IMR Press / FBL / Volume 28 / Issue 10 / DOI: 10.31083/j.fbl2810270
Open Access Original Research
LncRNA PART1 Regulates Ovarian Carcinoma Development via the miR-150-5p/MYB Axis
Jing Wang1,2,†Yun Han3,†Tingting Zhang4Jing Li5,*Boqun Xu1,6,*
Show Less
1 Obstetric and Gynecological Department, The Second Affiliated Hospital of Nanjing Medical University, 210000 Nanjing, Jiangsu, China
2 Obstetric and Gynecological Department, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, 210000 Nanjing, Jiangsu, China
3 Obstetrics and Gynecological Department, The Second Affiliated Hospital of Nantong University, 226000 Nantong, Jiangsu, China
4 Obstetric and Gynecological Department, Suqian People's Hospital of Nanjing Drum-Tower Hospital Group, 223800 Suqian, Jiangsu, China
5 State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 210000 Nanjing, Jiangsu, China
6 Obstetrics and Gynecological Department, Affiliated Sir Run Run Hospital of Nanjing Medical University, 210000 Nanjing, Jiangsu, China
*Correspondence: ljwth@njmu.edu.cn (Jing Li); boqun_xu@njmu.edu.cn (Boqun Xu)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2023, 28(10), 270; https://doi.org/10.31083/j.fbl2810270
Submitted: 1 December 2022 | Revised: 17 February 2023 | Accepted: 23 February 2023 | Published: 27 October 2023
(This article belongs to the Special Issue Diagnostic, Prognostic, and Therapeutic Biomarkers for Ovarian Cancer)
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Over the past few years, there have been many reports on the abnormal expression and functional relevance of long non-coding RNAs (lncRNAs) in tumors. The role played by lncRNAs in epithelial ovarian carcinoma (EOC) remains poorly understood, however the goal of the present work was to study molecular mechanisms that underlie involvement of prostate androgen-regulated transcript 1 (PART1) lncRNA in EOC development. Methods: A total of 25 tumor and 17 normal specimens were obtained from women undergoing surgery between 2015 and 2019 in the Second Affiliated Hospital, Nanjing Medical University. Expression levels for PART1 in EOC tissue and EOC cell lines were assessed using qRT-PCR. Assays for CCK-8, trans-well, colony forming and western blotting were used to investigate PART1, miR-150-5p and MYB (MYB proto-oncogene) for their invovement in EOC cell proliferation, migration and invasion. Luciferase reporter gene assay was also performed to investigate biological functions of PART1, miR-150-5p and MYB in EOC, and an animal xenograft model was employed to test tumorigenicity. Results: PART1 expression was increased in EOC relative to normal cells and correlated with EOC cell proliferation, migration and invasion. PART1 can sponge miR-150, thereby inhibiting growth of EOC by targeting MYB. The xenograft mouse model revealed that PART1 can regulate tumorigenesis in vivo. Conclusions: The PART1/miR-150/MYB axis is involved in EOC pathogenesis and could represent a new target to use in diagnosis and therapy.

Keywords
ovarian carcinoma
long non-coding RNAs
PART1
ceRNA
1. Introduction

Epithelial ovarian carcinoma (EOC) accounts for a large proportion of gynecologic malignancies, with an annual incidence of 3–12 new cases per 100,000 women [1, 2]. The most common treatment for EOC is maximum surgical cytoreduction combined with platinum chemotherapy. Despite the advances in surgery and chemoradiotherapy, 5-year survival rates for EOC patients are still poor due to the relatively late diagnosis of this disease and to acquired drug resistance [3, 4, 5, 6]. It is therefore crucial to identify the molecular mechanism associated with the development of EOC.

A large-scale human genomic sequencing study showed that just 2% is transcribed into RNA that codes for protein. Most of the remaining RNA transcripts are non-coding (ncRNA) [7]. Long ncRNA (lncRNA) is defined as non-coding transcript >200 nucleotides(nt) long [8]. The use of lncRNA as possible biomarkers in different tumor types has been proposed [9, 10]. Several studies have also reported an imbalance between lncRNAs and microRNAs (miRNAs) in lung, hepatic, pancreatic and gastric cancers, and recently in EOC [11, 12, 13, 14, 15, 16]. Other workers have shown that dysregulation between lncRNA and miRNA levels is closely correlated with several biological behaviors in cancer cells. These include aberrant proliferation, invasion and migration of cells, as well as chemical resistance. Such properties could be mediated through the signaling pathway for lncRNA-miRNA-mRNA in what is referred to as the competing endogenous RNA (ceRNA) hypothesis [17, 18, 19]. Expression of the lncRNA HOXD-AS1, is elevated in EOC, for example. This lncRNA was found to facilitate EOC cell invasion and migration, in addition to epithelial-mesenchymal transition, via HOXD-AS1/miR-186-5p/PIK3R3 signaling pathways [20], such as the ADAMTS9-AS2/miR-182-5p/FOXF2 pathway [21].

Here, we identified PART1/miR-150-5p/MYB axis in EOCs was identified, as well as the presence of a ceRNA mechanism. This axis was shown to have a major impact on EOC progression.

