Open Access

miR‑1299/NOTCH3/TUG1 feedback loop contributes to the malignant proliferation of ovarian cancer

Corrigendum in: /10.3892/or.2023.8533

  • Authors:
    • Yuqing Pei
    • Kexin Li
    • Xiaoying Lou
    • Yue Wu
    • Xin Dong
    • Wenpeng Wang
    • Ning Li
    • Donghong Zhang
    • Wei Cui
  • View Affiliations

  • Published online on: May 25, 2020     https://doi.org/10.3892/or.2020.7623
  • Pages: 438-448
  • Copyright: © Pei 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

Recent studies have revealed the oncogenic role of notch reporter 3 (NOTCH3) in ovarian cancer (OC). However, the possible regulators and mechanisms underlying notch receptor 3 (NOTCH3)‑mediated behaviors in OC remain to be completely investigated. In the present study, we aimed to identify regulators of NOTCH3 and their interactions underlying the pathogenesis of OC. Bioinformatics analysis and luciferase reporter assay were used to identify potential regulatory miRNAs and lncRNAs of NOTCH3 in OC. Several in vivo and in vitro assays were performed to evaluate their effects on the proliferative ability mediated by NOTCH3. We identified microRNA‑1299 (miR‑1299) as a novel negative regulator of NOTCH3. miR‑1299 was downregulated in OC and was found to be considerably correlated with tumor differentiation. Upregulation of miR‑1299 inhibited cell proliferation, colony formation, and 5‑ethynyl‑2'‑deoxyuridine (EdU) incorporation, as well as induced cell cycle arrest in the G0G1 phase in OC cells. Overexpression of miR‑1299 in xenograft mouse models suppressed tumor growth in vivo. The lncRNA taurine upregulated gene 1 (TUG1), acting as a sponge of miR‑1299, was found to upregulate NOTCH3 expression and promote cell proliferation in OC through the competing endogenous RNA mechanism. In addition, TUG1 was found to be a potential downstream target of NOTCH3, forming a miR‑1299/NOTCH3/TUG1 feedback loop in the development of OC. Collectively, our findings improve the understanding of NOTCH3‑mediated regulation in OC pathogenesis and facilitate the development of miRNA‑ and lncRNA‑directed diagnostics and therapeutics against this disease.

Introduction

Ovarian cancer (OC) continues to be one of the most lethal malignant tumors among women worldwide. In the United States, OC accounts for only 2.5% of all malignancies among females but for 5% of cancer deaths, making it the fifth leading cause of cancer-related deaths in females (1,2). In China, the mortality rate of OC is ranked tenth among females throughout the country (3). The low survival rates in OC are largely driven by a late-stage diagnosis and a high rate of tumor recurrence. Although most OC patients have no evidence of disease after first treatment, approximately 19% of early OC patients and 60 to 85% of advanced OC patients experience a relapse, and recurrent OC after treatment is almost incurable (46). Current treatment for OC is not confined merely to surgery and classical chemotherapy. Targeted therapy, such as polyADP ribose polymerase (PARP) inhibitors, as well as immunotherapy, are promising treatments (710).

The Notch signaling pathway is an important system that regulates cell proliferation, differentiation, and apoptosis. Integrated genomic analyses revealed that Notch is the main signaling pathway involved in the pathophysiology of OC (11,12). The expression of notch reporter 3 (NOTCH3) was found to be elevated in OC and closely correlated with the clinical stage, pathological grade, lymph node metastasis, drug-resistant recurrence, and survival rate of OC patients (1315). NOTCH3 promotes the malignant progression of OC by enhancing the proliferation of tumor cells, maintaining their stemness, and resisting apoptosis (16,17). A better understanding of the tumor-specific regulation of NOTCH3 is crucial and may contribute to targeted therapy for OC. However, limited information regarding the molecules that regulate the expression of NOTCH3 is available.

Approximately 70% of the human genome can be transcribed, while less than 2% of the genome encodes proteins, generating thousands of non-coding transcripts (ncRNAs) (18,19). Among them, microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are key regulators of gene expression at the post-transcriptional level. miRNAs are single-stranded non-coding small RNAs with a length of 17–22 nt. They are destabilizers and repressors of translation of mRNAs and regulate diverse biological functions, including proliferation, migration, apoptosis, angiogenesis, and metabolic progression in various cancers, including OC (2024). lncRNAs are ncRNAs that are longer than 200 nt. They are implicated in multiple cancers and play critical roles in tumor initiation and progression (2528). Recent studies have revealed that lncRNAs can act as competing endogenous RNA (ceRNA) or miRNA ‘sponges’ to modulate crucial genes in cancer (2932), providing a feasible way to understand the regulatory mechanisms underlying cancer pathogenesis and identify novel diagnostic biomarkers or potential therapeutic candidates.

Given the significant oncogenic role of NOTCH3 in OC, we aimed to determine the functional significance of NOTCH3-mediated regulatory ncRNAs in OC. In this study, we found that microRNA-1299 (miR-1299) was a novel negative regulator of NOTCH3 expression in OC with clinical significance. Downregulation of miR-1299 promoted the malignant progression of OC by tumor cell proliferation. The lncRNA taurine upregulated gene 1 (TUG1) was found to function as a ceRNA, regulating NOTCH3 expression by sponging miR-1299 in OC. TUG1 was also identified as a potential downstream target of NOTCH3 and formed a miR-1299/NOTCH3/TUG1 feedback loop. Our results elucidate the regulatory mechanism underlying NOTCH3 expression in OC and provide potential therapeutic targets for the anticancer therapy of OC.

Materials and methods

Cell lines/cell culture

The human OC cell lines A2780, CAOV3, and SKOV3 were purchased from the National Infrastructure of Cell Line Resources in China (Beijing, China). Cells were cultured in DMEM or RPMI-1640 medium with 10% FBS (Thermo Fisher Scientific, Inc.) and incubated at 37°C with 5% CO2. To block Notch signaling in OC cells, the γ-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT, #HY-13027; MedChem Express) dissolved in DMSO was added in the medium.

