Skip to main content
  • Research article
  • Open access
  • Published:

LINC00852 promotes the proliferation and invasion of ovarian cancer cells by competitively binding with miR-140-3p to regulate AGTR1 expression

Abstract

Background

Dysregulation of long non-coding RNAs (lncRNAs) has been identified in ovarian cancer. However, the expression and biological functions of LINC00852 in ovarian cancer are not understood.

Methods

The expressions of LINC00852, miR-140-3p and AGTR1 mRNA in ovarian cancer tissues and cells were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR) assay. Gain- and loss-of-function assays were performed to explore the biological functions of LINC00852 and miR-140-3p in the progression of ovarian cancer in vitro. The bindings between LINC00852 and miR-140-3p were confirmed by luciferase reporter gene assay, RNA immunoprecipitation (RIP) assay and RNA pull-down assay.

Results

We found that LINC00852 expression was significantly up-regulated in ovarian cancer tissues and cells, whereas miR-140-3p expression was significantly down-regulated in ovarian cancer tissues. Functionally, LINC00852 knockdown inhibited the viability, proliferation and invasion of ovarian cancer cells, and promoted the apoptosis of ovarian cancer cells. Further investigation showed that LINC00852 interacted with miR-140-3p, and miR-140-3p overexpression suppressed the viability, proliferation and invasion of ovarian cancer cells. In addition, miR-140-3p interacted with AGTR1 and negatively regulated its level in ovarian cancer cells. Mechanistically, we found that LINC00852 acted as a ceRNA of miR-140-3p to promote AGTR1 expression and activate MEK/ERK/STAT3 pathway. Finally, LINC00852 knockdown inhibited the growth and invasion ovarian cancer in vivo.

Conclusion

LINC00852/miR-140-3p/AGTR1 is an important pathway to promote the proliferation and invasion of ovarian cancer.

Peer Review reports

Background

Ovarian cancer is the second most common cancer death in female genital system, and there will be approximately 21,750 estimated new cases and 13,940 estimated deaths in USA by the latest data from 2020 Cancer Statistics [1]. Ovarian cancer is usually diagnosed at advanced stages due to the lack of typical clinical symptom and effective clinical diagnosis method at early stages [2, 3]. It has been found that the development of ovarian cancer is closely related with the proliferation, invasion and migration of ovarian cancer cells [4]. The metastasis of ovarian cancer usually leads to the recurrence and poor prognosis of ovarian cancer [5]. Although standard surgery, radiotherapy and adjuvant chemotherapy have been applied to treat ovarian cancer patients [6, 7], the survival rate is still not satisfactory [8]. So, the metastasis and recurrence are needed to be controlled to improve the prognosis.

Previous studies have verified that many long non-coding RNAs (lncRNAs), such as lncRNA PTAR, lncRNA ABHD11-AS and lncRNA FLVCR1-AS1, promote the proliferation, invasion, migration, metastasis of ovarian cancer [9, 10]. LINC00852 is a newly found lncRNA which is firstly discovered in lung adenocarcinoma [11]. LINC00852 is overexpressed in lung adenocarcinoma spinal metastasis tissues and functions as an oncogene by promoting the proliferation, migration, and invasion of lung adenocarcinoma cells [11]. In addition, a recent study has shown that LINC00852 expression is up-regulated in osteosarcoma and promotes the proliferation, migration, and invasion of osteosarcoma cancer cells [12]. Moreover, LINC00852 can act as a ceRNA and competitively binding to miR-7-5p to exert its function in osteosarcoma [12]. However, whether LINC00852 modulates the proliferation, migration, and invasion of ovarian cancer is not discovered.

MicroRNA-140-3p (miR-140-3p) is a cancer-related miRNA that acts as a tumor suppressor and suppresses the proliferation and migration in a variety of cancers, such as colorectal cancer, breast cancer, and non-small cell lung cancer [13, 14]. A lot of studies have reported that miR-140-3p is decreased in cancer tissues and cancer cells, and miR-140-3p overexpression can inhibit the growth and tumorigenesis of cancers by directly inhibiting its target genes [15, 16]. Importantly, Gregory et al. have identified that miR-140-3p expression is down-regulated in ovarian cancer tissue by miRNA microarray analysis [17]. However, the role of miR-140-3p in the regulation of the proliferation and migration of ovarian cancer is not understood.