2. Material and Methods
2.1 Subjects and EOC Clinical Specimens

Tumor (n = 25) and normal (n = 17) tissue were obtained from women undergoing surgery for EOC between 2015 and 2019 at our institution. None had received preoperative chemoradiotherapy. Histopathology was used to confirm specimens prior to storage in a liquid nitrogen tank at –196 °C until the extraction of RNA. All patients gave informed consent. The major clinicopathological features of cohort are indicated below (Table 1). For the relationship between MYB, miR-150-5p and clinicopathological features, see Supplementary Tables 1,2. Ethical approval for the study was given by the host institute (No. 2017AE02133, approval date December 3, 2017) and the Declaration of Helsinki guidelines were followed.

Table 1.LncRNA PART1 and the Clinicopathological Features of epithelial ovarian carcinoma (EOC) Patients.
Variables Cases (25) LncRNA PART1 expressional level p
Low/Negative High
<3.994 3.994
Age (years) >0.05
55 11 6 5
>55 14 6 8
FIGO stage <0.05
I–II 10 8 2
III–IV 15 4 11
Histological grade <0.05
1–2 13 9 4
3 12 3 9

FIGO stage, International Federation of Gynecology and Obstetrics stage.

2.2 Cell Culture and Treatments

The human cell lines studied here were provided by the Chinese Science Academy (Shanghai, China) and Zhong Qiao Xin Zhou (Shanghai, China). These consisted of normal ovarian surface epithelial cells (IOSE80, ZQ0721, Zhong Qiao Xin Zhou, Shanghai, China) and three EOC cell lines ((A2780, ZQ0486, Zhong Qiao Xin Zhou, Shanghai, China), (SKOV3, TCHu185, Chinese Science Academy, Shanghai, China) and (OVCAR3, TCHu228, Chinese Science Academy, Shanghai, China)). SKOV3 was grown in McCoy’s 5A medium (Gibco, New York, USA) with 10% fetal bovine serum (FBS), while remaining cell lines were grown in Dulbecco’s modified Eagle medium (DMEM) with high-glucose and 10% FBS. Cells were kept at 37 °C in a humidified incubator with 5% CO2 concentration. Mycoplasma testing has been done for the cell lines used. The cell lines used have been authenticated and Short Tandem Repeat (STR) was used for the authentication.

2.3 RNA Isolation for Quantitative RT-PCR (qRT-PCR)

RNA was extracted with Trizol (Invitrogen, Carlsbad, USA) and cDNA for qPCR was prepared using HiScript II SuperMix (Vazyme, Nanjing, China). qRT-PCR (Q6, Life) was carried out with SYBR® Green PCR Master Mix (Applied Biosystems, Waltham, USA) and primer sequences as follow: PART1 forward 5CTCCTGCGGTTTCCCATT3, reverse 5ATCTCACCAGACACCTGCCTAC3; miR-150-5p forward 5CGGGCTCTCCCAACCCTTGT3, reverse 5CAGCCACAAAAGAGCACAAT3; MYB forward 5GGCACACAAGAGACTGGGGA3, reverse 5CGACCTTCCGACGCATTGTA3; glyceraldehyde-3-phosphate dehydrogenase (GAPDH) forward 5ACCCACTCCTCCACCTTTGAC3, reverse 5TGTTGCTGTAGCCAAATTCGTT3; U6 forward 5CTCGCTTCGGCAGCACA3, reverse 5AACGCTTCACGAATTTGCGT3. The cycling conditions were: stage 1 (pre-denaturation): 95 °C, 10 min; stage 2 (cyclic reaction, 40 times): 95 °C for 10 sec; 60 °C for 30 sec; stage 3 (dissolution curve): 95 °C for 15 sec; 60 °C for 60 sec; 95 °C for 15 sec. Expression levels for lncRNA, miRNA and mRNA were detected quantitatively by the 2-ΔΔCt method [22], with GAPDH the internal references.

2.4 Predicting Target Gene

StarBase v3.0 (https://starbase.sysu.edu.cn/index.php) and miRcode (http://www.mircode.org/index.php) were employed to predict potential miRNAs that bind to PART1 [23, 24]. StarBase v3.0 was also used to identify candidate mRNAs that bind to miR-150-5p. This software integrates 5 different miRNA prediction softwares (TargetScan, PicTar, microT, miRmap, miRanda/mirSVR). The GEPIA database was used to analyze the expression levels of the target genes for miR-150-5p [25].

2.5 Lentivirus, siRNA, miR-150-5p Inhibitor and Transfection

PART1-shRNA-lentiviruses (sequence: sh1:GAAC TCAATTACGACTACATA; sh2:CCAGAUGAGACUAC GAUAATT; sh3:GAACAGAGUUGACUUUGUGTT; sh4:GCAAAGUAUCCAAGACCAATT) used for the in vivo experiments were provided by GenePharma (Shanghai, China), as were the siRNAs that target PART1. Transfection of cells was performed with Lipofectamine 3000 (Invitrogen, Carlsbad, USA). MiR-150-5p inhibitors (sequence: CACUGGUACAAGGGUUGGGAGA) were also from GenePharma. Triplicate experiments were performed throughout.

2.6 CCK-8 Assay

CCK-8 assay was used to quantify the proliferation rate of cells. Following inoculation into 96-well culture plates (1.5 × 103 cells/well), cells were grown in 100 µL DMEM containing 10% FBS. CCK-8 solution (10 µL) was added 0, 24, 48, 72 and 96 h after cellular attachment. The 450 nm absorbance was subsequently read following 2 h of incubation by Enzyme label (Multiskan FC, Thermo, Waltham, USA).