Clinical samples

Thirty-five epithelial serous OC and 16 pathologically confirmed normal ovary tissues were collected from patients (medium age 54 years; range 30–72 years and medium age 51 years; range 35–64 years, respectively) undergoing surgery at the Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (Beijing, China) from July 2018 to June 2019. All OC cases were diagnosed by pathological evaluation and had complete clinical information. All tissue samples were immediately frozen in liquid nitrogen after resection from patients and stored at −80°C for RNA extraction. This study was conducted according to the ethical guidelines of the 1975 Declaration of Helsinki and approved by the Ethics Committee of Peking Union Medical College Cancer Hospital (Beijing, China) (grant no: NCC2017G-115). Written informed consent was obtained from all of the subjects.

The OC samples were divided into high and low expression of miR-1299 and NOTCH3 groups by the cut-off value, which was defined as the cohort median.

Oligonucleotide transfection, plasmid construction, and lentiviral infection

miR-1299 mimics, inhibitors, and scramble miRNA controls were purchased from GenePharma (Shanghai, China). The lentivirus-containing short hairpin RNA (shRNA) targeting TUG1 or NOTCH3, and the plasmid expressing the active intracellular domain of NOTCH3 (NICD3) were purchased from GeneChem (Shanghai, China). The wild-type and mutated 3′UTR sequence of NOTCH3 was cloned into the pmirGLO vector (Promega) to construct recombinant plasmids named pmirGLO-NOTCH3-WT and pmirGLO-NOTCH3-MUT. Sequences of lncRNA TUG1 and XIST containing the predictive binding sites of miR-1299 and containing point mutations at the site region were cloned into the pmirGLO vector and were named pmirGLO-TUG1-WT, pmirGLO-XIST-WT, and pmirGLO-TUG1-MUT. OC cells were transfected with oligonucleotides using Lipofectamine® RNAiMAX reagent (Thermo Fisher Scientific, Inc.) and plasmids using TurboFect™ transfection reagent (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions.

RNA extraction and RT-qPCR

Total RNA was extracted from OC tissues and cultured cell lines using TRIzol reagent (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. mRNA and lncRNA levels were quantified with RT-qPCR using the SYBR Premix Ex Taq reverse transcription PCR kit (Takara, China), and β-actin was used as an internal control. miRNA was quantified with RT-qPCR using the hairpin-it™ microRNA and U6 snRNA normalization RT-PCR quantitation kit (GenePharma). Each assay was carried out in triplicates in a Light Cycler 480 Instrument (Roche), and the relative expression of mRNA and miRNA was calculated using the 2−ΔΔCq method (33). The primers for RT-qPCR are shown in Table SI.

Cell cycle, proliferation, and colony formation assays

For cell cycle analysis, cells were collected and adjusted to 1×106/ml 48 h after transfection. Ethanol (70%) was used to fix the cells, and RNase A (100 µg/ml) was used to remove RNAs. Finally, cells were stained with propidium iodide (PI) (50 µg/ml) at room temperature for 30 min and analyzed using LSRII flow cytometry (BD Biosciences). Cell viability was detected using the Cell Counting Kit-8 (CCK-8) (Dojindo, Japan) according to the manufacturer's instructions. Twenty-four hours after transfection, the cells were seeded in 96-well plates at 3,000 cells/well. The proliferative ability of cells was determined at 0, 24, 48, 72 and 96 h by measuring the absorbance values at a wavelength of 450 nm. For colony formation assay, OC cells (800 cells per well) were seeded into 6-well plates 24 h after transfection. After 10 days of incubation, the cells were fixed in methanol and stained with 0.1% crystal violet. The colonies were counted using a GBOX F3 gel documentation system (Syngene, Cambridge, UK).

Immunofluorescence (IF) assay

A total of 10,000 cells were seeded on glass coverslips in 24-well plates and grown overnight. For the EdU incorporation assay, 10 µM EdU was used to treat the cells, and an EdU cell proliferation kit (Sangon) was used for measurement. Cellular nuclei were stained using 5 µg/ml Hoechst 33342 at 26°C for 30 min. Images were observed under a DMI 4000 confocal laser scanning microscope at ×200 magnification (Leica, Frankfurt, Germany).

Luciferase reporter assay

Cells were seeded in 96-well plates at 1.5×104 cells/well. When the cells reached 60% confluence, vectors containing the wild-type or mutant 3′UTR of NOTCH3 and wild-type or mutant binding site sequence of TUG1 were co-transfected with miR-1299 mimics and scramble miRNA using Turbofect (Thermo Fisher Scientific, Inc.). Forty-eight hours after transfection, luciferase activity was measured using the Dual-Luciferase Reporter assay system (Promega) and expressed as the ratio of firefly and Renilla luciferase activities.

Western blotting

Proteins were extracted from cells or tissues using RIPA lysis buffer (Solarbio) with protease inhibitors and phosphatase inhibitors. A total of 30 µg proteins were loaded per lane, separated on 10% SDS-PAGE gels, and blotted on polyvinylidene difluoride membrane. After being blocked with 5% skim milk for 2 h at room temperature, the membranes were incubated with primary antibodies (dilution 1:1,000) overnight at 4°C. The primary antibodies used in this study were NOTCH3 antibody (product code ab23426; Abcam) and GAPDH (cat. no. 2118; Cell Signaling Technology, Inc.). Anti-mouse IgG, peroxidase-linked antibody was used as secondary antibody (dilution 1:2,500; cat. no. 5174; Cell Signaling Technology, Inc.) and was incubated at room temperature for 1 h. The blots were detected using a chemiluminescence kit (cat. no. 34577; Thermo Fisher Scientific, Inc.) and imaged using MiniChemi 610 system (Sage Creation Science, Co., Ltd.).

