdown of TINCR was found to significantly suppress the migratory (P<0.05; Fig. 2D) and invasive (P<0.05; Fig. 2E) abilities of CAOV-3 and SKOV3 cells compared with the control. These results demonstrated that TINCR may play tumor-promoting roles in the growth and metastasis of EOC cells in vitro.

TINCR acts as a competing endogenous RNA for miR-355 in EOC cells

It is well documented that lncRNAs serve as molecular sponges by interacting with miRNAs (38). To understand the mechanisms underlying the role of TINCR in regulating EOC progression, bioinformatics analysis was performed to search for miRNAs with the potential for complementary base pairing with TINCR. miR-335 (Fig. 3A) was found to be a putative target of TINCR, based on the presence of a putative binding site for miR-335 in TINCR. miR-335 was selected for further experimental identification because that this miRNA exerts important roles in the malignancy of EOC (39-41). To confirm this hypothesis, a luciferase reporter assay was conducted to determine whether TINCR could interact with miR-335 in EOC cells. These results showed that, in CAOV-3 and SKOV3 cells, the transfection of miR-335 mimics significantly reduced the luciferase activity of TINCR-wt compared with the corresponding control (P<0.05), whereas the luciferase activity of TINCR-mut was unaffected after miR-335 overexpression (Fig. 3B). In the RIP assay, TINCR and miR-335 were significantly more abundant in Ago2-precipitated pellets than in IgG-precipitated pellets (P<0.05; Fig. 3C), indicating that miR-335 is a TINCR-targeting miRNA. Furthermore, RT-qPCR analysis indicated that the knockdown of TINCR led to a significant increase in the expression of miR-335 in CAOV-3 and SKOV3 cells compared with the control (P<0.05; Fig. 3D). TINCR expression was significantly suppressed in CAOV-3 and SKOV3 cells transfected with miR-335 mimics compared with the control (P<0.05; Fig. 3E). To further elucidate the association between TINCR and miR-335 expression, we measured miR-335 expression in EOC tissues and cell lines using RT-qPCR. miR-335 expression was found to be significantly lower in EOC tissues (P<0.05; Fig. 3F) and cell lines (P<0.05; Fig. 3G) compared with adjacent normal tissues and NOEC, respectively. Of note, a significant negative correlation was observed between the expression levels of miR-335 and TINCR in the same EOC tissues (R2=0.4676, P<0.0001; Fig. 3H). These results demonstrated that miR-355 was sponged by TINCR in EOC.

miR-335 exerts an inhibitory effect on the growth and metastasis of EOC cells in vitro. Having demonstrated that miR-335 was sponged by TINCR in EOC, we then explored the role of miR-335 in the malignant phenotype of EOC cells. miR-335 mimics were transfected into CAOV-3 and SKOV3 cells. RT-qPCR analysis showed that miR-335 was significantly upregulated in CAOV-3 and SKOV3 cells following transfection with miR-335 mimics (P<0.05; Fig. 4A). Using a series of functional assays, we demonstrated that restoring miR-335 expression attenuated CAOV-3 and SKOV3 cell proliferation (P<0.05; Fig. 4B), increased apoptosis (P<0.05; Fig. 4C), and inhibited cell migration (P<0.05; Fig. 4C) and invasion (P<0.05; Fig. 4D) in vitro. These results further supported the notion that TINCR functions as a regulator of EOC progression by sponging miR-335.

