Open Access

Long noncoding RNA SNHG14 promotes the aggressiveness of retinoblastoma by sponging microRNA‑124 and thereby upregulating STAT3

Retraction in: /10.3892/ijmm.2022.5126

  • Authors:
    • Xiaowen Sun
    • Hui Shen
    • Shubin Liu
    • Jing Gao
    • Shuyan Zhang
  • View Affiliations

  • Published online on: March 20, 2020     https://doi.org/10.3892/ijmm.2020.4547
  • Pages: 1685-1696
  • Copyright: © Sun et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

A long noncoding RNA called small nucleolar RNA host gene 14 (SNHG14) has been validated as a key regulator of cellular processes in multiple types of human cancer. However, to the best of our knowledge, the expression status and specific roles of SNHG14 in retinoblastoma (RB) have not been studied. The aims of the present study were to determine the expression status of SNHG14 in RB, assess the effects of SNHG14 on malignant characteristics of RB cells and investigate the mechanisms of action of SNHG14 in RB. SNHG14 expression levels in RB tissue samples and cell lines were measured by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). Cell proliferation, apoptosis, migration and invasion in vitro, and tumor growth in vivo were quantitated by the Cell Counting Kit‑8 assay, flow cytometry, migration and invasion assays, and mouse tumor xenograft experiments, respectively. The target microRNA (miRNA) of SNHG14 was predicted by bioinformatics analysis and was subsequently validated by a luciferase reporter assay, RNA immunoprecipitation (RIP) assay, RT‑qPCR, and western blot analysis. SNHG14 was identified to be significantly overexpressed in RB tissues and cell lines. SNHG14 overexpression was markedly associated with the intraocular international retinoblastoma classification stage, optic nerve invasion, and differentiation grade among patients with RB. The patients in the SNHG14 high‑expression group exhibited shorter overall survival compared with the SNHG14 low‑expression group. Functional analysis revealed that SNHG14 silencing inhibited cell proliferation, migration and invasion, and increased apoptosis in vitro, and decreased tumor growth in vivo. SNHG14 directly interacted with, and functioned as a competing endogenous RNA (ceRNA) of, miR‑124, consequently upregulating signal transducer and activator of transcription 3 (STAT3). miR‑124 inhibition and STAT3 expression recovery attenuated the effects of the SNHG14 silencing on RB cells. In conclusion, SNHG14 served as a ceRNA to upregulate STAT3 by sponging miR‑124. Therefore, targeting the SNHG14/miR‑124/STAT3 pathway may be an effective therapeutic strategy against RB.

Introduction

Retinoblastoma (RB), a malignant tumor derived from photo-receptor precursor cells, is the most common intraocular human cancer among infants and children (1). It accounts for ~4% of all pediatric human malignant tumors (2). The typical clinical manifestations of RB are leukocoria and strabismus, and its morbidity rate ranges from 1/15,000-1/20,000 among live births worldwide (3). At present, the primary therapeutic techniques for patients with RB are enucleation, laser photocoagulation, chemotherapy and focal therapy (4). The pathogenesis of RB is a complicated process that involves gene mutations, activation of oncogenes, and inactivation of tumor suppressor genes (5-7); however, the detailed mechanism is yet to be determined. Although the primary option for RB management has yielded satisfactory results for the patients, long-term survival is poor among the majority of patients with the RB diagnosed at advanced stages (8). Therefore, further studies on the mechanisms of the genesis and progression of RB are urgently required for identifying early interventions and therapeutic targets.

Long noncoding RNAs (lncRNAs) measure >200 nucleotides long and have no protein-coding ability (9). They can regulate gene expression at epigenetic, transcriptional and post-transcriptional levels (10). LncRNAs have been demonstrated to exert regulatory effects on the initiation and progression of various human cancer types (11-13). Particularly, a number of lncRNAs are dysregulated in RB and serve either tumor-suppressive or oncogenic roles. For example, small nucleolar RNA host gene 16 (14), homeobox A11 antisense RNA (15), and FEZ family zinc finger 1 antisense RNA 1 (16) are overexpressed in RB and promote its aggressive characteristics. By contrast, Pvt1 oncogene (17), metallothionein 1J, pseudogene (18), and H19 imprinted maternally expressed transcript (19) are expressed weakly and inhibit RB carcinogenesis. Considering the important activities of lncRNAs in RB, these polynucleotides may be potential targets for the diagnosis, therapy and prognosis of RB.

MicroRNAs (miRNAs) are a class of noncoding small RNAs that act as gene expression regulators and have been studied extensively in the last decade (20). miRNAs participate in the modulation of gene expression by directly interacting with the 3′-untranslated region of their target mRNAs, thereby inhibiting translation or facilitating mRNA degradation (21). miRNAs are widely expressed in eukaryotes and are involved in a variety of pathological processes, including tumorigenesis and tumor progression (22,23). In recent years, an increasing number of studies revealed that a variety of miRNAs are aberrantly expressed and perform an important function in the malignancy of RB (24-26). Therefore, miRNAs involved in RB carcinogenesis and progression should be explored to facilitate the identification of effective diagnostic biomarkers and therapeutic strategies.

An lncRNA called small nucleolar RNA host gene 14 (SNHG14) has been validated as a key regulator of cellular processes in multiple types of human cancer (27-35). However, to the best of our knowledge, the expression status and detailed roles of SNHG14 in RB have not been determined. The present study first investigated whether SNHG14 is dysregulated in RB by measuring its expression in RB tissue samples and cell lines. Secondly, functional assays were performed to determine the biological activities of SNHG14 in RB cells. Third, the regulatory mechanisms of action of SNHG14 in RB were explored. These analyses identified the involvement of the SNHG14/miR-124/signal transducer and activator of transcription 3 (STAT3) pathway in RB and may have revealed a new mechanism underlying RB carcinogenesis and progression. Among the miRNAs that may interact with SNHG14, miR-124 was selected for validation as it has been demonstrated to be downregulated in RB and to exert tumor-suppressive actions (36).