In vivo animal experiments

Eight four-week-old female BALB/c nude mice (body weight range, 17.1–18.2 g) were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd. and randomly divided into two groups. The mice were housed with filtered air, 12 h light/dark cycle, constant temperature (25°C) and had free access to sterilized food and water. After 24-h transfection, 2×106 cells containing miR-1299 or NC agomir were infected into the right armpit of the mice. miR-1299 agomir or NC agomir (RiboBio) was directly injected into the implanted tumor at the dose of 2 nmol/30 µl phosphate-buffered saline (PBS) per mouse every 6 days for 6 times. Tumor growth was monitored by measuring the tumor volume (V) every 6 days with a Vernier caliper and calculated as: V=length × width2/2. All the mice were anesthetized with sodium pentobarbital (500 mg/kg, intraperitoneally) and sacrificed by cervical vertebra dislocation on day 42 or when the tumor volume reached the threshold of 1,500 mm3, and tumors were weighed and snap-frozen for protein and RNA extraction.

Animal experiments were conducted with the approval of the Animal Ethics Committee of Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (ACC2019A060) and in accordance with the Guide for the Care and Use of Laboratory Animals by the US National Institutes of Health.

Statistical analysis

SPSS software version 25.0 (IBM Corp.) and GraphPad Prism 8 (GraphPad Software, Inc.) were used for statistical analysis. Each experiment was performed at least in triplicate, and numerical data are expressed as means ± SD. The difference in clinicopathological features between two groups was determined by independent-sample Student's t-test or Mann-Whitney U test for continuous variables and the Chi-square test or Fisher exact test for categorical variables. One-way ANOVA analysis with Tukey post hoc test was used for comparisons among multiple groups. Pearson's correlation analysis was used to examine the relationship between two gene expression levels. A value of P<0.05 was indicative of statistical significance.

Results

miR-1299 is a negative regulator of NOTCH3 in OC with clinical significance

To investigate the potential miRNAs that regulate NOTCH3, we firstly performed a bioinformatics analysis using the miRWalk database (34), which integrated predicted gene-miRNA target information from 13 databases and compared the results with miRNAs reported to be significantly downregulated in previously published miRNA profiles (NCBI/GEO/GSE47841) (35) (Table SII). The two screening methods overlapped on only 11 miRNAs among which miR-1299 had the highest score in miRWalk. Therefore, we focused our subsequent analysis on miR-1299 as a putative regulator of NOTCH3 in OC. We measured the level of mature miR-1299 and NOTCH3 in 35 fresh OC tissues and 16 normal ovarian tissues, as well as in four OC cell lines. miR-1299 expression was significantly downregulated in the OC tissues compared with that in the normal tissue (P<0.05, Fig. 1A). There was an inverse correlation between the level of miR-1299 and NOTCH3 both in OC tissues (Fig. 1B) and in OC cell lines (Fig. 1C).

To confirm the regulation of miR-1299 on NOTCH3 expression, we first transfected miR-1299 mimics and NC scramble into miR-1299 low-expressing cell A2780 and CAOV3. RT-qPCR results showed that NOTCH3 and its pathway genes were largely reduced in the A2780 (Fig. 1D) and CAOV3 cells (Fig. S1) when transfected with miR-1299 mimics, and western blotting results confirmed the decreased protein level of NOTCH3 (Fig. 1E). In turn, when we knocked down NOTCH3 or overexpressed NICD3 in OC cells, we observed a subsequent increase or decrease in the miR-1299 level (Fig. S2). Furthermore, we cloned wild-type 3′UTR sequence of NOTCH3 and 3′UTR containing point mutations in the putative binding sites into a luciferase reporter plasmid (pmirGLO-NOTCH3-WT and pmirGLO-NOTCH3-MUT) (Fig. 1F). Both plasmids were co-transfected with miR-1299 mimics or NC mimics separately in A2780 cells. The results showed that overexpression of miR-1299 suppressed luciferase activity significantly in cells transfected with pmirGLO-NOTCH3-WT plasmid but not in those transfected with pmirGLO-NOTCH3-mutant vectors (Fig. 1G). These observations suggested that miR-1299 bound directly to the predicted binding sites in the NOTCH3 3′UTR region and negatively regulated NOTCH3 expression.

The association of miR-1299 level and clinicopathological features was also analyzed in OC patients (Table I). Low expression of miR-1299 was significantly correlated with low tumor differentiation. Decreased miR-1299 level was associated with younger age, advanced tumor stage, and necessity to receive neoadjuvant chemotherapy before surgery; however, the association was not statistically significant because of the limited number of cases. Thus, we considered miR-1299 as a regulator of NOTCH3 and hypothesized that decreased expression of miR-1299 promoted OC progression and development.

Table I.

Association between miR-1299 levels and clinicopathological characteristics of the OC patients.

Table I.

Association between miR-1299 levels and clinicopathological characteristics of the OC patients.

miR-1299 expression

CharacteristicsHigh expression (n=17)Low expression (n=18)P-value
Mean age (years)55.2±2.852.9±1.80.121
Tumor stage
  I–II  3  00.057
  III1313
  IV  1  5
Tumor differentiation 0.045
  High/median  4  0
  Low1318
Nodal metastasis 0.305
  No12  9
  Yes  5  9
Neoadjuvant chemotherapy 0.060
  No1510
  Yes  2  8
Serum CA125 level at initial diagnosis534.1 (108.0, 1,362.0)1017.3 (197.4, 1,676.0)0.211

[i] High and low expression of miR-1299 was determined by the cut-off value for miR-1299, which was defined as the cohort median. OC, ovarian cancer; CA125, cancer antigen 125. Significant P-values (P<0.05) are indicated in bold print.

miR-1299 inhibits OC cell proliferation, colony formation, and cell cycle in vitro