Angiotensin II Receptor Type 1 (AGTR1) is a receptor for angiotensin II and mediates the major cardiovascular effects of angiotensin II which acts as an effective vasopressor hormone and a major regulator of aldosterone secretion [18, 19]. According to the previous reports, AGTR1 exerts important functions in promoting the proliferation, invasion, migration and angiogenesis of cancer cells, such as glioma cells, breast cancer, cells and pancreatic cancer [20, 21]. On the contrary, AGTR1 antagonists can suppress the angiogenesis, migration and invasion of lung adenocarcinoma [22, 23]. Besides, AGTR1 has been found to promote the proliferation, migration and metastasis of ovarian cancer by triggering ERK1/2 and AKT signaling pathways, and AGTR1 overexpression predicts a poor prognosis of ovarian cancer [24].

In the present study, we investigated the role of LINC00852/miR-140-3p/AGTR1 pathway in ovarian cancer, and found LINC00852 acted as a ceRNA of miR-140-3p to repress miR-140-3p expression thereby promoting AGTR1 expression to promote the growth and invasion of ovarian cancer in vitro and in vivo.

Methods

Sample collection

Eighty-five ovarian cancer tissue and adjacent normal tissue samples were collected from ovarian cancer patients who underwent surgical resection at Cancer Hospital of China Medical University. Written informed consent was obtained from all patients. Tumor samples were confirmed by two pathologists independently and immediately frozen in liquid nitrogen. No patients received preoperative chemotherapy or radiotherapy. This study was approved by the Ethics Committee of Cancer Hospital of China Medical University.

Cell culture and transfection

Normal human ovarian epithelial cell line IOSE80 and human ovarian cancer cell lines (A2780, SKOV-3, OV-90, and CAOV3) were used in this study. The cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM; Gibco, CA, USA) or Roswell Park Memorial Institute (RPMI) 1640 medium (Gibco, CA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco, CA, USA), 2% sodium pyruvate (Gibco, CA, USA), 1% penicillin-streptomycin (Gibco, CA, USA) in an incubator containing 5% CO2 at 37 °C.

The vectors used for LINC00852 knockdown (sh, short hairpin)-LINC00852-1, sh-LINC00852-2, and sh-LINC00852-3), LINC00852 negative control (sh-NC), LINC00852 overexpression (oe-LINC00852 vecter was constructed by LINC00852 inserting into pcDNA3.1) and its negative control (oe-NC), miR-140-3p overexpression (miR-140-3p mimic) and its negative control (mimic NC), miR-140-3p knockdown (miR-140-3p inhibitor) and its negative control (inhibitor NC), and AGTR1 knockdown (sh-AGTR1) were synthesized by RiboBio (Guangdong, China). Transfection experiments were conducted using Lipofectamine 2000 (Invitrogen, CA, USA) according to the manufacturer’s instructions.

CCK-8 assay

SKOV-3 and OV-90 cells with different transfections were seeded into 96-well plates at a concentration of 1.5 × 104/mL. Forty-eight hours later, SKOV-3 and OV-90 cells were added with CCK-8 solution (10 μL; Sigma-Aldrich, MI, USA) and incubated for 2 h. The absorbance was measured at 450 nm by a microplate reader (Bio-Rad, CA, USA) at 24 h, 48 h and 72 h.

Colony formation assay

SKOV-3 and OV-90 cells with different transfections were seeded into 6-well plates at a concentration of 200 cells per well and cultured for 14 days. Then, SKOV-3 and OV-90 cell colonies were fixed with 80% methanol for 30 min and stained with 0.25% crystal violet at room temperature for 30 min.

Hoechst 33342 staining

The apoptosis of SKOV-3 and OV-90 cells with different transfections was confirmed by Hoechst 33342 staining according to previous report [25]. SKOV-3 and OV-90 cells were seeded in culture dish, added with 10 μL Hoechst 33342 solution (Beyotime Biotechnology, Nantong, China), and cultured for 10 min at 25 °C. A fluorescence microscopy (Olympus, Tokyo, Japan) was used to observe the changes in morphology of SKOV-3 and OV-90 cells (chromatin condensation, fragmentation and cell shrinkage). The apoptotic cancer cells were counted from 400 cells in 12 fields/well, and apoptosis rate (%) = apoptotic cancer cells/total cancer cells× 100.