Materials and methods

Patients and tissue samples

In total, 43 RB tissue samples were obtained from patients (mean age, 11 years; age range, 1-34 years; 21: 22 male: Female ratio) with RB between February 2012 and May 2014 at the People's Hospital of Rizhao. Normal retinas were collected from 11 patients (mean age, 28 years; age range, 17-49 years; 7: 4 male: Female ratio) with globe rupture between April 2012 and August 2016. All the patients were treated with enucleation and had not received preoperative radiotherapy or chemotherapy. Patients that had been treated with radiotherapy, chemotherapy or other anti-cancer therapies were excluded from the study. International Intraocular Retinoblastoma Classification (IIRC) was utilized to classify the severity of RB (37). Following surgical resection, all tissue samples were immediately immersed into liquid nitrogen and stored at −80°C for subsequent analysis. The present study was conducted with the approval of the Clinical Research Ethics Committee of People's Hospital of Rizhao and following the principles of the Declaration of Helsinki. Written informed consent was obtained from all patients or their parents/legal guardians prior to surgical resection.

Cell lines and transient transfection

A total of 3 RB cell lines (Y79, SO-RB50 and Weri-RB-1) and the normal retinal pigmented epithelial ARPE-19 cell line were purchased from the American Type Culture Collection and were maintained at 37°C in a humidified atmosphere containing 5% CO2. The cells were grown in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% of fetal bovine serum (FBS) and 1% of a penicillin-streptomycin solution (all from Gibco; Thermo Fisher Scientific, Inc.).

To induce SNHG14 knockdown, small interfering RNA (siRNA) targeting SNHG14 (si-SNHG14) and its negative control (NC) nonsense sequence (si-NC) were chemically synthesized by Guangzhou RiboBio Co., Ltd. The si-SNHG14 sequence was 5′-GCA CAA UAU CUU UGA ACU A-3′ and the si-NC sequence was 5′-UUC UCC GAA CGU GUC ACG UTT-3′. An miR-124 agomir (agomir-124), NC agomir (agomir-NC), miR-124 antagomir (antagomir-124), and antagomir-NC were purchased from Shanghai GenePharma Co., Ltd. The agomir-124 sequence was 5′-CCG UAA GUG GCG CAC GGA AU-3′ and the agomir-NC sequence was 5′-UUG UAC UAC ACA AAA GUA CUG-3′. The antagomir-124 sequence was 5′-GGC AUU CAC CGC GUG CCU UA-3′ and the antagomir-NC sequence was 5′-CAG UAC UUU UGU GUA GUA CAA-3′. The empty pcDNA3.1 vector and pcDNA3.1 carrying the full-length STAT3 sequence (pcDNA3.1-STAT3, hereafter: Pc-STAT3) were purchased from Shanghai GeneChem Co., Ltd. The aforementioned siRNAs (100 pmol), agomir (50 nM), and/or plasmid (4 µg) were transfected into cells using Lipofectamine 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). Transfected cells were cultured at 7°C in a humidified atmosphere containing 5% CO2. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR), flow cytometry analysis, migration and invasion assays were performed at 48 h post-transfection. A Cell Counting Kit-8 (CCK-8) assay and western blot analysis were conducted at 24 and 72 h after transfection, respectively. The mouse xenograft tumor model was constructed after 24 h culture.

RT-qPCR

TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was used for total RNA isolation from tissues or cells. To determine miR-124 expression, total RNA was reverse-transcribed into complementary DNA using the miScript Reverse Transcription kit (Qiagen GmbH). The temperature protocols for reverse transcription were as follows: 37°C for 60 min, 95°C for 5 min and maintenance at 4°C. Following this step, the complementary DNA was analyzed by qPCR using the miScript SYBR-Green PCR kit (Qiagen GmbH). The thermocycling conditions were as follows: 95°C for 2 min, followed by 95°C for 10 sec, 55°C for 30 sec and 72°C for 30 sec, for 40 cycles. Small nuclear RNA U6 served as the endogenous control for miR-124 expression. To examine SNHG14 expression, the PrimeScript RT Reagent kit (Takara Biotechnology Co., Ltd.) and the SYBR Premix Ex Taq™ kit (Takara Biotechnology Co., Ltd.) were utilized for reverse transcription and qPCR, respectively. The temperature protocols for reverse transcription were as follows: 37°C for 15 min and 85°C for 5 sec, and the thermocycling conditions for the qPCR step were as follows: 5 min at 95°C, followed by 40 cycles of 95°C for 30 sec and 65°C for 45 sec. SNHG14 expression was normalized to that of GAPDH. All reactions were carried out on the Applied Biosystems 7500 real-time PCR system (Thermo Fisher Scientific, Inc.). Relative gene expression was analyzed using the 2−ΔΔCq method (38).

The primers were designed as follows: miR-124 forward, 5′-GCT AAG GCA CGC GGT G-3′; miR-124 reverse, 5′-GTG CAG GGT CCG AGG T-3′; U6 forward, 5′-CTC GCT TCG GCA GCA CAT ATA CT-3′; U6 reverse, 5′-ACG CTT CAC GAA TTT GCG TGT C-3′; SNHG14 forward, 5′-GGG TGT TTA CGT AGA CCA GAA CC-3′; SNHG14 reverse, 5′-CTT CCA AAA GCC TTC TGC CTT AG-3′; GAPDH forward, 5′-GCA CCG TCA AGG CTG AGA AC-3′; and GAPDH reverse, 5′-AGG GAT CTC GCT CCT GGA A-3′.

CCK-8 assay

Transfected cells were collected at 24 h post-transfection and seeded at 2,000/well in 96-well plates. The CCK-8 assay was conducted to measure cellular proliferation at 4 time points: 0, 24, 48 and 72 h after seeding. At these time points, the culture medium was replaced with fresh DMEM containing 10 µl CCK-8 solution (Dojindo Molecular Technologies, Inc.), followed by incubation for 2 h at 37°C and 5% CO2. The optical density value was then detected at a wavelength of 450 nm. Each assay was performed in triplicate, and every group was analyzed in 3 replicate wells.

Flow-cytometric analysis of apoptosis

A total of 1.5×106 transfected cells were incubated with 0.25% trypsin, harvested and rinsed twice with pre-chilled PBS. Then, the apoptotic rate was evaluated with the Annexin V-fluorescein isothiocyanate (FITC) Apoptosis Detection kit (BioLegend, Inc.). Briefly, transfected cells were resuspended in 100 µl binding buffer prior to counterstaining with 5 µl Annexin V-FITC and 5 µl propidium iodide solution. The stained cells were analyzed on a flow cytometer (FACScan; BD Biosciences). Data were analyzed using CellQuest™ software v.5.1 (BD Biosciences).

Migration and invasion assays

A total of 5×104 cells were resuspended in 200 µl FBS-free DMEM and were subjected to the migration and invasion assays. For the migration assay, the cells were inoculated into the upper chambers of Transwell inserts (8 µm pore size; Corning Incorporated), while Matrigel (BD Biosciences)-precoated Transwell inserts were employed for determining the invasive ability and were loaded with equal numbers of cells.