Subsequently, we investigated the role of miR-1299 in OC by transfecting miR-1299 mimics in OC cell lines A2780 and CAOV3 (Fig. 2A), both of which had an endogenous low miR-1299 expression. Overexpression of miR-1299 in OC cells significantly reduced cell proliferation, as observed with a CCK-8 assay compared to cells transfected with scrambled miRNA (NC mimic) (Fig. 2B). Colony formation assay showed that treatment with miR-1299 mimics decreased the number of colonies formed by OC cell lines (Fig. 2C). EdU incorporation assay revealed that miR-1299 inhibited DNA synthesis in cell proliferation (Fig. 2D). Additionally, cell cycle analysis showed that transfection of miR-1299 mimics significantly blocked the cells in the G0/G1 phase, accompanied by a smaller population in S and G2 phases (Fig. 2E).

miR-1299 suppresses tumor growth in vivo

We further examined the impact of miR-1299 on OC in a xenograft nude mouse model. A2780 cells transfected with miR-1299 or NC mimics were implanted subcutaneously in BALB/c nude mice. Starting on day 8 post-implantation, miR-1299 or NC mimics were injected intratumorally every 6 days for 6 times. In vivo tumor growth was evaluated by measuring tumor volume and final weight. We observed that treatment with miR-1299 mimics significantly inhibited tumor growth in vivo. The tumor volume and weight were significantly lower in the miR-1299 mimic group than in the NC group (Fig. 3A-C), substantiating the tumor suppressor function of miR-1299 in OC tumorigenesis. Moreover, decreased NOTCH3 protein level in the xenograft tissues with overexpression of miR-1299 was further confirmed (Fig. 3D).

lncRNA TUG1 functions as a sponge of miR-1299 and promotes cell proliferation in OC

To investigate the potential regulators of NOTCH3 by ceRNA mechanism, we first performed a comprehensive bioinformatics analysis using predicted miR-1299-targeted lncRNAs from DIANA databases (36), and OC-relevant lncRNAs from Starbase database (37) and lncRNA disease database (38) (Fig. 4A). The three-screening data overlapped on only two lncRNAs, TUG1 and XIST (X-inactive specific transcript). Therefore, we constructed luciferase reporter plasmids containing the two highest scoring putative miR-1299 binding sites in TUG1 (pmirGLO-TUG1-WT1 and pmirGLO-TUG1-WT2) and XIST (pmirGLO-XIST-WT1 and pmirGLO-XIST-WT2). After co-transfecting the plasmids with miR-1299 mimics or NC mimics in A2780, we found that overexpression of miR-1299 suppressed luciferase activity significantly in two wild-type plasmids of TUG1 (Fig. 4B and C) but not in those of XIST (Fig. S3). We also constructed plasmids carrying point mutations in either binding site of TUG1 (pmirGLO-TUG1-MUT1 and pmirGLO-TUG1-MUT2), and no significant changes in luciferase activity was observed in cells transfected with the two mutant plasmids (Fig. 4B and C). RT-qPCR further revealed that miR-1299 could be upregulated in OC cells after knockdown of TUG1 by lentiviral shRNA particles (Fig. 4D and E). Taken together, these results indicated that the lncRNA TUG1 was a direct sponge of miR-1299 in OC.

We next evaluated the potential tumorigenicity of TUG1 in OC. As expected, downregulation of TUG1 by shRNA resulted in the inhibition of OC cells in terms of cell proliferation, colony formation, EdU incorporation, and cell cycle, which could be partially rescued by inhibition of miR-1299 (Fig. 4F-I). Moreover, knockdown of TUG1 reduced NOTCH3 expression in OC cells, and miR-1299 inhibitors partially rescued the NOTCH3 level (Fig. 4J). This suggested that TUG1, acting as a ceRNA, affects NOTCH3 expression and promotes cell proliferation by sponging miR-1299.

TUG1 is a potential target of NOTCH3 and forms a miR-1299/NOTCH3/TUG1 feedback loop

In the NOTCH signaling pathway, after the cleavage of γ-secretase, the intracellular domain of NOTCH3 protein (NICD3) is translocated to the nucleus and binds the transcription factor complex RBP Jκ (39). When we analyzed the transcriptional start site (TSS, −2 kb to +1 kb) of TUG1 in the JASPAR CORE database (40), we found 5 RBP Jκ binding motifs around its TSS (Fig. 5A), indicating that TUG1 may be directly regulated by NOTCH3. Additionally, there was a significant positive correlation between the expression of TUG1 and NOTCH3 both in our tissues and in the TCGA database (41) (Fig. 5B and C). When we treated A2780 cells with DAPT, an inhibitor of γ-secretase, that can block the NOTCH signaling pathway without interference in NOTCH3 mRNA activity, we found a subsequent decrease in TUG1 expression in a concentration gradient manner (Fig. 5D). Therefore, we considered TUG1 as a potential downstream target of NOTCH3, and that there may exist a miR-1299/NOTCH3/TUG1 feedback loop in the development of OC (Fig. 5E).

Discussion

Recently, miR-1299 was found to be downregulated in several types of cancer and to act as a tumor suppressor. Zhu et al (42) showed that in hepatocellular cancer, miR-1299 overexpression inhibited cell proliferation and arrested the cell cycle in the G0/G1 phase, while miR-1299 knockdown promoted cell proliferation and accelerated G1/S transition. In breast cancer, miR-1299 was found to inhibit CDK6 expression and to bind to its 3′UTR region, suppressing cell proliferation and migration ability (43). A similar tumor suppressive role of miR-1299 was found in esophageal squamous cell carcinoma (44), prostate cancer (45), triple-negative breast cancer (46), and cholangiocarcinoma (47). In this study, we demonstrated that miR-1299 was significantly decreased in OC tissues and was associated with clinical features, and overexpression of miR-1299 inhibited tumor growth both in vitro and in vivo, suggesting that miR-1299 also acted as a tumor suppressor in ovarian cancer (OC). In addition, we first demonstrated that miR-1299 was a negative regulator of NOTCH3, a vital oncogene in OC. Transcriptome analysis showed that suppression of NOTCH signaling in ovarian and breast cancer cells led to downregulation of genes in pathways involved in cell-cycle regulation (48). Both NOTCH3 siRNA and pathway inhibitors caused cell cycle arrest by reducing cyclin D1 and cyclin D3 levels while elevating p21 and p27 levels. In this study, we observed a similar cellular phenotype and protein change (data not shown) after transfection of miR-1299. However, there is also a limitation to our study as we used miR-1299 mimics to transfect two cell lines, and did not use inhibitors in the functional experiments considering the low expression of miR-1299 in OC.