2.7 Transwell Assays

Transwell chambers (Millipore, Darmstadt, Germany) used for cell invasion assays were first pretreated with 50 µL of a 1:9 Matrigel/DMEM solution (BD, New Jersey, USA). Subsequently, 1 × 105 cells were dispersed in DMEM without FBS (1 mL) and 200 µL of cellular solution was placed in the upper chamber. Following this, DMEM containing 10% FBS (600 µL) was placed into the lower chamber to act as a chemotactic agent. 48 h later, residual cells in the upper chamber were scraped off, while invading cells were immobilized in 4% paraformaldehyde then dyed in 2% crystal violet. The invading cells were counted by light microscopy (D-35578, Leica, Weztlar, Germany). The experimental procedure used to quantify the migration of cells was identical to the above, but with no Matrigel pretreatment.

2.8 Colony Formation Assay

Cells were dispersed into a single-cell suspension at 48 h after transfected. The colony forming assay was carried out by incubating 1 × 103 cells at 37 °C for two weeks in a culture dish containing 10% FBS medium. The cells were then stabilized, dyed using 0.1% crystal violet, and the colonies counted manually. Experiments were repeated three times for all groups.

2.9 Western Blotting Assay

Five × 105 cells per well were inoculated in 6-well plates, grown for 48 h, then rinsed in PBS and subsequently immersed in ice-cold lysis buffer. The concentration of protein in the lysate was quantified with the bicinchoninic acid (BCA) method (P0012S, Beyotime, Shanghai, China). Protein from each sample was separated with 10% SDS-PAGE then transferred to polyvinylidene fluoride film (162017, Bio-Rad, Hercules, USA). The film was subsequently incubated for 2 h with a solution of 5% non-fat dried milk in TBS with 0.1% Tween-20 to block non-specific proteins. MYB- (1:2000, ab109127, abcam, Cambridgeshire, UK) or GAPDH-specific antibody (1:5000, 60004-1-Ig, Proteintech, Wuhan, China) was then incubated with the polyvinylidene fluoride film for 24 h at 4 °C. The film was subsequently rinsed before incubation for 2 h with horseradish peroxidase (HRP)-conjugated secondary antibody (1:5000, Proteintech, 60004-1-Ig, Wuhan, China) at 37 °C. Immunoblot analysis was performed using an enhanced chemiluminescence reagent (34579, Invitrogen, Carlsbad, USA) and the film was subsequently irradiated with X-rays by WB Imaging Instrument (5260, Tanon, Shanghai, China). Image J software (1.8.0, NIH, USA) was used to analyse protein expression levels, with GAPDH used as the reference.

2.10 Dual-Luciferase Reporter Gene Assay

A2780 and SKOV3 cell lines were grown in 6-well plates. Wild-type (WT) and mutant fragments (MUT) from the 3-untranslated region of PART1 (PART1- WT: GUAAUCCCAGCACUUUGGGAGG; PART1-MUT: GUAAUCCCAGCACUCGCA CGAG) and MYB (MYB-WT: GAAACUUUUCAUGAAUGGGAGA; MYB-MUT: GAAACUUUUCAUGAACACACCA) that were related to the binding site for miR-150-5p were synthesized and added to pMIR-REPORT vectors. Reporter gene assay was conducted as per our previous study [26]. Normalization of luciferase activity was carried out in relation with Renilla luciferase activity.

2.11 Xenograft Model

Female BALB/c nude mice aged 4–5 weeks were obtained from Animal Core Facility, Nanjing Medical University. Each mouse (5 mice per group) received a subcutaneous injection in the right armpit with a 200 µL suspension containing 5 × 105 A2780 ovarian cancer cells. Tumor growth rate was evaluated at regular intervals starting from the 6th day. Tumor size was quantified with vernier calipers, where L was the longest diameter and W the longest transverse diameter perpendicular to the longest diameter when the tumor is viewed as an ellipse. Tumor volume was subsequently estimated with the formula: V = π/6×L×W×W. Animal experimentation conformed with the guidelines for “Animal Research: Reporting of In Vivo Experiments (ARRIVE)”. The animal ethics approval number was 1811051, and the approval date was April 2018.

2.12 Statistical Analyses

Experiments were conducted in triplicate. SPSS 15.0 (IBM, Chicago, USA) was used for statistical analyses, with values expressed as the mean ± standard deviation (SD). One-way-ANOVA analysis of variance was used to evaluate the differences among at least three groups. Least Significance Difference (LSD) was used with ANOVA. Student’s t test was used to determine the differences between groups. Pearson correlation analysis was used to analyse correlations between data. The normal distribution was tested using the Shapiro-Wilk (S-W test) by SPSS. p-values < 0.05 were considered to show statistical significance.

3. Results
3.1 Increased LncRNA PART1 Expression in Ovarian Carcinoma

LncRNA PART1 expression in clinical specimens was quantified by qRT-PCR. Fig. 1A shows the expression was significantly higher in EOC relative to normal ovarian surface epithelial (OSE) tissue. LncRNA PART1 expression was also higher in advanced stage EOC relative to lower stage tumors (Fig. 1B). Finally, the expression level in three EOC cell lines (OVCAR3, SKOV3, A2780) was greater relative to the OSE cell line (IOSE80), particularly in SKOV3 and A2780 cells (Fig. 1C).

Fig. 1.