Transwell assay

Mitomycin C (10 g/ml) was added to the cell culture medium to inhibit cell replication, which ruled out the changes in intercellular space caused by cell proliferation or apoptosis [26]. SKOV-3 and OV-90 cells (2 × 105 cells/mL) were added to the upper chambers of Transwell (8-μm-diameter pore membrane; Corning, NY, USA) coated with Matrigel. SKOV-3 and OV-90 cells were allowed to invade for 24 h, and cotton swabs were used to scrub cancer cells that did not penetrated the filters. Then, chambers were fixed with 4% paraformaldehyde (Beyotime Biotechnology, Nantong, China) for 2 min, stained with 0.3% crystal violet (Beyotime Biotechnology, Nantong, China) for 2 min and observed under a light microscope (Olympus, Tokyo, Japan).

qRT-PCR

Total RNAs were extracted from tumor tissues and ovarian cancer cells (SKOV-3 and OV-90 cells) using Trizol Reagent (Invitrogen, CA, USA). Complementary DNA (cDNA) was synthesized using High-Capacity RNA-to-cDNA Kit (Applied Biosystems, CA, USA) according to the manufacturer’s instruction. qRT-PCR reactions were conducted on an ABI StepOne Real-time PCR System (Applied Biosystems, CA, USA) using PowerUp SYBR Green Master Mix with the primers as the following: LINC00852 forward, 5′-CGTTGCCTACAGTCAAGTCAGT-3′, reverse, 5′-GCCATGGTTCCCTTACTGATAC-3′; miR-140-3p forward, 5′-ACACTCCAGCTGGGAGGCGGGGCGCCGCGGGA-3′, reverse, 5′-CTCAACTGGTGTCGTGGA-3′; AGTR1 forward, 5′-CCTCAGATAATGTAAGCTCATCCAC-3′, reverse, 5′-GCTGCAGAGGAATGTTCTCTT-3′; U6 forward, 5′-CTCGCTTCGGCAGCACA-3′, reverse, 5′-AACGCTTCACGAATTTGCGT-3′; GAPDH forward, 5′-TGTTCGTCATGGGTGTGAAC-3′, reverse, 5′-ATGGCATGGACTGTGGTCAT-3′. Relative gene expressions of LINC00852, miR-140-3p and AGTR1 were calculated using the2-ΔΔCt method and normalized by GAPDH or U6.

Western blotting

Proteins were extracted from tumor tissues and ovarian cancer cells (SKOV-3 and OV-90 cells) using RIPA lysis and extraction buffer (ThermoFisher Scientific, CA, USA). The concentration of proteins was measured using BCA protein assay kit (ThermoFisher Scientific, CA, USA). Proteins were separated by 10% sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred onto polyvinylidene fluoride membranes (PVDF; Invitrogen, CA, USA). The blots were blocked with 5% skimmed milk, and incubated with primary antibodies overnight at 4 °C: MMP-2 (1:1000; Abcam, Cambridge, UK), MMP-9 (1:5000; Abcam, Cambridge, UK), Ki67 (1:1000; Abcam, Cambridge, UK), PCNA (1:5000; Abcam, Cambridge, UK), MEK (1:1000; Cell Signaling Technology, MA, USA), p-MEK (1:1000; Cell Signaling Technology, MA, USA), ERK1/2 (1:1000; Abcam, Cambridge, U), p-ERK1/2 (1:1000; Cell Signaling Technology, MA, USA), STAT3 (1:1000; Cell Signaling Technology, MA, USA), p-STAT3 (1:2000; Cell Signaling Technology, MA, USA), GAPDH (1:1000; Cell Signaling Technology, MA, USA). The blots were then incubated with goat anti-rabbit or anti-mouse secondary antibody (Cell Signaling Technologies, MA, USA), visualized by an enhanced chemiluminescence kit (ThermoFisher Scientific, CA, USA) and normalized to GAPDH expression. The membranes were firstly cut, and then stained with antibodies. Each complete membrane has GAPDH loading control. The bands were photographed with a ChemiDoc™XRS Imaging System (Bio-Rad, CA, USA) and quantified by Quantity One software (Bio-Rad, CA, USA).