FGF2 is a direct target gene of miR-335 in EOC cells

As miRNAs function by regulating the expression of their target genes, the potential target of miR-335 was predicted using bioinformatics analysis. FGF2, which has complementary binding sequences for miR-335 (Fig. 5A), was chosen for further investigation as this gene has been shown to be involved in the aggressive behavior of EOC (42,43). miR-335 overexpression significantly decreased the luciferase activity of the plasmid containing the wt miR-335-binding site in CAOV-3 and SKOV3 cells (P<0.05). However, there were no significant effects on the luciferase activity of the FGF2-mut reporter plasmid (Fig. 5B). In addition, FGF2 mRNA (P<0.05; Fig. 5C) and protein (P<0.05; Fig. 5D) expression levels were significantly downregulated in CAOV-3 and SKOV3 cells after miR-335 overexpression compared with the control, as demonstrated by RT-qPCR and western blotting analyses, respectively. Furthermore, the expression levels of FGF2 mRNA (P<0.05; Fig. 5E) and protein (P<0.05; Fig. 5F) were increased in all four tested EOC cell lines than that in NOEC. In addition, FGF2 mRNA was significantly upregulated in EOC tissues compared with adjacent normal tissues (P<0.05; Fig. 5G). The levels of FGF2 mRNA in EOC tissues exhibited an inverse correlation with miR-335 levels (R2=0.3664, P<0.0001; Fig. 5H). These results provided sufficient evidence indicating FGF2 as a direct target gene of miR-335 in EOC cells.

FGF2 is required for the miR-335-associated malignant phenotype in EOC cells

A series of rescue experiments were performed to determine whether miR-335 has tumor-suppressing effects on EOC cells through the regulation of FGF2. To this end, FGF2 protein expression was restored in miR-335 mimic-transfected CAOV-3 and SKOV3 cells by co-transfecting cells with the FGF2 overexpression plasmid, pc-FGF2 (P<0.05; Fig. 6A). Functional experiments of FGF2 overexpression showed that the proliferation (P<0.05; Fig. 6B), apoptosis (P<0.05; Fig. 6C), migration (P<0.05; Fig. 7A), and invasion (P<0.05; Fig. 7B) of CAOV-3 and SKOV3 cells exhibited opposing effects to those of miR-335 overexpression, Thus, miR-335 may exert its tumor-suppressing effects on EOC progression, at least partly, by decreasing FGF2 expression.

Decreasing TINCR expression inhibits EOC progression by decreasing the sponging of miR-335 and subsequently, decreasing FGF2 expression

Rescue assays were performed to determine whether TINCR knockdown elicited inhibitory effects on EOC cells due to the reduced sponging of miR-335. si-TINCR was co-transfected with an miR-335 inhibitor or an NC inhibitor into CAOV-3 and SKOV3 cells. Transfection of an miR-335 inhibitor significantly silenced the expression of miR-335 in CAOV-3 and SKOV3 cells (P<0.05; Fig. 8A). miR-335 expression was upregulated in CAOV-3 and SKOV3 cells by transfection of si-TINCR, while its expression was significantly decreased in the two cell lines by co-transfection of the miR-335 inhibitor (P<0.05; Fig. 8B). In addition, RT-qPCR and western blot analyses showed that silencing TINCR expression significantly decreased FGF2 expression in CAOV-3 and SKOV3 cells, at both the mRNA (P<0.05; Fig. 8C) and protein (P<0.05; Fig. 8D and E) levels compared with the controls; however, co-transfection of miR-335 inhibitor abrogated the influence of TINCR knockdown on FGF2 expression. Furthermore, functional assays showed that the inhibition of miR-335 significantly abolished the effects of TINCR silencing on the proliferation (P<0.05; Fig. 8F), apoptosis (P<0.05; Fig. 8G), migration (P<0.05; Fig. 8H), and invasion (P<0.05; Fig. 8I) of CAOV-3 and SKOV3 cells in vitro. Collectively, these results suggested that decreasing TINCR expression suppressed the expression of FGF2 by decreasing the sponging of miR-335, i.e., increasing miR-335 expression in EOC cells, resulting in the restriction of EOC progression.