Elevated expression of lncRNA PART1 in EOC detected with qRT-PCR. (A) PART1 expression in EOC and normal ovarian tissue. (B) PART1 expression in EOCs at different FIGO stages. (C) PART1 expression in EOC (OVCAR3, SKOV3, A2780) and ovarian surface epithelial (OSE) (IOSE80) cells. *p < 0.05, **p < 0.01.

3.2 LncRNA PART1 Knockdown Reduces EOC Cell Proliferation, Migration and Invasion

PART1 expression in SKOV3 and A2780 cells was reduced following transfection with a knockdown vector (sh-PART1). A control vector (sh-NC) was also used, and transfection efficiency confirmed using qRT-PCR. Among them, sh-1, sh-2, sh-3 and sh-4 were compared with sh-NC, respectively, and sh-2 had the best transfection efficiency (Fig. 2A,B). Results from colony forming assays and CCK-8 assays demonstrated that lncRNA PART1 knockdown inhibited EOC cell proliferation (Fig. 2C–E). Furthermore, transwell assay revealed that lncRNA PART1 knockdown also inhibited the migration and invasion ability of cells (Fig. 2F–I). Hence, the above results imply that PART1 enhances the proliferation, migration and invasion of EOC cells in vitro.

Fig. 2.

LncRNA PART1 knockdown reduces the proliferation, migration and invasion of A2780 and SKOV3 cells. (A,B) PART1 expression in cells transfected with sh-PART1 (including sh-PART1-1, sh-PART1-2, h-PART1-3 and sh-PART1-4) as measured by qRT-PCR. (C) Proliferation of cells following transfection with sh-PART1 or sh-NC (control), as determined using colony-forming assay. (D,E) Proliferation of cells following transfection with sh-PART1 or sh-NC, as determined by CCK8 assay. (F,G) Migration of cells following transfection with sh-PART1 or sh-NC, as determined by transwell assay. (H,I) Invasion of cells following transfection with sh-PART1 or sh-NC, shown using transwell assay. *p < 0.05, **p < 0.01.

3.3 LncRNA PART1 Sponges miR-150-5p

The findings from starBase revealed the presence of binding sites between lncRNA PART1 and miR-150-5p (Fig. 3A). Luciferase reporter gene assay also demonstrated that miR-150-5p decreased luciferase activity in lncRNA PART1-WT cells, but not lncRNA PART1-MUT cells (Fig. 3B). miR-150-5p expression was significantly reduced in EOC tissue relative to normal tissue (Fig. 3C). Pearson’s analysis demonstrated an inverse correlation between PART1 and miR-150-5p levels in EOC tissue (Fig. 3D). Similar results to EOC tissues were obtained for miR-150-5p expression in OSE and EOC cells. The expression levels in the three EOC cell lines (OVCAR3, SKOV3, A2780) were lower than those in the OSE80 cell line, especially in SKOV3 and A2780 cells (Fig. 3E). miR-150-5p expression was increased in SKOV3 and A2780 cells in which lncRNA PART1 was down-regulated (Fig. 3F). Together, these results indicate lncRNA PART1 is able to sponge miR-150-5p in EOC cells.

Fig. 3.

LncRNA PART1 can sponge miR-150-5p. (A) Putative binding sites between miR-150-5p and PART1 identified with online databases. (B) miR-150-5p reduces luciferase activity of PART1-WT, but not PART1-MUT, as shown by luciferase reporter gene assay. (C) miR-150-5p expression in EOC and normal ovarian tissue, as quantified by qRT-PCR. (D) Correlation analysis of relative expression levels for PART1 and miR-150-5p in clinical specimens. (E) miR-150-5p expression levels in EOC cells (OVCAR3, SKOV3, A2780) and OSE cells (IOSE80), as measured with qRT-PCR. (F) miR-150-5p expression in A2780 and SKOV3 cells after transfection with sh-PART1 or with sh-NC (negative control), as measured with qRT-PCR. *p < 0.05, **p < 0.01.

3.4 LncRNA PART1 Enhances EOC Progression by Binding to miR-150-5p

Functional assays were performed to determine whether lncRNA PART1 acts in A2780 and SKOV3 cells by binding to and therefore effectively removing miR-150-5p. Transfection efficiencies for miR-150-5p inhibitor, sh-PART1 and corresponding controls were confirmed using qRT-PCR (Fig. 4A,B). CCK-8 assays revealed that lncRNA PART1 down-regulation decreased EOC cell proliferation. This was partially reduced by miR-150-5p inhibitor (Fig. 4C,D). Similarly, results from transwell assays revealed miR-150-5p inhibitor can attenuate inhibition of migration and invasion caused by lncRNA PART1 downregulation (Fig. 4E–H). These findings suggest that lncRNA PART1 can enhance EOC cell proliferation, migration and invasion through binding and thus inactivation of miR-150-5p.

Fig. 4.

LncRNA PART1 enhances the progression of EOC by binding miR-150-5p. (A,B) miR-150-5p expression in A2780 and SKOV3 cells after transfection with miR-150-5p inhibitor, sh-PART1 or the corresponding controls, as determined with qRT-PCR. (C,D) A2780 and SKOV3 cell proliferation after transfection with miR-150-5p inhibitor, sh-PART1 or the corresponding controls, as measured with the CCK8 assay. (E,F) Migration of A2780 and SKOV3 cells following transfection with miR-150-5p inhibitor, sh-PART1 or the corresponding controls, as measured with the transwell assay. (G,H) Invasion of A2780 and SKOV3 cells following transfection with miR-150-5p inhibitor, sh-PART1 or the corresponding controls, as measured with the transwell assay. *p < 0.05, **p < 0.01.