A total of 50 µl Matrigel was evenly smeared onto the upper chambers of the Transwell inserts and incubated at 37°C for 1 h. The complete culture medium (supplemented with 20% of FBS) was added into the lower chambers and functioned as a chemoattractant. After 24 h incubation, the non-traversing cells remaining on the upper surface of the insert were gently wiped off with a cotton swab. The migratory and invasive cells were fixed in 4% formaldehyde at room temperature for 15 min and stained in a 0.1% crystal violet solution at room temperature for 15 min. Following extensive washing (5 min), the migratory and invasive cells were counted in 5 randomly selected visual fields under an IX71 inverted light microscope (Olympus Corporation) at magnification, ×200.

Mouse xenograft tumor model

All experimental procedures involving animals were conducted in accordance with the Animal Protection Law of the People's Republic of China-2009 for experimental animals (39,40) and were approved by the Animal Care Committee of People's Hospital of Rizhao. A total of 6 female BALB/c mice (average weight, 20 g; age, 4-6 weeks) were purchased from the Animal Experimental Center of Jilin University and were randomly assigned to two groups: Si-SNHG14 and si-NC. In the si-SNHG14 group, a total of 5×106 si-SNHG14-transfected cells were injected subcutaneously into the flank of the mice, whereas si-NC-transfected cells were injected into the flank of mice in the si-NC group. Each group contained 3 nude mice. The animals were maintained under specific pathogen-free conditions at 25°C and 50% humidity, with a 10: 14 light: Dark cycle and ad libitum food/water access. The width and length of the resultant tumor xenografts were monitored with an interval of 4 days, and their volume was calculated via the following formula: Volume = (length × width2)/2. A total of 4 weeks after injection, all mice were euthanized via cervical dislocation; tumor xenografts were then excised and weighed. The humane endpoints used in the study included: Tumor diameter >15 mm; tumor ulceration; and extreme weight loss. None of these endpoints were observed in the mice during the study period.

RNA immunoprecipitation (RIP) assay

This assay was performed using the Magna RIP RNA-Binding Protein Immunoprecipitation kit (cat. no. 17-700; EMD Millipore). In brief, cells were incubated with RIP lysis buffer, and the obtained cellular lysates were next probed with magnetic beads conjugated with a human anti-AGO2 antibody or control IgG (1:5,000 dilution; cat. no. 03-110; EMD Millipore). The cell lysates were then treated with proteinase K buffer (150 µl) at 55°C for 30 min to digest the protein. The expression of SNHG14 and miR-124 in RIP-derived immunoprecipitated RNA was measured by RT-qPCR, as aforementioned.

Bioinformatics analysis

starBase 3.0 (http://starbase.sysu.edu.cn/), an open-source platform for studying the lncRNA-miRNA interactions, was applied to search for the target(s) of SNHG14.

Luciferase reporter assay

The fragments of SNHG14 containing predicted wild-type (wt) and mutant (mut) miR-124-binding sequences were amplified by Shanghai GenePharma Co., Ltd., and inserted into the luciferase reporter gene of the pmirGLO vector (Promega Corporation), producing the reporter plasmids SNHG14-wt and SNHG14-mut, respectively. Following this, the gener-ated plasmids were co-transfected with either agomir-124 or agomir-NC into RB cells using Lipofectamine 2000. After 48 h of incubation, firefly luciferase activity was normalized to that of Renilla luciferase as determined with the Dual-Luciferase® Reporter Assay kit (Promega Corporation).

Western blot analysis

Radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology) was used for the extraction of total protein. The concentration of total protein was quantified via the BCA kit (Beyotime Institute of Biotechnology). Equal amounts of proteins (30 µg) were loaded, separated by 10% SDS-PAGE and then transferred to polyvinylidene difluoride membranes (EMD Millipore), followed by blocking for 2 h with 5% fat-free milk at room temperature and probing with primary antibodies overnight 4°C. Next, after 2 h incubation with a goat anti-rabbit immunoglobulin G horseradish peroxidase-conjugated secondary antibody (cat. no. sc-516102; 1:5,000 dilution; Santa Cruz Biotechnology, Inc.) at room temperature, the protein signals were detected using the Pierce™ ECL Western Blotting Substrate (Pierce; Thermo Fisher Scientific, Inc.). The primary antibodies used in this assay included anti-STAT3 (cat. no. sc-8019; 1:1,000 dilution; Santa Cruz Biotechnology Inc.) and anti-GAPDH (cat. no. sc-47724; 1:1,000 dilution; Santa Cruz Biotechnology, Inc.). GAPDH served as an endogenous control for the quantification of STAT3 protein expression. Quantity One software v.4.62 (Bio-Rad Laboratories, Inc.) was used for densitometry analysis.

Statistical analysis

Data were presented as mean ± standard deviation, and analyzed using SPSS statistics software (version 21.0; IBM Corp.). The comparisons between two groups were conducted via Student's t-test, whereas comparisons between multiple groups were performed by one-way analysis of variance followed by Bonferroni's post hoc test. The association between SNHG14 expression and clinical parameters among the patients with RB was assessed by the χ2 test. Spearman's correlation between SNHG14 and miR-124 expression levels in RB tissues was calculated next. Survival curves were created by Kaplan-Meier analysis and compared using the log-rank test. P<0.05 was considered to indicate a statistically significant difference.

Results

SNHG14 expression is high in RB tumors and cell lines

The expression of SNHG14 in the RB tissue samples and normal retinas was examined by RT-qPCR. The results indicated that the expression of SNHG14 was increased in RB tissue samples compared with in normal retinas (Fig. 1A). In addition, SNHG14 expression was examined in 3 RB cell lines: Y79; SO-RB50; and Weri-RB-1. The normal retinal pigmented epithelial ARPE-19 cell line served as the control. As compared with the ARPE-19 cells, all 3 RB cell lines exhibited marked SNHG14 upregulation (Fig. 1B). All of the patients in the study cohort were assigned to either an SNHG14 high-expression or SNHG14 low-expression group based on the median level of SNHG14 among the RB tissue samples, which served as the cutoff value (median SNHG14 expression in RB tissues; 2.21). The correlation between SNHG14 levels and clinical parameters of the patients with RB was investigated. The analysis indicated that an increased level of SNHG14 was significantly associated with the IIRC stage (P=0.004), optic nerve invasion (P=0.006) and differentiation grade (P=0.031; Table I). Notably, the patients with RB in the SNHG14 high-expression group exhibited shorter overall survival times compared with the patients in the SNHG14 low-expression group (Fig. 1C; P=0.032). These results implied that SNHG14 may be implicated in RB carcinogenesis.