Detection of gelatin zymography by mono-dimensional gel electrophoresis

The enzymatic activity of MMP-2 and MMP-9 in SKOV3 and OV-90 cells was conducted with gelatine zymography. Proteins (20 μg) extracted from ovarian cancer cells (SKOV-3 and OV-90 cells) were used for each zymographic assay. Under non-reducing condition, mono-dimensional gelatine zymography was conducted on SDS-PAGE (7.5%) copolymerized with gelatine (0.1%). After electrophoresis, SDS was removed from the gels and zymograms were developed for 18 h at 37 °C. Gels were then stained with Coomassie Brilliant Blue for 30 min at 25 °C and visualized after destaining in methanol/acetic acid/H2O.

Flourescence in situ hybridization (FISH) assay

FISH was conducted according to previous report [27]. Digoxin (Sigma-Aldrich, MI, USA)-labeled LINC00852 complementary DNA probe was synthesized in vitro. SKOV-3 cells were grown on the slides. After washed with phosphate buffer solution (PBS) for three times, SKOV-3 cells grown on the slides were fixed with 4% paraformaldehyde (Beyotime Biotechnology, Nantong, China). Then, the slides were treated with protease reagent (Invitrogen, CA, USA) and hybridized with digoxin-labeled LINC00852 probe for 12 h at 40 °C. Images were observed by a confocal microscope (Olympus, Tokyo, Japan) and the magnification of images was 400 × objective.

Dual luciferase reporter gene assay

The luciferase reporter gene assay was conducted according to previous report [28]. The sequences of miR-140-3p or AGTR1 3′-UTR was sub-cloned into pGL3 luciferase reporter vectors (Promega, WI, USA). SKOV-3 cells were seeded into 48-well plates at a concentration of 3 × 104 cells/well, and transfected with wide type miR-140-3p vector (pGL3-miR-140-3p-wt) or mutant miR-140-3p vector (pGL3-miR-140-3p-mut), wide type 3′-UTR of AGTR1 vector (pGL3-AGTR1-wt), mutant 3′-UTR of AGTR1 vector (pGL3-AGTR1-mut) using Lipofectamine 2000 reagent (Invitrogen, CA, USA). Forty-eight hours later, luciferase activity was detected by dual luciferase reporter assay system (Progema, WI, USA).

RNA immunoprecipitation (RIP) assay

RIP assay was conducted as previously described [29]. SKOV-3 cells (2 × 107) were collected to perform RIP assay using an AGO2 antibody (Millipore, MA, USA). AGO2 antibody (5 μg) for each RIP was used in RIP assay, and normal rabbit IgG was used as negative control. The co-precipitated RNAs were isolated and detected by qRT-PCR.

RNA pull-down assay

SKOV-3 cells were transfected with biotin-labeled wild-type miR-140-3p probe (bio-miR-140-3p-wt), biotin-labeled mutant miR-140-3p probe (bio-miR-140-3p-mut) or negative control probe (bio-probe NC). After 48 h, SKOV-3 cells were collected and lysed using RIPA lysis and extraction buffer (ThermoFisher Scientific, CA, USA). Then, cell lysates were incubated with Streptavidin magnetic beads (Pierce, CA, USA) for 12 h at 4 °C. After elution of Streptavidin magnetic beads, the bound RNAs were detected using qRT-PCR.

Xenograft mouse model

Ovarian cancer xenograft mouse model was established by subcutaneously injecting BALB/c nude mice (4–5 weeks, weight 20 ± 0.5 g, female) with 1 × 107 SKOV-3 cells transfected with sh-LINC00852 or sh-NC (sh-LINC00852 and sh-NC groups), with five mice in each group. For control group, BALB/c nude mice were injected with SKOV-3 cells without transfection. BALB/c nude mice were obtained from Charles River (Beijing, China), and kept in individual cages with standard chow and ad libitum access to drinking water under a sterile condition. Tumor volumes were assessed by measuring the length and width of the tumor with calipers (tumor volume [mm3] = 0.5 × length×width2). The length and width were detected between 09.00 am to 10.00 am and testing order was randomized daily by two investigators who are unaware of grouping. Six weeks later, all mice were euthanized by an overdose of 100 mg/kg sodium pentobarbital through intravenous injection. And the tumors were excised, photographed and weighed. The animal experiment was approved by the Animal Care and Use Committee of Cancer Hospital of China Medical University.