3.5 MiR-150-5p Inhibits OEC Cell Proliferation, Migration and Invasion by Targeting MYB

We investigated miR-150-5p target genes to test the ceRNA hypothesis. 22 genes were screened out by StarBase v3.0. Then three of these genes, MTCH2, MYB and NDC1, were found to be elevated in ovarian cancer tissues relative to normal ovarian tissues through the GEPIA database (Supplementary Fig. 1). MYB is one of the more classical, malignant progression-promoting oncogenes in a variety of tumors, such as: breast, liver, colon and lung cancers [27, 28, 29, 30]. Next, MYB was selected as the object of study (Fig. 5A). The luciferase reporter gene assay revealed that miR-150-5p decreased luciferase activity for MYB-WT, but there was no significant effect for MYB-MUT (Fig. 5B). The expression of MYB was elevated in EOC compared to OSE tissue (Fig. 5C). Moreover, MYB expression was inversely associated miR-150-5p expression in EOC specimens, as shown by Pearson’s correlation analysis (Fig. 5D). Three EOC cell lines (OVCAR3, SKOV3, A2780) were compared with OSE cell line (OSE80), respectively. qRT-PCR also demonstrated that MYB expression in EOC cells was higher than in OSE cells (Fig. 5E). Together, the data indicate that MYB is a target for miR-150-5p. Functional assays showed the impacts of si-MYB on the proliferation, migration and invasion of EOC cells were partly attenuated by miR-150-5p inhibitor (Fig. 6A–H). Overall, these findings demonstrate that miR-150-5p can reduce proliferation, migration and invasion of EOC cells through targeting MYB.

Fig. 5.

MYB is a target for miR-150-5p. (A) Putative binding sites for miR-150-5p on MYB, identified with online databases. (B) miR-150-5p reduces luciferase activity of MYB-WT in EOC cell lines, but not for MYB-MUT, as shown by luciferase reporter assay. (C) MYB expression levels in EOC and normal ovarian tissue, as measured with qRT-PCR. (D) Correlation between MYB and miR-150-5p expression in clinical specimens, as determined using Pearson’s analysis. (E) MYB expression in EOC (OVCAR3, SKOV3, A2780) and OSE (IOSE80) cells, as measured with qRT-PCR. **p < 0.01.

Fig. 6.

MYB enhances the proliferation, migration and invasion of A2780 and SKOV3 cells in vitro. (A,B) MYB expression in A2780 and SKOV3 cells following transfection with si-MYB, or si-MYB plus miR-150-5p inhibitor, as measured with qRT-PCR and Western blotting. (C,D) Proliferation of A2780 and SKOV3 cells following transfection with si-MYB, or si-MYB plus miR-150-5p inhibitor, as measured with the CCK8 assay. (E,F) Migration of A2780 and SKOV3 cells following transfection with si-MYB, or si-MYB plus miR-150-5p inhibitor, as determined using the transwell assay. (G,H) Invasion by A2780 and SKOV3 cells following transfection with si-MYB, or si-MYB plus miR-150-5p inhibitor, as measured using transwell assays. **p < 0.01.

3.6 Knockdown of lncRNA PART1 Suppresses the Growth of Ovarian Carcinoma In Vivo

To investigate whether lncRNA PART1 is involved in EOC growth in vivo, female nude mice received subcutaneous injections of A2780 cells following transfection with sh-PART1 or sh-NC (negative control). Tumor size was estimated every four days and the tumor growth rate determined after 30 days. sh-PART1 was found to suppress cancer growth in the nude mice compared to controls (Fig. 7A–C). In addition, mean tumor volume and weight were smaller with sh-PART1(Fig. 7D,E). Overall, these finding indicate that lncRNA PART1 can enhance the growth of EOC in vivo.

Fig. 7.

MYB promotes in vivo growth of EOC. (A,B) EOC growth in mice injected with A2780 cells that had been transfected with sh-PART1 or with sh-NC. (C–E) PART1 downregulation resulted in a lower tumor growth rate (volume and weight) than observed in controls. **p < 0.01.

4. Discussion

EOC shows a high degree of malignancy in female patients [31]. The lack of obvious symptoms during the initial stages of EOC mean that is easily be ignored by patients. As a consequence, most women are diagnosed with middle and late stages of this cancer, thus greatly increasing the difficulty of treatment [32, 33]. A large body of research has revealed a major role for lncRNAs in the pathogenesis of human malignancies [34, 35]. Novel transcriptome sequencing technology has resulted in the discovery of an ever-increasing number of lncRNAs. There is now strong research evidence showing the involvement of lncRNAs in several biological behaviors including the regulation of gene transcription, epigenetic regulation, ontogenetic regulation, and chromatin modification [36, 37, 38]. Several research groups have demonstrated that progression of several tumor types, including EOC, correlates with aberrant expression of lncRNAs [39]. Some lncRNAs have also been suggested as markers for early cancer detection and diagnosis [40, 41, 42]. PART1 is increasingly recognized to be associated in the pathogenesis of many cancer types. Recently, Zhao et al. [43] published that lncRNA PART1 can suppress the growth of esophageal cancer, while Chen et al. [44] reported it could also increase proliferation of lung cancer cells. Presently, we confirmed that lncRNA PART1 expression was elevated in EOC compared to normal tissue. Knockdown of PART1 expression in EOC cells markedly inhibited EOC cell proliferation, migration and invasion in vitro. An EOC xenograft animal model was also established and was used to show that lncRNA PART1 knockdown lowered the growth rate of xenografts.