Table I

Associations between SNHG14 expression and clini-copathological parameters of patients with retinoblastoma.

Table I

Associations between SNHG14 expression and clini-copathological parameters of patients with retinoblastoma.

ParametersSNHG14 expression
High (n=22)Low (n=21)P-value
Age, years0.537
 <31512
 ≥379
Sex0.763
 Male1011
 Female1210
IIRC stage0.004
 Early stage (A, B, C)312
 Advanced Stage (D, E)199
Optic nerve invasion0.006
 Negative716
 Positive155
Differentiation grade0.031
 Well/moderately916
 Poorly/undifferentiated135

[i] SNHG14, small nucleolar RNA host gene 14.

Silencing of SNHG14 inhibits RB cell proliferation, migration and invasion, and promotes apoptosis

As SNHG14 was identified to be markedly overexpressed in the Y79 and Weri-RB-1 cell lines, these 2 cell lines were selected for subsequent experiments. To examine the functions of SNHG14 in RB progression, si-SNHG14 was transfected into the Y79 and Weri-RB-1 cells to silence endogenous SNHG14 expression. The transfection efficiency was verified by RT-qPCR, which indicated that SNHG14 was knocked down efficiently in the Y79 and Weri-RB-1 cells following transfection with si-SNHG14 (Fig. 2A). The CCK-8 assay was performed to quantify the effect of SNHG14 silencing on RB cell proliferation, and it was revealed that the downregulation of SNHG14 inhibited the proliferation of the Y79 and Weri-RB-1 cells (Fig. 2B). The apoptosis status of the SNHG14-deficient Y79 and Weri-RB-1 cells was examined via flow cytometry. Silencing of SNHG14 enhanced the apoptosis levels of the Y79 and Weri-RB-1 cells (Fig. 2C). Furthermore, the migration and invasion assays revealed that the migratory (Fig. 2D) and invasive (Fig. 2E) abilities were markedly suppressed by SNHG14 silencing. Overall, these results suggested that SNHG14 knockdown inhibited the malignant properties of RB cells in vitro.

SNHG14 functions as a molecular sponge of miR-124 in RB cells

Previous studies have indicated that lncRNAs are implicated in cancer progression by acting as competing endogenous RNAs (ceRNAs) on certain miRNAs, thereby relieving the miRNA-induced repression of their target genes (41-43). To elucidate the mechanisms by which SNHG14 affected the malignant properties of RB cells, bioinformatics analysis was conducted to predict the miRNA(s) that can directly interact with SNHG14. Among the candidates, miR-124 was selected for validation, as this miRNA has been demonstrated to be downregulated in RB and to exert tumor-suppressive actions (36). The putative binding site, the complementary sequence between SNHG14 and miR-124, is presented in Fig. 3A. First, the transfection efficiency of agomir-124 was confirmed via RT-qPCR, and the results indicated that transfection with agomir-124 notably increased the expression of miR-124 in both the Y79 and Weri-RB-1 cells (Fig. 3B).

Figure 3

SNHG14 functions as a sponge for miR-124 and thereby upregulates STAT3. (A) The predicted miR-124-binding sequences in SNHG14 and the designed mut sequence (SNHG14-mut). (B) The expression of miR-124 was examined in Y79 and Weri-RB-1 cells that were transfected with either agomir-124 or agomir-NC. **P<0.01 vs. the agomir-NC group. (C) The luciferase reporter assay was conducted to evaluate the interaction between SNHG14 and miR-124 in RB cells. Luciferase activities were determined in Y79 and Weri-RB-1 cells following co-transfection with either the SNHG14-wt or SNHG14-mut reporter plasmid and either agomir-124 or agomir-NC. *P<0.05 vs. the agomir-NC group. (D) The RIP assay was performed to validate the interaction between SNHG14 and miR-124 in RB. SNHG14 was identified to be preferentially enriched on AGO2-containing beads following the immunoprecipitation in the lysates of Y79 and Weri-RB-1 cells. **P<0.01 vs. the IgG group. (E) The expression of miR-124 was examined in RB tissue samples (n=43) and normal retinas (n=11). *P 0.05 vs. normal retinas. (F) The correlation between SNHG14 and miR-124 levels in RB tissue samples was assessed by Spearman's correlation analysis. R2=0.3864. P<0.0001. (G) miR-124 expression in SNHG14-deficient Y79 and Weri-RB-1 cells was measured by reverse transcription-quantitative polymerase chain reaction. **P<0.01 vs. the si-NC group. (H) Western blot analysis was conducted to examine STAT3 protein expression in Y79 and Weri-RB-1 cells following transfection with either si-SNHG14 or si-NC. **P<0.01 vs. the si-NC group. SNHG14, small nucleolar RNA host gene 14; miR, microRNA; STAT3, signal transducer and activator of transcription 3; wt, wild type; mut, mutant; NC, negative control; RB, retinoblastoma; RIP, RNA immunoprecipitation; si, small interfering RNA.

The luciferase reporter assay was performed to confirm the potential SNHG14/miR-124 interaction: Agomir-124 or agomir-NC and either SNHG14-wt or SNHG14-mut plasmids were co-transfected into the Y79 and Weri-RB-1 cells. The luciferase activities of the Y79 and Weri-RB-1 cells transfected with the SNHG14-wt reporter plasmid were markedly decreased following miR-124 overexpression (P<0.05), whereas no change was observed in the cells transfected with the SNHG14-mut plasmid (Fig. 3C). The results from the RIP assay in the Y79 and Weri-RB-1 cells revealed that SNHG14 was preferentially enriched on the AGO2-containing beads (Fig. 3D), suggesting that SNHG14 is likely bound to the miR-124 RNA-induced silencing complex. Furthermore, miR-124 was much more weakly expressed in RB tissue samples in comparison with normal retinas (Fig. 3E), which was in agreement with a previous study (36). As demonstrated in Fig. 3F, Spearman's correlation analysis indicated an inverse correlation between SNHG14 and miR-124 expression levels among the RB tissue samples (R2=0.3864; P<0.0001). Following this, the regulatory effect of SNHG14 on the miR-124 expression in RB cells was examined by RT-qPCR. The downregulation of SNHG14 markedly increased miR-124 accumulation in Y79 and Weri-RB-1 cells (Fig. 3G). STAT3 has been previously demonstrated to be a direct target gene of miR-124 in RB cells (36); accordingly, STAT3 protein expression levels were measured in SNHG14-deficient Y79 and Weri-RB-1 cells. The results of the western blot analysis indicated that a decrease in SNHG14 expression significantly decreased the protein expression levels of STAT3 in the Y79 and Weri-RB-1 cells (Fig. 3H). When taken together, these results suggested that SNHG14 may act as an miR-124 sponge in RB cells and consequently increase STAT3 expression.