The lncRNA taurine upregulated gene 1 (TUG1), a 7.1-kb lncRNA, was first discovered in a genomic screen for genes upregulated by taurine treatment in mouse retinal cells (49). Recently, TUG1 was characterized as a new oncogene and found to be upregulated in multiple human cancers (5056). TUG1 was upregulated in OC tissues and cells and was positively correlated with advanced disease and poor prognosis. Knockdown of TUG1 significantly inhibited cell proliferation and epithelial-mesenchymal transition (EMT) and induced cell apoptosis in OC (5658). The biological functions of lncRNAs largely rely on their distinct subcellular localization. Cytoplasmic lncRNAs can regulate mRNA stability or translation by acting as sponges for miRNAs (59). TUG1 localizes both in the nucleus and the cytoplasm, indicating that it could have a multi-oncogenic role. Using bioinformatics analyses and luciferase reporter assays, our study provided the first evidence that TUG1 directly bound to and inhibited miR-1299 expression. Knockdown of TUG1 was found to inhibit cell proliferation, colony formation, and cell cycle progression, which could be partially rescued by inhibition of miR-1299. Our observations were consistent with previous studies and confirmed the involvement of TUG1 in the ceRNA mechanism. Notably, there have been several other miRNAs negatively regulated by TUG1 in cancers (6062), among which quite a few miRNAs have been reported to have a tumor suppressive role in OC (6365). TUG1 could sponge more than one miRNA in OC and affect the development of OC through different signaling pathways.

Interestingly, we found that TUG1 possessed five RBP Jk motifs around its promoter region, which can be bound by the NOTCH intracellular domain and trigger transcriptional activation by classical NOTCH signaling pathway. Additionally, treatment with DAPT, an inhibitor of γ-secretase, in OC cells decreased the TUG1 level in a concentration gradient-dependent manner. Therefore, we hypothesized that there could be a miR-1299/NOTCH3/TUG1 feedback loop in OC development. Downregulation of miR-1299 in OC upregulated NOTCH3 level, and overexpression of NOTCH3 activated the expression of various oncogenes by the NOTCH signaling pathway, including the downstream molecule TUG1. In addition, overexpression of TUG1 was a sponge for miR-1299, triggering a positive feedback in tumorigenesis. A recent study in glioma showed that TUG1 was a NOTCH-regulated lncRNA (66), which confirmed our hypothesis. However, we have provided preliminary evidence on this feedback loop hypothesis. The direct regulation of TUG1 by NOTCH3 at the molecular level requires further experimental evidence such as RIP or RNA pulldown results. Therefore, further confirmation of the miR-1299/NOTCH3/TUG1 feedback loop, as well as its role in other important issues, including stemness and drug-resistance in OC, will be the main aim of our future work.

To date, the possible regulators and mechanisms underlying NOTCH3-mediated regulatory behaviors in OC have not been completely elucidated. Our study showed that miR-1299 is a novel negative regulator of NOTCH3 and is downregulated in OC. Overexpression of miR-1299 was found to play a tumor suppressor role both in vitro and in vivo by partially inhibiting cell proliferation. lncRNA TUG1 acted as a sponge for miR-1299 and promoted cell proliferation by upregulating NOTCH3. TUG1 was also a potential target of NOTCH3, forming a miR-1299/NOTCH3/TUG1 feedback loop in OC cells. Our findings improve the understanding of OC pathogenesis and facilitate the development of miRNA- and lncRNA-targeted diagnostics and therapeutics against this disease.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgement

Not applicable.

Funding

This work was supported by grants from the National Natural Science Foundation of China (grant no. 81772272) and the Graduate Innovation Fund of Peking Union Medical College (grant no. 2018-1002-01-27).

Availability of data and materials

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

Authors' contributions

YP, DZ and WC designed the research and analyzed the data. YP, KL and XL performed the cell function experiments. YP and XD performed the animal experiments. YW, WW and NL collected the OC and normal ovary tissues and reviewed the clinical features. YP prepared the images and drafted the original manuscript. WC and DZ reviewed and edited the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The present study was approved by the Research Ethics Committee of the Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing [grant no: NCC2017G-115], and informed consent was obtained from all participating patients. Animal experiments were conducted with the approval of the Animal Ethics Committee of Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (ACC2019A060) and in accordance with the Guide for the Care and Use of Laboratory Animals by the US National Institutes of Health.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

OC

ovarian cancer

NOTCH3

notch receptor 3

lncRNA

long noncoding RNA

miRNA

microRNA

ceRNA

competing endogenous RNA

NICD3

NOTCH3 intracellular domain

TSS

transcriptional start site

EMT

epithelial-mesenchymal transition

TUG1

taurine upregulated gene 1

References

1 

Torre LA, Trabert B, DeSantis CE, Miller KD, Samimi G, Runowicz CD, Gaudet MM, Jemal A and Siegel RL: Ovarian cancer statistics, 2018. CA Cancer J Clin. 68:284–296. 2018. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Zheng RS, Sun KX, Zhang SW, Zeng HM, Zou XN, Chen R, Gu XY, Wei WW and He J: Report of cancer epidemiology in China, 2015. Zhonghua Zhong Liu Za Zhi. 41:19–28. 2019.(In Chinese). PubMed/NCBI

4 

Marchetti C, Palaia I, De Felice F, Musella A, Donfracesco C, Vertechy L, Romito A, Piacenti I, Musio D, Muzii L, et al: Tyrosine-kinases inhibitors in recurrent platinum-resistant ovarian cancer patients. Cancer Treat Rev. 42:41–46. 2016. View Article : Google Scholar : PubMed/NCBI