Salmena et al. [19] demonstrated that lncRNAs could act as a sponge to absorb miRNAs, thereby indirectly regulating mRNA expression. An increasing number of interrelationships in the lncRNA-miRNA-mRNA ceRNA axis have recently been found. Several investigators have suggested a role for ceRNA during progression of multiple tumor types including bladder, colorectal and cervical cancer [45, 46, 47]. With major advances in sequencing technology, bioinformatics now plays an important role in both foundational and clinical medicine [48, 49]. In the current study, the online databases starbase V3.0 and miRcode were used to identify miR-150-5p as being a downstream target of lncRNA PART1. LncRNA PART1expression was higher in EOC tissue and cells, but this was decreased by miR-150-5p. Functional studies also revealed that lncRNA PART1 had oncogene-like properties in EOCs. Both PART1 sh-RNA and miR-150-5p mimic reduced MYB-WT luciferase activity in a reporter gene assay, while the inhibitory action of PART1 sh-RNA was restored with miR-150 inhibitor. Furthermore, qRT-PCR and Western blot revealed that lncRNA PART1 knockdown reduced MYB expression. This effect was partly attenuated with miR-150 inhibitor. In addition, functional assays showed the effect of sh-PART1 on the proliferation, migration and invasion ability of EOC cells was abolished with miR-150 inhibitors. These results indicate PART1 can act like a ceRNA, thus regulating the miR-150/MYB axis and hence EOC progression.

This study has several limitations. The study cohort of EOC tissues was relatively small and hence the five different EOC subtypes (low grade plasmacytosis, high grade plasmacytosis, endometrioid, clear cell, mucinous) could not be evaluated individually. The fallopian tube is the main source of high-grade serous ovarian carcinoma (HGSOC), which is the most frequent type of ovarian cancer. Therefore clinical tissue selection cannot be limited to normal ovarian tissue alone. Moreover, the development of ovarian cancer involves complex molecular regulation pathways that need further investigation.

5. Conclusions

Our work suggests a tumorigenic role for lncRNA PART1 in EOC. It has revealed a potential mechanism by which lncRNA PART1 binds competitively to miR-150-5p, thereby upregulating the downstream oncogene MYB (Fig. 8). This research confirms a role for lncRNA PART1 in the development and pathogenesis of EOC, as well as identifying the likely molecular pathway.

Fig. 8.

Schematic model showing the putative role for lncRNA PART1 in the proliferation, migration and invasion of EOC.

Availability of Data and Materials

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

Author Contributions

JW, YH and TZ performed the in vivo studies. JW and YH performed the in vitro studies. JL and BX contributed to the design and analysis of the study and revised the manuscript. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work to take public responsibility for appropriate portions of the content and agreed to be accountable for all aspects of the work in ensuring that questions related to its accuracy or integrity.

Ethics Approval and Consent to Participate

All patients were provided with written informed consent. This study obtained ethical approval from the Second Affiliated Hospital of Nanjing Medical University. All tests were carried out in accordance with the Declaration of Helsinki. Ethics Committee of the Second Affiliated Hospital of Nanjing Medical University, number 2017AE02133.

All animal experiments complied with the Animal Research: Reporting of In vivo Experiments (ARRIVE) guidelines. AND all experiments involving use of mice were conducted in accordance with animal protocol (certificate number: 1811051) approved by the Animal Care and Use Committee of the Model Animal Research Center of Key Lab of Reproductive Medicine.

Acknowledgment

Not applicable.