miR-124 silencing neutralizes the effects of SNHG14 knockdown on RB cells

A series of rescue experiments were conducted to further investigate the interaction between SNHG14 and miR-124, and to clarify the molecular events underlying the oncogenic roles of SNHG14 in RB cells. si-SNHG14 was co-transfected with either antagomir-124 or antagomir-NC into the Y79 and Weri-RB-1 cells. Firstly, it was confirmed that transfection with antagomir-124 resulted in efficient miR-124 silencing in the Y79 and Weri-RB-1 cells (Fig. 4A). The upregulation of miR-124 induced by SNHG14 knockdown was identified to be attenuated in the Y79 and Weri-RB-1 cells that were co-transfected with antagomir-124 (Fig. 4B). Subsequent functional experiments revealed that SNHG14 silencing inhibited Y79 and Weri-RB-1 cell proliferation (Fig. 4C), induced their apoptosis (Fig. 4D), and attenuated their migratory (Fig. 4E) and invasive (Fig. 4F) capabilities. Conversely, these si-SNHG14-mediated effects on cell proliferation, apoptosis, migration and invasion were reversed by antagomir-124 co-transfection. These data suggested that SNHG14 performs its oncogenic functions during RB progression via the downregulation of miR-124.

Downregulation of SNHG14 inhibits the malignant characteristics of RB cells in vitro via the miR-124/STAT3 regulatory loop

To further clarify the participation of the SNHG14/miR-124/STAT3 axis in the malignancy of RB, si-SNHG14, in combination with either STAT3 overexpression plasmid pc-STAT3 or the empty pcDNA3.1 vector, was introduced into the Y79 and Weri-RB-1 cells. The results of the western blot analysis indicated that si-SNHG14 significantly decreased the protein expression of STAT3 in the Y79 and Weri-RB-1 cells, but co-transfection with pc-STAT3 abrogated this effect (Fig. 5A). Furthermore, recovery of STAT3 expression attenuated the effects of SNHG14 silencing on Y79 and Weri-RB-1 cell proliferation (Fig. 5B), apoptosis (Fig. 5C), migration (Fig. 5D) and invasion (Fig. 5E) in vitro. Therefore, SNHG14 participates in RB carcinogenesis in vitro by serving as a ceRNA that sponges miR-124, consequently inhibiting the miR-124-induced repression of its direct target gene STAT3.

Decrease in SNHG14 expression inhibits RB tumor growth in vivo

The mouse xenograft tumor model was constructed to assess the biological effect of SNHG14 on the in vivo tumor growth of RB cells. Y79 cells transfected with either si-SNHG14 or si-NC were subcutaneously inoculated into the flank of nude mice. On day 28, all the mice were euthanized, and a representative image of the formed tumor xenografts is presented in Fig. 6A. The volume of the tumor xenografts was markedly decreased in the si-SNHG14 group compared with in the si-NC group (Fig. 6B). In addition, the mean tumor weight was decreased in the si-SNHG14 group compared with the si-NC group (Fig. 6C). RT-qPCR analysis suggested that the tumor xenografts derived from si-SNHG14-transfected Y79 cells presented decreased SNHG14 (Fig. 6D) and increased miR-124 (Fig. 6E). Western blot analysis revealed that the levels of STAT3 protein were decreased in the tumor xenografts obtained from the si-SNHG14 group (Fig. 6F). These results suggested that SNHG14 downregulation inhibited the tumor growth of RB cells in vivo, and that this effect was mediated by the upregulation of miR-124 and downregulation of STAT3.

Discussion

Previous evidence has highlighted the aberrant expression of lncRNAs in RB, and that these aberrations serve crucial roles in the RB carcinogenesis and progression (44-46). LncRNAs are implicated in the regulation of a wide range of biological activities and perform either tumor-suppressive or oncogenic functions (17,47,48). Therefore, the therapies that target lncRNAs hold promise for the management of RB. In the present study, we hypothesized that a known cancer-associated lncRNAs, SNHG14, may serve crucial roles in the modulation of RB progression. Therefore, SNHG14 expression levels were measured in RB tumors and its clinical value was assessed among patients with RB. Then, the effect of SNHG14 on the malignant characteristics of RB cells in vitro and in vivo was examined in a series of functional experiments. Finally, a systemic approach was applied to explore the mechanisms behind the oncogenic activities of SNHG14 in RB in vivo.

SNHG14 expression is low in glioma tissues and cell lines (27). Conversely, SNHG14 expression is high in non-small cell lung cancer (NSCLC) cells (28,49,50), and SNHG14 upregulation is associated with tumor size and TNM stage (28). Patients with NSCLC featuring high SNHG14 expression exhibit shorter overall survival and recurrence-free survival times compared with the patients with low SNHG14 expression (28). SNHG14 is known to be overexpressed in cervical cancer and to be significantly associated with tumor stage, lymph node metastasis and shorter overall survival (29,30). SNHG14 also exhibits high expression levels in ovarian (31,32), breast (33), bladder (34) and gastric cancer (35). Nevertheless, to the best of our knowledge, the expression profile and clinical significance of SNHG14 in RB have never been determined. The present study identified that SNHG14 was overexpressed in RB tissues and cell lines. SNHG14 overexpression was associated with the IIRC stage, optic nerve invasion, differentiation grade and poor clinical outcomes among the patients with RB. These results suggest that SNHG14 is a potential biomarker for the diagnosis and prognosis of RB.