Loss of TINCR hindered EOC tumor growth in vivo

In vivo xenograft experiments were performed to analyze the role of TINCR in tumor growth in vivo. Decreasing the expression of TINCR significantly inhibited the growth of EOC tumors, compared with tumors from mice injected with si-NC-transfected cells (P<0.05; Fig. 9A and B). At the experimental endpoint, tumor xenografts were resected and weighed. The tumor xenograft weight significantly decreased after TINCR knockdown compared with the control (P<0.05; Fig. 9C). In addition, the expression levels of TINCR, miR-335 and FGF2 in the tumor xenografts were determined using RT-qPCR. In tumors from mice injected with si-TINCR-transfected cells, TINCR (P<0.05; Fig. 9D) and FGF2 mRNA (P<0.05; Fig. 9E) expression was significantly downregulated compared with the control, while miR-335 expression (P<0.05; Fig. 9F) was upregulated. Furthermore, western blotting analysis indicated that FGF2 protein expression was significantly reduced in tumor xenografts derived from mice injected with si-TINCR-transfected cells (P<0.05; Fig. 9G). Collectively, these data indicated that the loss of TINCR impaired EOC tumor growth in vivo by regulating the miR-335/FGF2 axis.

Discussion

An increasing number of studies have demonstrated the important regulatory roles of lncRNAs in carcinogenesis and cancer progression (44-46). A variety of lncRNAs are aberrantly expressed in EOC and play dispensable roles in regulating a wide range of biological activities, such as cell proliferation, the cell cycle, apoptosis, metastasis, and epithelial-mesenchymal transition (47-49). Therefore, the identification of the specific roles of lncRNAs in the pathogenesis of EOC may facilitate the development of effective targets for the treatment of EOC patients (50-52). However, only a small percentage of the lncRNAs dysregulated in EOC have been investigated in detail. To the best of our knowledge, our study is the first to investigate the expression of TINCR in EOC, and TINCR was subsequently evaluated for its clinical and prognostic value in patients with EOC. More importantly, the function of lncRNAs in the progression of EOC and the relevant underlying mechanisms were explored using a series of experiments.

TINCR expression is reduced in prostate (31) and colorectal (32,33) cancers. Reduced TINCR expression has been associated with multiple malignant clinical parameters in patients with prostate cancer (31). Prostate cancer patients with low TINCR expression have a poorer prognosis than those with high TINCR expression (31). By contrast, TINCR is upregulated in hepatocellular carcinoma, and high TINCR expression levels are significantly correlated with tumor size, tumor differentiation, TNM stage, and vascular invasion (34). Hepatocellular carcinoma patients with high TINCR expression levels have shorter disease-free survival times and reduced overall survival than those with low TINCR expression levels (34). Increased levels of TINCR expression have also been observed in breast (35) and gastric (36) cancers. However, the expression profile of TINCR in EOC remains unclear. Herein, we found that TINCR was upregulated in EOC, and was associated with tumor size, FIGO stage and lymphatic metastasis. Notably, EOC patients with high levels of TINCR expression had shorter overall survival times than those with low TINCR expression levels. These findings suggested that TINCR may be an effective indicator for predicting the prognosis of patients with EOC.

TINCR exerts inhibitory effects on the pathogenesis of cancer. For instance, TINCR was implicated in the regulation of thyroid hormone receptor interactor 13 expression and therefore, suppresses prostate cancer cell growth and metastasis in vitro (31). TINCR has been shown to inhibit colorectal cancer cell proliferation, migration and invasion in vitro, induce cell apoptosis in vitro, and decrease tumor growth and metastasis in vivo (32,33). These regulatory effects occurred through the regulation of the miR-107/CD36 axis and promotion of EpCAM cleavage (32,33). By contrast, TINCR has been shown to play oncogenic roles in breast cancer and to participate in the regulation of cell proliferation, anchorage-independent growth, apoptosis, migration and invasion in vitro, as well as tumor growth in vivo (35). A study of gastric cancer has indicated that the loss of TINCR expression reduces cell proliferation, induces apoptosis, and hinders tumor growth in vivo, due to the decreased sponging of miR-375 (36). These inconsistent observations prompted our interest in investigating the effect of TINCR on the aggressive behavior of EOC. Our results indicated that TINCR knockdown inhibited the proliferation, migration and invasion of EOC cells in vitro, but promoted their apoptosis. In addition, decreasing TINCR expression impaired EOC tumor growth in vivo. These findings suggested that the targeting of TINCR is a promising therapeutic approach for treating patients with EOC.