5 

du Bois A, Reuss A, Pujade-Lauraine E, Harter P, Ray-Coquard I and Pfisterer J: Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer: A combined exploratory analysis of 3 prospectively randomized phase 3 multicenter trials: By the Arbeitsgemeinschaft Gynaekologische Onkologie Studiengruppe Ovarialkarzinom (AGO-OVAR) and the Groupe d'Investigateurs Nationaux Pour les Etudes des Cancers de l'Ovaire (GINECO). Cancer. 115:1234–1244. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Zang RY, Harter P, Chi DS, Sehouli J, Jiang R, Tropé CG, Ayhan A, Cormio G, Xing Y, Wollschlaeger KM, et al: Predictors of survival in patients with recurrent ovarian cancer undergoing secondary cytoreductive surgery based on the pooled analysis of an international collaborative cohort. Br J Cancer. 105:890–896. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Ottevanger PB: Ovarian cancer stem cells more questions than answers. Semin Cancer Biol. 44:67–71. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Mirza MR, Monk BJ, Herrstedt J, Oza AM, Mahner S, Redondo A, Fabbro M, Ledermann JA, Lorusso D, Vergote I, et al: Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med. 375:2154–2164. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Lee EK and Konstantinopoulos PA: Combined PARP and immune checkpoint inhibition in ovarian cancer. Trends Cancer. 5:524–528. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Mitsuhashi Y, Horiuchi A, Miyamoto T, Kashima H, Suzuki A and Shiozawa T: Prognostic significance of Notch signalling molecules and their involvement in the invasiveness of endometrial carcinoma cells. Histopathology. 60:826–837. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Cancer Genome Atlas Research Network, . Integrated genomic analyses of ovarian carcinoma. Nature. 474:609–615. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Park JT, Li M, Nakayama K, Mao TL, Davidson B, Zhang Z, Kurman RJ, Eberhart CG, Shih IeM and Wang TL: Notch3 gene amplification in ovarian cancer. Cancer Res. 66:6312–6318. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Liu Z, Yun R, Yu X, Hu H, Huang G, Tan B and Chen T: Overexpression of Notch3 and pS6 is associated with poor prognosis in human ovarian epithelial cancer. Mediators Inflamm. 2016:59534982016. View Article : Google Scholar : PubMed/NCBI

14 

Jung SG, Kwon YD, Song JA, Back MJ, Lee SY, Lee C, Hwang YY and An HJ: Prognostic significance of Notch 3 gene expression in ovarian serous carcinoma. Cancer Sci. 101:1977–1983. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Park JT, Chen X, Tropè CG, Davidson B, Shih IeM and Wang TL: Notch3 overexpression is related to the recurrence of ovarian cancer and confers resistance to carboplatin. Am J Pathol. 177:1087–1094. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Hu W, Liu T, Ivan C, Sun Y, Huang J, Mangala LS, Miyake T, Dalton HJ, Pradeep S, Rupaimoole R, et al: Notch3 pathway alterations in ovarian cancer. Cancer Res. 74:3282–3293. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Gupta N, Xu Z, El-Sehemy A, Steed H and Fu Y: Notch3 induces epithelial-mesenchymal transition and attenuates carboplatin-induced apoptosis in ovarian cancer cells. Gynecol Oncol. 130:200–206. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al: Landscape of transcription in human cells. Nature. 489:101–108. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Derrien T, Guigó R and Johnson R: The long non-coding RNAs: A new (P)layer in the ‘Dark Matter’. Front Genet. 2:1072012. View Article : Google Scholar : PubMed/NCBI

20 

Yang Y, Ishak Gabra MB, Hanse EA, Lowman XH, Tran TQ, Li H, Milman N, Liu J, Reid MA, Locasale JW, et al: MiR-135 suppresses glycolysis and promotes pancreatic cancer cell adaptation to metabolic stress by targeting phosphofructokinase-1. Nat Commun. 10:8092019. View Article : Google Scholar : PubMed/NCBI

21 

Tang W, Zhou W, Xiang L, Wu X, Zhang P, Wang J, Liu G, Zhang W, Peng Y, Huang X, et al: The p300/YY1/miR-500a-5p/HDAC2 signalling axis regulates cell proliferation in human colorectal cancer. Nat Commun. 10:6632019. View Article : Google Scholar : PubMed/NCBI

22 

Huang T, Wan X, Alvarez AA, James CD, Song X, Yang Y, Sastry N, Nakano I, Sulman EP, Hu B and Cheng SY: MIR93 (microRNA-93) regulates tumorigenicity and therapy response of glioblastoma by targeting autophagy. Autophagy. 15:1100–1111. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Zhang Z, Zhang L, Wang B, Wei R, Wang Y, Wan J, Zhang C, Zhao L, Zhu X, Zhang Y, et al: MiR-337-3p suppresses proliferation of epithelial ovarian cancer by targeting PIK3CA and PIK3CB. Cancer Lett. 469:54–67. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Tung CH, Kuo LW, Huang MF, Wu YY, Tsai YT, Wu JE, Hsu KF, Chen YL and Hong TM: MicroRNA-150-5p promotes cell motility by inhibiting c-Myb-mediated Slug suppression and is a prognostic biomarker for recurrent ovarian cancer. Oncogene. 39:862–876. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Lin C and Yang L: Long noncoding RNA in cancer: Wiring signaling circuitry. Trends Cell Biol. 28:287–301. 2018. View Article : Google Scholar : PubMed/NCBI

26 

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

27 

Schmitt AM and Chang HY: Long noncoding RNAs in cancer pathways. Cancer Cell. 29:452–463. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Huarte M: The emerging role of lncRNAs in cancer. Nat Med. 21:1253–1261. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Wang CJ, Zhu CC, Xu J, Wang M, Zhao WY, Liu Q, Zhao G and Zhang ZZ: The lncRNA UCA1 promotes proliferation, migration, immune escape and inhibits apoptosis in gastric cancer by sponging anti-tumor miRNAs. Mol Cancer. 18:1152019. View Article : Google Scholar : PubMed/NCBI