The biological functions of SNHG14 have been well studied in a range of cancer types. SNHG14 sponges miR-92a-3p to exert tumor-suppressive effects on the progression of glioma by inhibiting cell viability and invasion and by promoting apoptosis (27). By contrast, SNHG14 was validated as an oncogenic lncRNA in NSCLC and identified to participate in the regulation of cell proliferation, cell cycle, apoptosis, colony formation and gefitinib chemosensitivity in vitro, and tumor growth in vivo (28). These effects are mediated by the ceRNA effect on miR-340 (28), by ATP binding cassette subfamily B member 1 upregulation via the sponging of miR-206-3p (49) and by affecting the regulatory loop (50). In cervical cancer, SNHG14 silencing decreased tumor cell viability, proliferation, migration and invasion, and facilitated apoptosis through modulation of the miR-206/tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein zeta axis and activation of the Janus kinase-STAT pathway (29,30). Besides, SNHG14 exerts oncogenic actions on the malignancy of ovarian (31,32), breast (33), bladder (34), and gastric (35). However, data on the detailed involvement of SNHG14 in the malignant properties of RB is limited. In the present study, functional assays revealed that the depletion of SNHG14 decreased the levels of RB cell proliferation, migration and invasion in vitro, promoted apoptosis in vitro and inhibited tumor growth in vivo.

Studies investigating the mechanisms of action of SNHG14 on RB malignancy are crucial for understanding the roles of SNHG14 in RB pathogenesis and for the identification of effective therapeutic targets. In the present study, SNHG14 was identified to serve as a molecular sponge of miR-124 in RB cells, thereby upregulating STAT3. miR-124 expression has been demonstrated to be low in RB tumors and cell lines (36). miR-124 exerts tumor-suppressive effects on RB progression by repressing RB cell proliferation, migration and invasion, and by inducing apoptosis (36). Several lncRNAs, including nuclear paraspeckle assembly transcript 1 (51), X inactive specific transcript (52) and metastasis associated lung adenocarcinoma transcript 1 (53,54), have been identified to sponge miR-124 and to contribute to RB aggressiveness in vitro and in vivo. The results of the present study revealed that inhibition of miR-124 abrogated the SNHG14 knockdown-induced suppression of RB cell proliferation, migration and invasion, and reversed the pro-apoptotic effects of the SNHG14 knock-down on RB cells.

A mechanistic investigation has confirmed STAT3 as a direct target gene of miR-124 in RB cells (36). STAT3, a key transcription factor in the STAT family, is a signal mediator that is activated by various cytokines, growth factors and interferons (55). It is overexpressed in RB, performs oncogenic roles during RB progression, and is involved in the regulation of multiple physiological and pathological processes in tumor development (56-59). The results of the present study demonstrated clearly that STAT3 expression is directly controlled by the SNHG14/miR-124 regulatory loop in RB.

The present study has certain limitations. The effect of SNHG14 on the metastasis of RB cells in vivo was not examined directly. This will be addressed future studies.

In conclusion, the present study demonstrated that SNHG14 was overexpressed in RB, and that it was associated with poor clinical outcomes among patients with RB. SNHG14 serves as an oncogenic lncRNA in RB cells in vitro and in vivo. The regulatory role of SNHG14 in RB malignancy is partly mediated by its function as a ceRNA of miR-124, and consequent upregulation of STAT3. These results may offer a novel perspective on the targeted therapy of RB.

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

SZ and XS designed the study. XS and HS conducted RT-qPCR, CCK-8 assay, RIP assay and luciferase reporter assay. Flow cytometric analysis, and migration and invasion assays were carried out by SL. JG constructed the mouse xenograft tumor model and conducted western blotting. Statistical analysis was conducted by SZ. All authors read and approved the final manuscript.

Ethics approval and informed consent

The present study was conducted with the approval of the Clinical Research Ethics Committee of People's Hospital of Rizhao and was performed following the principles of the Declaration of Helsinki. All participants provided written informed consent for participation prior to surgical resection. All experimental procedures involving animals were conducted in accordance with the Animal Protection Law of the People's Republic of China-2009 for experimental animals and were approved by the Animal Care Committee of People's Hospital of Rizhao.

Patient consent for publication

All participants provided written informed consent for publication prior to surgical resection.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kamihara J, Bourdeaut F, Foulkes WD, Molenaar JJ, Mossé YP, Nakagawara A, Parareda A, Scollon SR, Schneider KW, Skalet AH, et al: Retinoblastoma and neuroblastoma predisposition and surveillance. Clin Cancer Res. 23:e98–e106. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Ramirez-Ortiz MA, Lansingh VC, Eckert KA, Haik BG, Phillips BX, Bosch-Canto V, González-Pérez G, Villavicencio-Torres A and Etulain-González A: Systematic review of the current status of programs and general knowledge of diagnosis and management of retinoblastoma. Bol Med Hosp Infant Mex. 74:41–54. 2017.

3 

Bansal A, Noronha V, Krishnakumar S and Khetan V: A tale of absent crystalline lens in an eye with retinoblastoma. Ocul Oncol Pathol. 4:250–253. 2018. View Article : Google Scholar

4 

Errico A: Cancer therapy: Retinoblastoma-chemotherapy increases the risk of secondary cancer. Nat Rev Clin Oncol. 11:6232014. View Article : Google Scholar

5 

Rojanaporn D, Boontawon T, Chareonsirisuthigul T, Thanapanpanich O, Attaseth T, Saengwimol D, Anurathapan U, Sujirakul T, Kaewkhaw R and Hongeng S: Spectrum of germline RB1 mutations and clinical manifestations in retinoblastoma patients from Thailand. Mol Vis. 24:778–788. 2018.

6 

Soliman SE, Racher H, Zhang C, MacDonald H and Gallie BL: Genetics and molecular diagnostics in retinoblastoma-an update. Asia Pac J Ophthalmol (Phila). 6:197–207. 2017.