The identification of the mechanisms underlying the tumor-promoting effects of TINCR in EOC is important for the development of novel therapeutic targets. Thus far, the lncRNA-miRNA-mRNA pathway is considered the most widespread regulatory molecular mechanism for lncRNA. In the present study, TINCR was shown to function as a molecular sponge of miR-335 in EOC cells, via the suppression of FGF2. miR-335 was previously reported to be expressed at low levels in EOC, and low miR-335 expression levels were associated with shorter overall and relapse-free survival periods (39). Multivariate analyses have confirmed that miR-335 is an independent prognostic factor for poor overall and relapse-free survival (39). miR-335 is closely involved in the malignancy of EOC by inhibiting the survival, migration and invasion of EOC cells, and increasing their sensitivity to cisplatin (40,41). Our findings also confirmed that miR-335 directly targeted FGF2 to inhibit the generation of malignant phenotypes of EOC cells. More importantly, miR-335 knockdown abolished the si-TINCR-mediated suppression of EOC cell proliferation, migration and invasion, and eliminated the pro-apoptotic effects of si-TINCR on EOC cells. Taken together, these results led us to conclude that TINCR regulated the aggressive behavior of EOC cells in vitro and in vivo via the miR-335/FGF2 axis.

FGF2 is a member of the FGF family and revealed to be a prototypic growth factor (53). FGF2 has been reported to be overexpressed in multiple human cancer types, including renal cell carcinoma (54), breast cancer (55), colorectal cancer (56), and lung cancer (57). In EOC, FGF2 expresses at high levels (58), and exert tumor-promoting roles in the oncogenicity of EOC (42,43). Herein, we revealed that FGF2 is directly regulated by the TINCR/miR-335 axis in EOC and is involved in multiple cancer-related pathological behaviors.

This study includes several limitations. First, we demonstrated that the miR-335/FGF2 axis was responsible for the tumor-promoting roles of TINCR in EOC progression; however, other miRNAs may also could be sponged by TINCR. In addition, we did not apply immunohistochemistry to detect the E-cad and Ki-67 in the tumor xenografts; furthermore, TUNEL analysis was not employed to determine tumor tissue apoptosis. As such, we aim resolve these limitations in the future.

In summary, this study demonstrated that, since TINCR acted as an endogenous sponge of miR-335, a decrease in TINCR expression resulted in an increase in miR-335 expression, thereby decreasing FGF2 expression and restricting EOC progression. Our current research provides novel data regarding the mechanisms underlying EOC pathogenesis and may help to identify potential targets for the treatment of EOC.

Funding

Not applicable.

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

YW designed the study. RL and YW performed the RT-qPCR, flow cytometry and in vivo xenograft experiments. YX performed the Transwell migration and invasion assays. XH and YL performed the other experiments. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The current study was approved by the Ethics Committee of The People's Hospital of Zhengzhou University and was carried out in accordance with the Declaration of Helsinki. Written informed consent was provided by all enrolled patients before their participation in the study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2019. CA Cancer J Clin. 69:7–34. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Ledermann JA, Raja FA, Fotopoulou C, Gonzalez-Martin A, Colombo N and Sessa C: Newly diagnosed and relapsed epithelial ovarian carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 24(Suppl 6): vi24–vi32. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Lupia M and Cavallaro U: Ovarian cancer stem cells: Still an elusive entity? Mol Cancer. 16:642017. View Article : Google Scholar : PubMed/NCBI

4 

La Vecchia C: Ovarian cancer: Epidemiology and risk factors. Eur J Cancer Prev. 26:55–62. 2017. View Article : Google Scholar

5 

Candido-dos-Reis FJ, Song H, Goode EL, Cunningham JM, Fridley BL, Larson MC, Alsop K, Dicks E, Harrington P, Ramus SJ, et al: Germline mutation in BRCA1 or BRCA2 and ten-year survival for women diagnosed with epithelial ovarian cancer. Clin Cancer Res. 21:652–657. 2015. View Article : Google Scholar :