Funding

This study was funded by National Natural Science Foundation of China (81873820) and 789 Excellent Talents Training Program of The Second Affiliated Hospital of Nanjing Medical University (789ZYRC202070208).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Coleman RL. Ovarian cancer in 2015: Insights into strategies for optimizing ovarian cancer care. Nature Reviews. Clinical Oncology. 2016; 13: 71–72.
[2]
Mao TL, Fan MH, Dlamini N, Liu CL. LncRNA MALAT1 Facilitates Ovarian Cancer Progression through Promoting Chemoresistance and Invasiveness in the Tumor Microenvironment. International Journal of Molecular Sciences. 2021; 22: 10201.
[3]
Li S, Jia H, Zhang Z, Wu D. DRAIC promotes growth of breast cancer by sponging miR-432-5p to upregulate SLBP. Cancer Gene Therapy. 2022; 29: 951–960.
[4]
Hua F, Li CH, Chen XG, Liu XP. Long Noncoding RNA CCAT2 Knockdown Suppresses Tumorous Progression by Sponging miR-424 in Epithelial Ovarian Cancer. Oncology Research. 2018; 26: 241–247.
[5]
Zhang S, Cheng J, Quan C, Wen H, Feng Z, Hu Q, et al. circCELSR1 (hsa_circ_0063809) Contributes to Paclitaxel Resistance of Ovarian Cancer Cells by Regulating FOXR2 Expression via miR-1252. Molecular Therapy. Nucleic Acids. 2020; 19: 718–730.
[6]
Ghafouri-Fard S, Shoorei H, Bahroudi Z, Abak A, Taheri M. The role of H19 lncRNA in conferring chemoresistance in cancer cells. Biomedicine & Pharmacotherapy. 2021; 138: 111447.
[7]
St Laurent G, Wahlestedt C, Kapranov P. The Landscape of long noncoding RNA classification. Trends in Genetics. 2015; 31: 239–251.
[8]
Hu X, Li Y, Kong D, Hu L, Liu D, Wu J. Long noncoding RNA CASC9 promotes LIN7A expression via miR-758-3p to facilitate the malignancy of ovarian cancer. Journal of Cellular Physiology. 2019; 234: 10800–10808.
[9]
Hao T, Huang S, Han F. LINC-PINT suppresses tumour cell proliferation, migration and invasion through targeting miR-374a-5p in ovarian cancer. Cell Biochemistry and Function. 2020; 38: 1089–1099.
[10]
Colvin EK, Howell VM, Mok SC, Samimi G, Vafaee F. Expression of long noncoding RNAs in cancer-associated fibroblasts linked to patient survival in ovarian cancer. Cancer Science. 2020; 111: 1805–1817.
[11]
Zhao L, Yang J, Liu X, Fu X, Ding Y, Wang C. Overexpression of Long Noncoding RNA E2F-Mediated Cell Proliferation Enhancing Long Noncoding RNA Is Involved in the Development of Chemoresistance of Cancer Cells to Carboplatin in Ovarian Endometrioid Adenocarcinoma. Cancer Biotherapy & Radiopharmaceuticals. 2019; 34: 566–571.
[12]
Chen Z, Chen X, Lu B, Gu Y, Chen Q, Lei T, et al. Up-regulated LINC01234 promotes non-small-cell lung cancer cell metastasis by activating VAV3 and repressing BTG2 expression. Journal of Hematology & Oncology. 2020; 13: 7.
[13]
Zuo X, Chen Z, Gao W, Zhang Y, Wang J, Wang J, et al. M6A-mediated upregulation of LINC00958 increases lipogenesis and acts as a nanotherapeutic target in hepatocellular carcinoma. Journal of Hematology & Oncology. 2020; 13: 5.
[14]
Xu DF, Wang LS, Zhou JH. Long non coding RNA CASC2 suppresses pancreatic cancer cell growth and progression by regulating the miR 24/MUC6 axis. International Journal of Oncology. 2020; 56: 494–507.
[15]
Shuai Y, Ma Z, Liu W, Yu T, Yan C, Jiang H, et al. TEAD4 modulated LncRNA MNX1-AS1 contributes to gastric cancer progression partly through suppressing BTG2 and activating BCL2. Molecular Cancer. 2020; 19: 6.
[16]
Lin H, Shen L, Lin Q, Dong C, Maswela B, Illahi GS, et al. SNHG5 enhances Paclitaxel sensitivity of ovarian cancer cells through sponging miR-23a. Biomedicine & Pharmacotherapy. 2020; 123: 109711.
[17]
Bartonicek N, Maag JLV, Dinger ME. Long noncoding RNAs in cancer: mechanisms of action and technological advancements. Molecular Cancer. 2016; 15: 43.
[18]
Yang M, Zhai Z, Guo S, Li X, Zhu Y, Wang Y. Long non-coding RNA FLJ33360 participates in ovarian cancer progression by sponging miR-30b-3p. OncoTargets and Therapy. 2019; 12: 4469–4480.
[19]
Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. A ceRNA hypothesis: the Rosetta Stone of a hidden RNA language? Cell. 2011; 146: 353–358.
[20]
Dong S, Wang R, Wang H, Ding Q, Zhou X, Wang J, et al. HOXD-AS1 promotes the epithelial to mesenchymal transition of ovarian cancer cells by regulating miR-186-5p and PIK3R3. Journal of Experimental & Clinical Cancer Research. 2019; 38: 110.
[21]
Wang A, Jin C, Li H, Qin Q, Li L. LncRNA ADAMTS9-AS2 regulates ovarian cancer progression by targeting miR-182-5p/FOXF2 signaling pathway. International Journal of Biological Macromolecules. 2018; 120: 1705–1713.
[22]
Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001; 25: 402–408.
[23]
Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Research. 2014; 42: D92–D97.
[24]
Jeggari A, Marks DS, Larsson E. miRcode: a map of putative microRNA target sites in the long non-coding transcriptome. Bioinformatics. 2012; 28: 2062–2063.
[25]
Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Research. 2017; 45: W98–W102.
[26]