7 

Wang Y, Yuan J, Yang L, Wang P, Wang X, Wu Y, Chen K, Ma R, Zhong Y, Guo X, et al: Inhibition of migration and invasion by berberine via inactivation of PI3K/Akt and p38 in human retino-blastoma cell line. Adv Clin Exp Med. 27:899–905. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Abramson DH, Shields CL, Munier FL and Chantada GL: Treatment of retinoblastoma in 2015: Agreement and disagreement. JAMA Ophthalmol. 133:1341–1347. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Iyer MK, Niknafs YS, Malik R, Singhal U, Sahu A, Hosono Y, Barrette TR, Prensner JR, Evans JR, Zhao S, et al: The land-scape of long noncoding RNAs in the human transcriptome. Nat Genet. 47:199–208. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Xing Y, Zhao Z, Zhu Y, Zhao L, Zhu A and Piao D: Comprehensive analysis of differential expression profiles of mRNAs and lncRNAs and identification of a 14-lncRNA prognostic signature for patients with colon adenocarcinoma. Oncol Rep. 39:2365–2375. 2018.PubMed/NCBI

11 

Wu Y, Shao A, Wang L, Hu K, Yu C, Pan C and Zhang S: The role of lncRNAs in the distant metastasis of breast cancer. Front Oncol. 9:4072019. View Article : Google Scholar : PubMed/NCBI

12 

Xuan W, Yu H, Zhang X and Song D: Crosstalk between the lncRNA UCA1 and microRNAs in cancer. FEBS Lett. 593:1901–1914. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Zhu W, Liu H, Wang X, Lu J and Yang W: Long noncoding RNAs in bladder cancer prognosis: A meta-analysis. Pathol Res Pract. 215:1524292019. View Article : Google Scholar : PubMed/NCBI

14 

Xu C, Hu C, Wang Y and Liu S: Long noncoding RNA SNHG16 promotes human retinoblastoma progression via sponging miR-140-5p. Biomed Pharmacother. 117:1091532019. View Article : Google Scholar : PubMed/NCBI

15 

Han N, Zuo L, Chen H, Zhang C, He P and Yan H: Long non-coding RNA homeobox A11 antisense RNA (HOXA11-AS) promotes retinoblastoma progression via sponging miR-506-3p. Onco Targets Ther. 12:3509–3517. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Quan LJ and Wang WJ: FEZF1-AS1 functions as an oncogenic lncRNA in retinoblastoma. Biosci Rep. 39:BSR201907542019. View Article : Google Scholar : PubMed/NCBI

17 

Wu XZ, Cui HP, Lv HJ and Feng L: Knockdown of lncRNA PVT1 inhibits retinoblastoma progression by sponging miR-488-3p. Biomed Pharmacother. 112:1086272019. View Article : Google Scholar : PubMed/NCBI

18 

Bi LL, Han F, Zhang XM and Li YY: LncRNA MT1JP acts as a tumor inhibitor via reciprocally regulating Wnt/β-Catenin pathway in retinoblastoma. Eur Rev Med Pharmacol Sci. 22:4204–4214. 2018.PubMed/NCBI

19 

Zhang A, Shang W, Nie Q, Li T and Li S: Long non-coding RNA H19 suppresses retinoblastoma progression via counteracting miR-17-92 cluster. J Cell Biochem. 119:3497–3509. 2018. View Article : Google Scholar

20 

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

21 

Croce CM: Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 10:704–714. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Chandra S, Vimal D, Sharma D, Rai V, Gupta SC and Chowdhuri DK: Role of miRNAs in development and disease: Lessons learnt from small organisms. Life Sci. 185:8–14. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Voorhoeve PM: MicroRNAs: Oncogenes, tumor suppressors or master regulators of cancer heterogeneity? Biochim Biophys Acta. 1805:72–86. 2010.

24 

Li W, Wang J, Zhang D, Zhang X, Xu J and Zhao L: MicroRNA-98 targets HMGA2 to inhibit the development of retinoblastoma through mediating Wnt/β-catenin pathway. Cancer Biomark. 25:79–88. 2019. View Article : Google Scholar

25 

Delsin LEA, Salomao KB, Pezuk JA and Brassesco MS: Expression profiles and prognostic value of miRNAs in retinoblastoma. J Cancer Res Clin Oncol. 145:1–10. 2019. View Article : Google Scholar

26 

Liao Y, Yin X, Deng Y and Peng X: MiR-140-5p suppresses retinoblastoma cell growth via inhibiting c-Met/AKT/mTOR pathway. Biosci Rep. 38:BSR201807762018. View Article : Google Scholar : PubMed/NCBI

27 

Wang Q, Teng Y, Wang R, Deng D, You Y, Peng Y, Shao N and Zhi F: The long non-coding RNA SNHG14 inhibits cell proliferation and invasion and promotes apoptosis by sponging miR-92a-3p in glioma. Oncotarget. 9:12112–12124. 2018.PubMed/NCBI

28 

Zhang Z, Wang Y, Zhang W, Li J, Liu W and Lu W: Long non-coding RNA SNHG14 exerts oncogenic functions in non-small cell lung cancer through acting as an miR-340 sponge. Biosci Rep. 39:BSR201809412019. View Article : Google Scholar :

29 

Ji N, Wang Y, Bao G, Yan J and Ji S: LncRNA SNHG14 promotes the progression of cervical cancer by regulating miR-206/YWHAZ. Pathol Res Pract. 215:668–675. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Zhang YY, Li M, Xu YD and Shang J: LncRNA SNHG14 promotes the development of cervical cancer and predicts poor prognosis. Eur Rev Med Pharmacol Sci. 23:3664–3671. 2019.PubMed/NCBI

31 

Li L, Zhang R and Li SJ: Long noncoding RNA SNHG14 promotes ovarian cancer cell proliferation and metastasis via sponging miR-219a-5p. Eur Rev Med Pharmacol Sci. 23:4136–4142. 2019.PubMed/NCBI

32 

Zhao YL and Huang YM: LncSNHG14 promotes ovarian cancer by targeting microRNA-125a-5p. Eur Rev Med Pharmacol Sci. 23:3235–3242. 2019.PubMed/NCBI

33 

Xie SD, Qin C, Jin LD, Wang QC, Shen J, Zhou JC, Chen YX, Huang AH, Zhao WH and Wang LB: Long noncoding RNA SNHG14 promotes breast cancer cell proliferation and invasion via sponging miR-193a-3p. Eur Rev Med Pharmacol Sci. 23:2461–2468. 2019.PubMed/NCBI

34 

Li J, Wang AS, Wang S, Wang CY, Xue S, Guan H, Li WY, Ma TT and Shan YX: LncSNHG14 promotes the development and progression of bladder cancer by targeting miRNA-150-5p. Eur Rev Med Pharmacol Sci. 23:1022–1029. 2019.PubMed/NCBI

35 

Liu Z, Yan Y, Cao S and Chen Y: Long non-coding RNA SNHG14 contributes to gastric cancer development through targeting miR-145/SOX9 axis. J Cell Biochem. 119:6905–6913. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Liu S, Hu C, Wang Y, Shi G, Li Y and Wu H: MiR-124 inhibits proliferation and invasion of human retinoblastoma cells by targeting STAT3. Oncol Rep. 36:2398–2404. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Ortiz MV and Dunkel IJ: Retinoblastoma. J Child Neurol. 31:227–236. 2016. View Article : Google Scholar