6 

Karnezis AN and Cho KR: Preclinical models of ovarian cancer: Pathogenesis, problems, and implications for prevention. Clin Obstet Gynecol. 60:789–800. 2017. View Article : Google Scholar : PubMed/NCBI

7 

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

8 

Luo M, Li Z, Wang W, Zeng Y, Liu Z and Qiu J: Long non-coding RNA H19 increases bladder cancer metastasis by associating with EZH2 and inhibiting E-cadherin expression. Cancer Lett. 333:213–221. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Lai MC, Yang Z, Zhou L, Zhu QQ, Xie HY, Zhang F, Wu LM, Chen LM and Zheng SS: Long non-coding RNA MALAT-1 over-expression predicts tumor recurrence of hepatocellular carcinoma after liver transplantation. Med Oncol. 29:1810–1816. 2012. View Article : Google Scholar

10 

Matouk IJ, Mezan S, Mizrahi A, Ohana P, Abu-Lail R, Fellig Y, Degroot N, Galun E and Hochberg A: The oncofetal H19 RNA connection: Hypoxia, p53 and cancer. Biochim Biophys Acta. 1803:443–451. 2010. View Article : Google Scholar : PubMed/NCBI

11 

ENCODE Project Consortium; Birney E, Stamatoyannopoulos JA, Dutta A, Guigó R, Gingeras TR, Margulies EH, Weng Z, Snyder M, Dermitzakis ET, et al: Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 447:799–816. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Gutschner T and Diederichs S: The hallmarks of cancer: A long non-coding RNA point of view. RNA Biol. 9:703–719. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Zhang X, Gejman R, Mahta A, Zhong Y, Rice KA, Zhou Y, Cheunsuchon P, Louis DN and Klibanski A: Maternally expressed gene 3, an imprinted noncoding RNA gene, is associated with meningioma pathogenesis and progression. Cancer Res. 70:2350–2358. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Yu G, Yao W, Gumireddy K, Li A, Wang J, Xiao W, Chen K, Xiao H, Li H, Tang K, et al: Pseudogene PTENP1 functions as a competing endogenous RNA to suppress clear-cell renal cell carcinoma progression. Mol Cancer Ther. 13:3086–3097. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Chen Y, Du H, Bao L and Liu W: LncRNA PVT1 promotes ovarian cancer progression by silencing miR-214. Cancer Biol Med. 15:238–250. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Yan H, Silva MA, Li H, Zhu L, Li P, Li X, Wang X, Gao J, Wang P and Zhang Z: Long noncoding RNA DQ786243 interacts with miR-506 and promotes progression of ovarian cancer through targeting cAMP responsive element binding protein 1. J Cell Biochem. 119:9764–9780. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Zhang C, Wang M, Shi C, Shi F and Pei C: Long non-coding RNA Linc00312 modulates the sensitivity of ovarian cancer to cisplatin via the Bcl-2/Caspase-3 signaling pathway. Biosci Trends. 12:309–316. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Qu C, Dai C, Guo Y, Qin R and Liu J: Long noncoding RNA SNHG15 serves as an oncogene and predicts poor prognosis in epithelial ovarian cancer. Onco Targets Ther. 12:101–111. 2019. View Article : Google Scholar

19 

Li J, Feng L, Tian C, Tang YL, Tang Y and Hu FQ: Long noncoding RNA-JPX predicts the poor prognosis of ovarian cancer patients and promotes tumor cell proliferation, invasion and migration by the PI3K/Akt/mTOR signaling pathway. Eur Rev Med Pharmacol Sci. 22:8135–8144. 2018.PubMed/NCBI

20 

Shi C and Wang M: LINC01118 modulates paclitaxel resistance of epithelial ovarian cancer by regulating miR-134/ABCC1. Med Sci Monit. 24:8831–8839. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Xue Z, Zhu X and Teng Y: Long noncoding RNA CASC2 inhibits progression and predicts favorable prognosis in epithelial ovarian cancer. Mol Med Rep. 18:5173–5181. 2018.PubMed/NCBI