30 

Lu W, Zhang H, Niu Y, Wu Y, Sun W, Li H, Kong J, Ding K, Shen HM, Wu H, et al: Long non-coding RNA linc00673 regulated non-small cell lung cancer proliferation, migration, invasion and epithelial mesenchymal transition by sponging miR-150-5p. Mol Cancer. 16:1182017. View Article : Google Scholar : PubMed/NCBI

31 

Du Z, Sun T, Hacisuleyman E, Fei T, Wang X, Brown M, Rinn JL, Lee MG, Chen Y, Kantoff PW and Liu XS: Integrative analyses reveal a long noncoding RNA-mediated sponge regulatory network in prostate cancer. Nat Commun. 7:109822016. View Article : Google Scholar : PubMed/NCBI

32 

Zheng ZQ, Li ZX, Zhou GQ, Lin L, Zhang LL, Lv JW, Huang XD, Liu RQ, Chen F, He XJ, et al: Long noncoding RNA FAM225A promotes nasopharyngeal carcinoma tumorigenesis and metastasis by acting as ceRNA to sponge miR-590-3p/miR-1275 and upregulate ITGB3. Cancer Res. 79:4612–4626. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Sticht C, De La Torre C, Parveen A and Gretz N: miRWalk: An online resource for prediction of microRNA binding sites. PLoS One. 13:e02062392018. View Article : Google Scholar : PubMed/NCBI

35 

Vilming Elgaaen B, Olstad OK, Haug KB, Brusletto B, Sandvik L, Staff AC, Gautvik KM and Davidson B: Global miRNA expression analysis of serous and clear cell ovarian carcinomas identifies differentially expressed miRNAs including miR-200c-3p as a prognostic marker. BMC Cancer. 14:802014. View Article : Google Scholar : PubMed/NCBI

36 

Paraskevopoulou MD, Georgakilas G, Kostoulas N, Reczko M, Maragkakis M, Dalamagas TM and Hatzigeorgiou AG: DIANA-LncBase: Experimentally verified and computationally predicted microRNA targets on long non-coding RNAs. Nucleic Acids Res. 41((Database Issue)): D239–D245. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Li JH, Liu S, Zhou H, Qu LH and Yang JH: starBase v2.0: Decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 42((Database Issue)): D92–D97. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Chen G, Wang Z, Wang D, Qiu C, Liu M, Chen X, Zhang Q, Yan G and Cui Q: LncRNADisease: A database for long-non-coding RNA-associated diseases. Nucleic Acids Res. 41((Database Issue)): D983–D986. 2013.PubMed/NCBI

39 

Groeneweg JW, Foster R, Growdon WB, Verheijen RH and Rueda BR: Notch signaling in serous ovarian cancer. J Ovarian Res. 7:952014. View Article : Google Scholar : PubMed/NCBI

40 

Khan A, Fornes O, Stigliani A, Gheorghe M, Castro-Mondragon JA, van der Lee R, Bessy A, Chèneby J, Kulkarni SR, Tan G, et al: JASPAR 2018: Update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 46:D12842018. View Article : Google Scholar : PubMed/NCBI

41 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Zhu H, Wang G, Zhou X, Song X, Gao H, Ma C, Chang H, Li H, Liu FF, Lu J and Ma J: miR-1299 suppresses cell proliferation of hepatocellular carcinoma (HCC) by targeting CDK6. Biomed Pharmacother. 83:792–797. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Liu LH, Tian QQ, Liu J, Zhou Y and Yong H: Upregulation of hsa_circ_0136666 contributes to breast cancer progression by sponging miR-1299 and targeting CDK6. J Cell Biochem. 120:12684–12693. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Meng L, Liu S, Ding P, Chang S and Sang M: Circular RNA ciRS-7 inhibits autophagy of ESCC cells by functioning as miR-1299 sponge to target EGFR signaling. J Cell Biochem. 121:1039–1049. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Morgan SL, Wyant GA and Dinulescu DM: ‘Take it up a NOTCH’: Novel strategies for cancer therapy. Cell Cycle. 12:191–192. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Sang M, Meng L, Liu S, Ding P, Chang S, Ju Y, Liu F, Gu L, Lian Y and Geng C: Circular RNA ciRS-7 maintains metastatic phenotypes as a ceRNA of miR-1299 to target MMPs. Mol Cancer Res. 16:1665–1675. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Shah MM, Zerlin M, Li BY, Herzog TJ, Kitajewski JK and Wright JD: The role of Notch and gamma-secretase inhibition in an ovarian cancer model. Anticancer Res. 33:801–808. 2013.PubMed/NCBI

48 

Chen X, Thiaville MM, Chen L, Stoeck A, Xuan J, Gao M, Shih IeM and Wang TL: Defining NOTCH3 target genes in ovarian cancer. Cancer Res. 72:2294–2303. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Young TL, Matsuda T and Cepko CL: The noncoding RNA taurine upregulated gene 1 is required for differentiation of the murine retina. Curr Biol. 15:501–512. 2005. View Article : Google Scholar : PubMed/NCBI

50 

Guo S, Zhang L, Zhang Y, Wu Z, He D, Li X and Wang Z: Long non-coding RNA TUG1 enhances chemosensitivity in non-small cell lung cancer by impairing microRNA-221-dependent PTEN inhibition. Aging (Albany NY). 11:7553–7569. 2019.PubMed/NCBI