Immunohistochemistry (IHC) analysis

Tumor tissue sections were de-paraffinized and rehydrated using descending concentrations of ethanol. Then, tumor tissue sections were added with 3% H2O2 for 30 min and blocked with 1% FBS for 1 h. After that, tumor tissue sections were incubated with anti-MMP-2 (1:100; Abcam, Cambridge, UK), anti-MMP-9 (1:1000; Abcam, Cambridge, UK), anti-Ki67 (1:1000; Abcam, Cambridge, UK) and anti-PCNA (1:200; Abcam, Cambridge, UK) overnight at 4 °C. Then, the sections were washed with PBS and incubated with secondary antibodies (1:500; Abcam, Cambridge, UK) for 1 h at room temperature. Finally, tumor tissue sections were stained with 3,3′-diaminobenzidine (DAB) staining solution and observed by a fluorescence microscope (Nikon, Tokyo, Japan).

Lung metastasis model

SKOV-3 cells (1 × 106/0.2 ml) transfected with sh-LINC00852-1 or sh-NC were injected into the tail veins of 15 BALB/c nude mice (4–5 weeks, weight 20 ± 0.5 g, female). BALB/c nude mice were obtained from Charles River (Beijing, China), and kept in individual cages with standard chow and ad libitum access to drinking water under a sterile condition. For control group, BALB/c nude mice were injected with SKOV-3 cells without transfection. There were five mice in each group. The mice were sacrificed 4 weeks after the inoculation and lungs were removed. Then, metastatic nodules were counted macroscopically.

Statistical analysis

GraphPad Prism 5.0 was used for data analysis. Data were expressed as mean ± standard deviation (SD). Comparisons for two groups were analyzed by Student’s t-test. Comparisons for multiple groups were analyzed by one-way analysis of variance ANOVA followed by Bonferroni post hoc test. P value less than 0.05 was considered statistically significant.

Results

LINC00852 is highly expressed in ovarian cancer

As shown in Fig. 1a, LINC00852 expression was significantly up-regulated in ovarian cancer tissues than adjacent normal tissues. Besides, LINC00852 expression was significantly up-regulated in human ovarian cancer cells (A2780, SKOV-3, OV-90 and CAOV3) than normal human ovarian epithelial cells IOSE80, with highest LINC00852 expression in SKOV-3 cells (Fig. 1b). Importantly, the analysis of LINC00852 expression and clinicopathological characteristics of ovarian cancer showed that LINC00852 expression was correlated with TNM stage and lymph node metastasis (Table 1).

Fig. 1
figure 1

LINC00852 is highly expressed in ovarian cancer. a LINC00852 expression in ovarian cancer tissues and adjacent normal tissues was detected by qRT-PCR. N = 85. b LINC00852 expression was detected in normal human ovarian epithelial cells IOSE80 and human ovarian cancer cells (A2780, SKOV-3, OV-90, and CAOV3) by qRT-PCR. *p < 0.05

Table 1 Correlation between LINC00852 expression and clinicopathological characteristics in ovarian cancer patients (N = 85)

LINC00852 knockdown inhibits the proliferation and invasion of ovarian cancer cells