22 

Wang C, Qi S, Xie C, Li C, Wang P and Liu D: Upregulation of long non-coding RNA XIST has anticancer effects on epithelial ovarian cancer cells through inverse downregulation of hsa-miR-214-3p. J Gynecol Oncol. 29:e992018. View Article : Google Scholar : PubMed/NCBI

23 

Wang YS, Ma LN, Sun JX, Liu N and Wang H: Long non-coding RNA CPS1-IT1 is a positive prognostic factor and inhibits epithelial ovarian cancer tumorigenesis. Eur Rev Med Pharmacol Sci. 21:3169–3175. 2017.PubMed/NCBI

24 

Qin Z, Zheng X and Fang Y: Long noncoding RNA TMPO-AS1 promotes progression of non-small cell lung cancer through regulating its natural antisense transcript TMPO. Biochem Biophys Res Commun. 516:486–493. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Montavon Sartorius C, Mirza U, Schotzau A, Mackay G, Fink D, Hacker NF and Heinzelmann-Schwarz V: Impact of the new FIGO 2013 classification on prognosis of stage I epithelial ovarian cancers. Cancer Manag Res. 10:4709–4718. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Kallen AN, Zhou XB, Xu J, Qiao C, Ma J, Yan L, Lu L, Liu C, Yi JS, Zhang H, et al: The imprinted H19 lncRNA antagonizes let-7 microRNAs. Mol cell. 52:101–112. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Men Y, Fan Y, Shen Y, Lu L and Kallen AN: The steroidogenic acute regulatory protein (StAR) is regulated by the H19/let-7 Axis. Endocrinology. 158:402–409. 2017. View Article : Google Scholar :

28 

Zuckerwise L, Li J, Lu L, Men Y, Geng T, Buhimschi CS, Buhimschi IA, Bukowski R, Guller S, Paidas M and Huang Y: H19 long noncoding RNA alters trophoblast cell migration and invasion by regulating TβR3 in placentae with fetal growth restriction. Oncotarget. 7:38398–38407. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Zheng Y, Lv P, Wang S, Cai Q, Zhang B and Huo F: LncRNA PLAC2 upregulates p53 to induce hepatocellular carcinoma cell apoptosis. Gene. 712:1439442019. View Article : Google Scholar : PubMed/NCBI

30 

Yang H, Fu G, Liu F, Hu C, Lin J, Tan Z, Fu Y, Ji F and Cao M: LncRNA THOR promotes tongue squamous cell carcinomas by stabilizing IGF2BP1 downstream targets. Biochimie. 165:9–18. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Dong L, Ding H, Li Y, Xue D and Liu Y: LncRNA TINCR is associated with clinical progression and serves as tumor suppressive role in prostate cancer. Cancer Manag Res. 10:2799–2807. 2018. View Article : Google Scholar : PubMed/NCBI

32 

Zhang X, Yao J, Shi H, Gao B and Zhang L: LncRNA TINCR/microRNA-107/CD36 regulates cell proliferation and apoptosis in colorectal cancer via PPAR signaling pathway based on bioinformatics analysis. Biol Chem. 400:663–675. 2019. View Article : Google Scholar

33 

Zhang ZY, Lu YX, Zhang ZY, Chang YY, Zheng L, Yuan L, Zhang F, Hu YH, Zhang WJ and Li XN: Loss of TINCR expression promotes proliferation, metastasis through activating EpCAM cleavage in colorectal cancer. Oncotarget. 7:22639–22649. 2016.PubMed/NCBI

34 

Tian F, Xu J, Xue F, Guan E and Xu X: TINCR expression is associated with unfavorable prognosis in patients with hepatocellular carcinoma. Biosci Rep. 37:BSR201703012017. View Article : Google Scholar : PubMed/NCBI