51 

Yu G, Zhou H, Yao W, Meng L and Lang B: lncRNA TUG1 Promotes cisplatin resistance by regulating CCND2 via epigenetically silencing miR-194-5p in bladder cancer. Mol Ther Nucleic Acids. 16:257–271. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Zhang Z, Wang X, Cao S, Han X, Wang Z, Zhao X, Liu X, Li G, Pan and Lei D: The long noncoding RNA TUG1 promotes laryngeal cancer proliferation and migration. Cell Physiol Biochem. 49:2511–2520. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Barbagallo C, Brex D, Caponnetto A, Cirnigliaro M, Scalia M, Magnano A, Caltabiano R, Barbagallo D, Biondi A, Cappellani A, et al: LncRNA UCA1, upregulated in CRC biopsies and downregulated in serum exosomes, controls mRNA expression by RNA-RNA interactions. Mol Ther Nucleic Acids. 12:229–241. 2018. View Article : Google Scholar : PubMed/NCBI

54 

He C, Liu Z, Jin L, Zhang F, Peng X, Xiao Y, Wang X, Lyu Q and Cai X: lncRNA TUG1-mediated miR-142-3p downregulation contributes to metastasis and the epithelial-to-mesenchymal transition of hepatocellular carcinoma by targeting ZEB1. Cell Physiol Biochem. 48:1928–1941. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Sun J, Hu J, Wang G, Yang Z, Zhao C, Zhang X and Wang J: LncRNA TUG1 promoted KIAA1199 expression via miR-600 to accelerate cell metastasis and epithelial-mesenchymal transition in colorectal cancer. J Exp Clin Cancer Res. 37:1062018. View Article : Google Scholar : PubMed/NCBI

56 

Xu Y, Ge Z, Zhang E, Zuo Q, Huang S, Yang N, Wu D, Zhang Y, Chen Y, Xu H, et al: The lncRNA TUG1 modulates proliferation in trophoblast cells via epigenetic suppression of RND3. Cell Death Dis. 8:e31042017. View Article : Google Scholar : PubMed/NCBI

57 

Kuang D, Zhang X, Hua S, Dong W and Li Z: Long non-coding RNA TUG1 regulates ovarian cancer proliferation and metastasis via affecting epithelial-mesenchymal transition. Exp Mol Pathol. 101:267–273. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Li TH, Zhang JJ, Liu SX and Chen Y: Long non-coding RNA taurine-upregulated gene 1 predicts unfavorable prognosis, promotes cells proliferation, and inhibits cells apoptosis in epithelial ovarian cancer. Medicine (Baltimore). 97:e05752018. View Article : Google Scholar : PubMed/NCBI

59 

Chen LL: Linking long noncoding RNA localization and function. Trends Biochem Sci. 41:761–772. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Ren K, Li Z, Li Y, Zhang W and Han X: Long noncoding RNA taurine-upregulated gene 1 promotes cell proliferation and invasion in gastric cancer via negatively modulating miRNA-145-5p. Oncol Res. 25:789–798. 2017. View Article : Google Scholar : PubMed/NCBI

61 

Wang Y, Yang T, Zhang Z, Lu M, Zhao W, Zeng X and Zhang W: Long non-coding RNA TUG1 promotes migration and invasion by acting as a ceRNA of miR-335-5p in osteosarcoma cells. Cancer Sci. 108:859–867. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Zhao L, Sun H, Kong H, Chen Z, Chen B and Zhou M: The lncRNA-TUG1/EZH2 axis promotes pancreatic cancer cell proliferation, migration and EMT phenotype formation through sponging miR-382. Cell Physiol Biochem. 42:2145–2158. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Sheng Q, Zhang Y, Wang Z, Ding J, Song Y and Zhao W: Cisplatin-mediated down-regulation of miR-145 contributes to up-regulation of PD-L1 via the c-Myc transcription factor in cisplatin-resistant ovarian carcinoma cells. Clin Exp Immunol. 200:45–52. 2020. View Article : Google Scholar : PubMed/NCBI

64 

Tan H, He Q, Gong G, Wang Y, Li J, Wang J, Zhu D and Wu X: miR-382 inhibits migration and invasion by targeting ROR1 through regulating EMT in ovarian cancer. Int J Oncol. 48:181–190. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Zhu X, Li Y, Xie C, Yin X, Liu Y, Cao Y, Fang Y, Lin X, Xu Y, Xu W, et al: miR-145 sensitizes ovarian cancer cells to paclitaxel by targeting Sp1 and Cdk6. Int J Cancer. 135:1286–1296. 2014. View Article : Google Scholar : PubMed/NCBI

66 

Katsushima K, Natsume A, Ohka F, Shinjo K, Hatanaka A, Ichimura N, Sato S, Takahashi S, Kimura H, Totoki Y, et al: Targeting the Notch-regulated non-coding RNA TUG1 for glioma treatment. Nat Commun. 7:136162016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2020
Volume 44 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Pei Y, Li K, Lou X, Wu Y, Dong X, Wang W, Li N, Zhang D and Cui W: miR‑1299/NOTCH3/TUG1 feedback loop contributes to the malignant proliferation of ovarian cancer Corrigendum in /10.3892/or.2023.8533. Oncol Rep 44: 438-448, 2020
APA
Pei, Y., Li, K., Lou, X., Wu, Y., Dong, X., Wang, W. ... Cui, W. (2020). miR‑1299/NOTCH3/TUG1 feedback loop contributes to the malignant proliferation of ovarian cancer Corrigendum in /10.3892/or.2023.8533. Oncology Reports, 44, 438-448. https://doi.org/10.3892/or.2020.7623
MLA
Pei, Y., Li, K., Lou, X., Wu, Y., Dong, X., Wang, W., Li, N., Zhang, D., Cui, W."miR‑1299/NOTCH3/TUG1 feedback loop contributes to the malignant proliferation of ovarian cancer Corrigendum in /10.3892/or.2023.8533". Oncology Reports 44.2 (2020): 438-448.
Chicago
Pei, Y., Li, K., Lou, X., Wu, Y., Dong, X., Wang, W., Li, N., Zhang, D., Cui, W."miR‑1299/NOTCH3/TUG1 feedback loop contributes to the malignant proliferation of ovarian cancer Corrigendum in /10.3892/or.2023.8533". Oncology Reports 44, no. 2 (2020): 438-448. https://doi.org/10.3892/or.2020.7623