38 

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

39 

Han X, Huang T and Han J: Long noncoding RNA VPS9D1-AS1 augments the malignant phenotype of non-small cell lung cancer by sponging microRNA-532-3p and thereby enhancing HMGA2 expression. Aging (Albany NY). 12:370–386. 2020. View Article : Google Scholar

40 

Jia J, Wang J, Yin M and Liu Y: MicroRNA-605 directly targets SOX9 to alleviate the aggressive phenotypes of glioblastoma multiforme cell lines by deactivating the PI3K/Akt pathway. Onco Targets Ther. 12:5437–5448. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Klec C, Prinz F and Pichler M: Involvement of the long noncoding RNA NEAT1 in carcinogenesis. Mol Oncol. 13:46–60. 2019. View Article : Google Scholar :

42 

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

43 

Shuwen H, Qing Z, Yan Z and Xi Y: Competitive endogenous RNA in colorectal cancer: A systematic review. Gene. 645:157–162. 2018. View Article : Google Scholar

44 

Su S, Gao J, Wang T, Wang J, Li H and Wang Z: Long non-coding RNA BANCR regulates growth and metastasis and is associated with poor prognosis in retinoblastoma. Tumour Biol. 36:7205–7211. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Gao Y and Lu X: Decreased expression of MEG3 contributes to retinoblastoma progression and affects retinoblastoma cell growth by regulating the activity of Wnt/beta-catenin pathway. Tumour Biol. 37:1461–1469. 2016. View Article : Google Scholar

46 

Dong C, Liu S, Lv Y, Zhang C, Gao H, Tan L and Wang H: Long non-coding RNA HOTAIR regulates proliferation and invasion via activating Notch signalling pathway in retinoblastoma. J Biosci. 41:677–687. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Lyu X, Ma Y, Wu F and Wang L and Wang L: LncRNA NKILA inhibits retinoblastoma by downregulating lncRNA XIST. Curr Eye Res. 44:975–979. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Yan G, Su Y, Ma Z, Yu L and Chen N: Long noncoding RNA LINC00202 promotes tumor progression by sponging miR-3619-5p in retinoblastoma. Cell Struct Funct. 44:51–60. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Wu K, Li J, Qi Y, Zhang C, Zhu D, Liu D and Zhao S: SNHG14 confers gefitinib resistance in non-small cell lung cancer by up-regulating ABCB1 via sponging miR-206-3p. Biomed Pharmacother. 116:1089952019. View Article : Google Scholar : PubMed/NCBI

50 

Jiao P, Hou J, Yao M, Wu J and Ren G: SNHG14 silencing suppresses the progression and promotes cisplatin sensitivity in non-small cell lung cancer. Biomed Pharmacother. 117:1091642019. View Article : Google Scholar : PubMed/NCBI

51 

Wang L, Yang D, Tian R and Zhang H: NEAT1 promotes retinoblastoma progression via modulating miR-124. J Cell Biochem. 120:15585–15593. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Hu C, Liu S, Han M, Wang Y and Xu C: Knockdown of lncRNA XIST inhibits retinoblastoma progression by modulating the miR-124/STAT3 axis. Biomed Pharmacother. 107:547–554. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Huang J, Yang Y, Fang F and Liu K: MALAT1 modulates the autophagy of retinoblastoma cell through miR-124-mediated stx17 regulation. J Cell Biochem. 119:3853–3863. 2018. View Article : Google Scholar

54 

Liu S, Yan G, Zhang J and Yu L: Knockdown of long noncoding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) inhibits proliferation, migration, and invasion and promotes apoptosis by targeting miR-124 in retino-blastoma. Oncol Res. 26:581–591. 2018. View Article : Google Scholar

55 

Wagner KU and Schmidt JW: The two faces of Janus kinases and their respective STATs in mammary gland development and cancer. J Carcinog. 10:322011. View Article : Google Scholar

56 

Xu B, Chen X, Tan J and Xu X: Effect of AG490 on JAK2/STAT3 signaling pathway in human retinoblastoma HXO-RB44 cell lines. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 43:1061–1067. 2018.In Chinese. PubMed/NCBI

57 

Li Y, Sun W, Han N, Zou Y and Yin D: Curcumin inhibits proliferation, migration, invasion and promotes apoptosis of retinoblastoma cell lines through modulation of miR-99a and JAK/STAT pathway. BMC Cancer. 18:12302018. View Article : Google Scholar : PubMed/NCBI

58 

Liu S, Zhang X, Hu C, Wang Y and Xu C: MiR-29a inhibits human retinoblastoma progression by targeting STAT3. Oncol Rep. 39:739–746. 2018.

59 

Jo DH and Kim JH, Cho CS, Cho YL, Jun HO, Yu YS, Min JK and Kim JH: STAT3 inhibition suppresses proliferation of retino-blastoma through down-regulation of positive feedback loop of STAT3/miR-17-92 clusters. Oncotarget. 5:11513–11525. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2020
Volume 45 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun X, Shen H, Liu S, Gao J and Zhang S: Long noncoding RNA SNHG14 promotes the aggressiveness of retinoblastoma by sponging microRNA‑124 and thereby upregulating STAT3 Retraction in /10.3892/ijmm.2022.5126. Int J Mol Med 45: 1685-1696, 2020
APA
Sun, X., Shen, H., Liu, S., Gao, J., & Zhang, S. (2020). Long noncoding RNA SNHG14 promotes the aggressiveness of retinoblastoma by sponging microRNA‑124 and thereby upregulating STAT3 Retraction in /10.3892/ijmm.2022.5126. International Journal of Molecular Medicine, 45, 1685-1696. https://doi.org/10.3892/ijmm.2020.4547
MLA
Sun, X., Shen, H., Liu, S., Gao, J., Zhang, S."Long noncoding RNA SNHG14 promotes the aggressiveness of retinoblastoma by sponging microRNA‑124 and thereby upregulating STAT3 Retraction in /10.3892/ijmm.2022.5126". International Journal of Molecular Medicine 45.6 (2020): 1685-1696.
Chicago
Sun, X., Shen, H., Liu, S., Gao, J., Zhang, S."Long noncoding RNA SNHG14 promotes the aggressiveness of retinoblastoma by sponging microRNA‑124 and thereby upregulating STAT3 Retraction in /10.3892/ijmm.2022.5126". International Journal of Molecular Medicine 45, no. 6 (2020): 1685-1696. https://doi.org/10.3892/ijmm.2020.4547