35 

Liu Y, Du Y, Hu X, Zhao L and Xia W: Up-regulation of ceRNA TINCR by SP1 contributes to tumorigenesis in breast cancer. BMC Cancer. 18:3672018. View Article : Google Scholar : PubMed/NCBI

36 

Chen Z, Liu H, Yang H, Gao Y, Zhang G and Hu J: The long noncoding RNA, TINCR, functions as a competing endogenous RNA to regulate PDK1 expression by sponging miR-375 in gastric cancer. Onco Targets Ther. 10:3353–3362. 2017. View Article : Google Scholar :

37 

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

38 

Chan JJ and Tay Y: Noncoding RNA: RNA regulatory networks in cancer. Int J Mol Sci. 19:pii: E13102018. View Article : Google Scholar

39 

Cao J, Cai J, Huang D, Han Q, Chen Y, Yang Q, Yang C, Kuang Y, Li D and Wang Z: miR-335 represents an independent prognostic marker in epithelial ovarian cancer. Am J Clin Pathol. 141:437–442. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Liu R, Guo H and Lu S: MiR-335-5p restores cisplatin sensitivity in ovarian cancer cells through targeting BCL2L2. Cancer Med. 7:4598–4609. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Cao J, Cai J, Huang D, Han Q, Yang Q, Li T, Ding H and Wang Z: miR-335 represents an invasion suppressor gene in ovarian cancer by targeting Bcl-w. Oncol Rep. 30:701–706. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Lau MT, So WK and Leung PC: Fibroblast growth factor 2 induces E-cadherin down-regulation via PI3K/Akt/mTOR and MAPK/ERK signaling in ovarian cancer cells. PLoS One. 8:e590832013. View Article : Google Scholar : PubMed/NCBI

43 

De Cecco L, Marchionni L, Gariboldi M, Reid JF, Lagonigro MS, Caramuta S, Ferrario C, Bussani E, Mezzanzanica D, Turatti F, et al: Gene expression profiling of advanced ovarian cancer: Characterization of a molecular signature involving fibroblast growth factor 2. Oncogene. 23:8171–8183. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Yu WD, Wang H, He QF, Xu Y and Wang XC: Long noncoding RNAs in cancer-immunity cycle. J Cell Physiol. 233:6518–6523. 2018. View Article : Google Scholar : PubMed/NCBI

Vallone C, Rigon G, Gulia C, Baffa A, Votino R, Morosetti G, Zaami S, Briganti V, Catania F, Gaffi M, et al: Non-coding RNAs and endometrial cancer. Genes (Basel). 9:pii: E1872018. View Article : Google Scholar

45 

Chen X, Sun Y, Cai R, Wang G, Shu X and Pang W: Long noncoding RNA: Multiple players in gene expression. BMB Rep. 51:280–289. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Chu ZP, Dai J, Jia LG, Li J, Zhang Y, Zhang ZY and Yan P: Increased expression of long noncoding RNA HMMR-AS1 in epithelial ovarian cancer: An independent prognostic factor. Eur Rev Med Pharmacol Sci. 22:8145–8150. 2018.PubMed/NCBI

47 

Hu X, Li Y, Kong D, Hu L, Liu D and Wu J: Long noncoding RNA CASC9 promotes LIN7A expression via miR-758-3p to facilitate the malignancy of ovarian cancer. J Cell Physiol. 234:10800–10808. 2019. View Article : Google Scholar

48 

Liu X, Wen J, Wang H and Wang Y: Long non-coding RNA LINC00460 promotes epithelial ovarian cancer progression by regulating microRNA-338-3p. Biomed Pharmacother. 108:1022–1028. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Liu S, Liu Y, Lu Q, Zhou X, Chen L and Liang W: The lncRNA TUG1 promotes epithelial ovarian cancer cell proliferation and invasion via the WNT/β-catenin pathway. Onco Targets Ther. 11:6845–6851. 2018. View Article : Google Scholar :