Han Y, You J, Han Y, Liu Y, Huang M, Lu X, et al. LINC00184 Promotes Ovarian Cancer Cells Proliferation and Cisplatin Resistance by Elevating CNTN1 Expression via Sponging miR-1305. OncoTargets and Therapy. 2021; 14: 2711–2726.
[27]
Li Y, Jin K, van Pelt GW, van Dam H, Yu X, Mesker WE, et al. c-MYB Enhances Breast Cancer Invasion and Metastasis through the Wnt/β-Catenin/Axin2 Pathway. Cancer Research. 2016; 76: 3364–3375.
[28]
Yongchun Z, Linwei T, Xicai W, Lianhua Y, Guangqiang Z, Ming Y, et al. MicroRNA-195 inhibits non-small cell lung cancer cell proliferation, migration and invasion by targeting MYB. Cancer Letters. 2014; 347: 65–74.
[29]
Zhang J, Luo N, Luo Y, Peng Z, Zhang T, Li S. microRNA-150 inhibits human CD133-positive liver cancer stem cells through negative regulation of the transcription factor c-MYB. International Journal of Oncology. 2012; 40: 747–756.
[30]
Ramsay RG, Thompson MA, Hayman JA, Reid G, Gonda TJ, Whitehead RH. MYB expression is higher in malignant human colonic carcinoma and premalignant adenomatous polyps than in normal mucosa. Cell Growth & Differentiation. 1992; 3: 723–730.
[31]
Jia Y, Lin R, Jin H, Si L, Jian W, Yu Q, et al. MicroRNA-34 suppresses proliferation of human ovarian cancer cells by triggering autophagy and apoptosis and inhibits cell invasion by targeting Notch 1. Biochimie. 2019; 160: 193–199.
[32]
Chi Y, Wang D, Wang J, Yu W, Yang J. Long Non-Coding RNA in the Pathogenesis of Cancers. Cells. 2019; 8: 1015.
[33]
Visvanathan K, Shaw P, May BJ, Bahadirli-Talbott A, Kaushiva A, Risch H, et al. Fallopian Tube Lesions in Women at High Risk for Ovarian Cancer: A Multicenter Study. Cancer Prevention Research. 2018; 11: 697–706.
[34]
Yao RW, Wang Y, Chen LL. Cellular functions of long noncoding RNAs. Nature Cell Biology. 2019; 21: 542–551.
[35]
Torres D, Wang C, Kumar A, Bakkum-Gamez JN, Weaver AL, McGree ME, et al. Factors that influence survival in high-grade serous ovarian cancer: A complex relationship between molecular subtype, disease dissemination, and operability. Gynecologic Oncology. 2018; 150: 227–232.
[36]
Yoshimoto R, Chhipi-Shrestha JK, Schneider-Poetsch T, Furuno M, Burroughs AM, Noma S, et al. Spliceostatin A interaction with SF3B limits U1 snRNP availability and causes premature cleavage and polyadenylation. Cell Chemical Biology. 2021; 28: 1356–1365.e4.
[37]
Xie W, Zhu H, Zhao M, Wang L, Li S, Zhao C, et al. Crucial roles of different RNA-binding hnRNP proteins in Stem Cells. International Journal of Biological Sciences. 2021; 17: 807–817.
[38]
Karam A, Ledermann JA, Kim JW, Sehouli J, Lu K, Gourley C, et al. Fifth Ovarian Cancer Consensus Conference of the Gynecologic Cancer InterGroup: first-line interventions. Annals of Oncology. 2017; 28: 711–717.
[39]
Wu DD, Chen X, Sun KX, Wang LL, Chen S, Zhao Y. Role of the lncRNA ABHD11-AS1 in the tumorigenesis and progression of epithelial ovarian cancer through targeted regulation of RhoC. Molecular Cancer. 2017; 16: 138.
[40]
Hou G, Zhao X, Li L, Yang Q, Liu X, Huang C, et al. SUMOylation of YTHDF2 promotes mRNA degradation and cancer progression by increasing its binding affinity with m6A-modified mRNAs. Nucleic Acids Research. 2021; 49: 2859–2877.
[41]
Wang FW, Cao CH, Han K, Zhao YX, Cai MY, Xiang ZC, et al. APC-activated long noncoding RNA inhibits colorectal carcinoma pathogenesis through reduction of exosome production. The Journal of Clinical Investigation. 2019; 129: 727–743.
[42]
Sas-Chen A, Aure MR, Leibovich L, Carvalho S, Enuka Y, Körner C, et al. LIMT is a novel metastasis inhibiting lncRNA suppressed by EGF and downregulated in aggressive breast cancer. EMBO Molecular Medicine. 2016; 8: 1052–1064.
[43]
Zhao Y, Zhang Q, Liu H, Wang N, Zhang X, Yang S. lncRNA PART1, manipulated by transcriptional factor FOXP2, suppresses proliferation and invasion in ESCC by regulating the miR 18a 5p/SOX6 signaling axis. Oncology Reports. 2021; 45: 1118–1132.
[44]
Chen Y, Zhou X, Huang C, Li L, Qin Y, Tian Z, et al. LncRNA PART1 promotes cell proliferation and progression in non-small-cell lung cancer cells via sponging miR-17-5p. Journal of Cellular Biochemistry. 2021; 122: 315–325.
[45]
Tang C, Cai Y, Jiang H, Lv Z, Yang C, Xu H, et al. LncRNA MAGI2-AS3 inhibits bladder cancer progression by targeting the miR-31-5p/TNS1 axis. Aging. 2020; 12: 25547–25563.
[46]
Qi X, Lin Y, Liu X, Chen J, Shen B. Biomarker Discovery for the Carcinogenic Heterogeneity Between Colon and Rectal Cancers Based on lncRNA-Associated ceRNA Network Analysis. Frontiers in Oncology. 2020; 10: 535985.
[47]
Xu Y, Zhu H, Ma H, Yuan L, Hu Q, Yang L. LINC01305 inhibits malignant progression of cervical cancer via miR-129-5p/Sox4 axis. American Journal of Translational Research. 2020; 12: 7581–7592.
[48]
Hosono Y, Niknafs YS, Prensner JR, Iyer MK, Dhanasekaran SM, Mehra R, et al. Oncogenic Role of THOR, a Conserved Cancer/Testis Long Non-coding RNA. Cell. 2017; 171: 1559–1572.e20.
[49]
Fang Y, Fullwood MJ. Roles, Functions, and Mechanisms of Long Non-coding RNAs in Cancer. Genomics, Proteomics & Bioinformatics. 2016; 14: 42–54.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top