50 

Wang J, Xu W, He Y, Xia Q and Liu S: LncRNA MEG3 impacts proliferation, invasion, and migration of ovarian cancer cells through regulating PTEN. Inflamm Res. 67:927–936. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Gordon MA, Babbs B, Cochrane DR, Bitler BG and Richer JK: The long non-coding RNA MALAT1 promotes ovarian cancer progression by regulating RBFOX2-mediated alternative splicing. Mol Carcinog. 58:196–205. 2019. View Article : Google Scholar

52 

Litwin M, Radwańska A, Paprocka M, Kieda C, Dobosz T, Witkiewicz W and Baczyńska D: The role of FGF2 in migration and tubulogenesis of endothelial progenitor cells in relation to pro-angiogenic growth factor production. Mol Cell Biochem. 410:131–142. 2015. View Article : Google Scholar : PubMed/NCBI

53 

Xu M, Gu M, Zhang K, Zhou J, Wang Z and Da J: miR-203 inhibition of renal cancer cell proliferation, migration and invasion by targeting of FGF2. Diagn Pathol. 10:242015. View Article : Google Scholar : PubMed/NCBI

54 

Sahores A, Figueroa V, May M, Liguori M, Rubstein A, Fuentes C, Jacobsen BM, Elía A, Rojas P, Sequeira GR, et al: Increased high molecular weight FGF2 in endocrine-resistant breast cancer. Horm Cancer. 9:338–348. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Zhang X, Xu J, Jiang T, Liu G, Wang D and Lu Y: MicroRNA-195 suppresses colorectal cancer cells proliferation via targeting FGF2 and regulating Wnt/β-catenin pathway. Am J Cancer Res. 6:2631–2640. 2016.

56 

Deng ZH, Cao HQ, Hu YB, Wen JF and Zhou JH: TRX is up-regulated by fibroblast growth factor-2 in lung carcinoma. APMIS. 119:57–65. 2011. View Article : Google Scholar

57 

Feng QL, Shi HR, Qiao LJ and Zhao J: Expression of hSef and FGF-2 in epithelial ovarian tumor. Zhonghua Zhong Liu Za Zhi. 33:770–774. 2011.In Chinese.

58 

Whitworth MK, Backen AC, Clamp AR, Wilson G, McVey R, Friedl A, Rapraeger AC, David G, McGown A, Slade RJ, et al: Regulation of fibroblast growth factor-2 activity by human ovarian cancer tumor endothelium. Clin Cancer Res. 11:4282–4288. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2019
Volume 55 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li R, Wang Y, Xu Y, He X and Li Y: Silencing the long noncoding RNA, TINCR, a molecular sponge of miR‑335, inhibits the malignant phenotype of epithelial ovarian cancer via FGF2 suppression Retraction in /10.3892/ijo.2022.5411. Int J Oncol 55: 1110-1124, 2019
APA
Li, R., Wang, Y., Xu, Y., He, X., & Li, Y. (2019). Silencing the long noncoding RNA, TINCR, a molecular sponge of miR‑335, inhibits the malignant phenotype of epithelial ovarian cancer via FGF2 suppression Retraction in /10.3892/ijo.2022.5411. International Journal of Oncology, 55, 1110-1124. https://doi.org/10.3892/ijo.2019.4875
MLA
Li, R., Wang, Y., Xu, Y., He, X., Li, Y."Silencing the long noncoding RNA, TINCR, a molecular sponge of miR‑335, inhibits the malignant phenotype of epithelial ovarian cancer via FGF2 suppression Retraction in /10.3892/ijo.2022.5411". International Journal of Oncology 55.5 (2019): 1110-1124.
Chicago
Li, R., Wang, Y., Xu, Y., He, X., Li, Y."Silencing the long noncoding RNA, TINCR, a molecular sponge of miR‑335, inhibits the malignant phenotype of epithelial ovarian cancer via FGF2 suppression Retraction in /10.3892/ijo.2022.5411". International Journal of Oncology 55, no. 5 (2019): 1110-1124. https://doi.org/10.3892/ijo.2019.4875