Since SK-OV3 and OV-90 cells are commonly used in the study of the growth and invasion in ovarian cancer [30,31,32,33] and LINC00852 expression is highly expressed in SKOV-3 and OV-90 cells, we use SKOV-3 and OV-90 cells for the following experiments. After SKOV-3 and OV-90 cells transfected with sh-LINC00852-1, sh-LINC00852-2 and sh-LINC00852-3, LINC00852 expression was significantly down-regulated in SKOV-3 and OV-90 cells (Fig. 2a), and results showed that sh-LINC00852-1 obtained the best transfection efficiency (Fig. 2a). The viability of SKOV-3 and OV-90 cells was significantly inhibited in sh-LINC00852-1 and sh-LINC00852-2 groups than control and sh-NC groups (Fig. 2b). The colony formation ability of SKOV-3 and OV-90 cells was suppressed in sh-LINC00852-1 and sh-LINC00852-2 groups than control and sh-NC groups (Fig. 2c). The apoptosis rate of SKOV-3 and OV-90 cells was significantly increased in sh-LINC00852-1 and sh-LINC00852-2 groups than control and sh-NC groups (Fig. 2d). The invasion rate of SKOV-3 and OV-90 cells was significantly decreased in sh-LINC00852-1 and sh-LINC00852-2 groups than control and sh-NC groups (Fig. 2e). Moreover, protein levels of invasion markers MMP-2, MMP-9, proliferation markers Ki67 and PCNA in SKOV-3 and OV-90 cells were significantly down-regulated in sh-LINC00852-1 and sh-LINC00852-2 groups than control and sh-NC groups (Fig. 2f and g), whereas apoptosis markers cleaved caspase-3 and cleaved PARP in SKOV-3 and OV-90 cells were significantly up-regulated in sh-LINC00852-1 and sh-LINC00852-2 groups than control and sh-NC groups (Fig. 2f and g). These findings indicated that sh-LINC00852 inhibited the proliferation and invasion of ovarian cancer cells. According to gelatine zymography analyses, enzymatic forms of MMP-2 and MMP-9 were reduced in sh-LINC00852 group than control and sh-NC groups (Fig. 2h).

Fig. 2
figure 2

LINC00852 knockdown inhibits the proliferation and invasion of ovarian cancer cells. a SKOV-3 and OV-90 cells were transfected with sh-LINC00852-1, sh-LINC00852-2, and sh-LINC00852-3, then LINC00852 expression was detected in SKOV-3 and OV-90 cells with different transfections. b The viability of SKOV-3 and OV-90 cells at different groups was detected by CCK-8 assay. c The colony formation ability of SKOV-3 and OV-90 cells at different groups was detected by colony formation assay. d The apoptosis rate of SKOV-3 and OV-90 cells at different groups was observed by Hoechst 33342 staining. e The invasion rate of SKOV-3 and OV-90 cells at different groups was detected by Transwell assay. f-g Protein levels of MMP-2, MMP-9, Ki67, PCNA, apoptosis markers cleaved caspase-3 and cleaved PARP in SKOV-3 and OV-90 cells at different groups were measured by Western blotting. H. Gelatine zymography analyses showed enzymatic forms of MMP-2 and MMP-9. *p < 0.05, compared with control

LINC00852 acts as a ceRNA of miR-140-3p

According to the online bioinformatics software (http://lncatlas.crg.eu), we found that LINC00852 localized mainly in the cytoplasm (Fig. 3a). FISH assay further confirmed the subcellular localization of LINC00852 in the cytoplasm (Fig. 3b). In addition, bioinformatics software (Starbase) predicted the binding sites between LINC00852 and miR-140-3p (Fig. 3c). Dual luciferase reporter gene assay showed that LINC00852 significantly decreased the luciferase activity of miR-140-3p-wt, whereas LINC00852 did not change the luciferase activity of miR-140-3p-Mut (Fig. 3d). In addition, RIP assay showed LINC00852 was enriched in Ago2 group than IgG group, indicating LINC00852 can bind with Ago2 (Fig. 3e). Compared with bio-miR-140-3p-mut or bio-probe NC, LINC00852 can be enriched by bio-miR-140-3p-wt (Fig. 3f).

Fig. 3
figure 3

LINC00852 acts as a ceRNA of miR-140-3p. a Online bioinformatics software (http://lncatlas.crg.eu) predicted the subcellular localization of LINC00852. b The subcellular location of LINC00852 was confirmed by FISH assay. Scale bars, 20 μm. c The binding sites between LINC00852 and miR-140-3p were predicted by bioinformatics software (Starbase). d The regulation of LINC00852 on miR-140-3p was detected by dual luciferase reporter gene assay. *p < 0.05, compared with oe-NC. e The binding between LINC00852 and Ago2 was detected by RIP assay. *p < 0.05, compared with IgG. F. RNA pull-down assay was used to detect the enrichment of LINC00852 by miR-140-3p. *p < 0.05, compared with bio-probe NC

miR-140-3p overexpression reverses the promotion effect of LINC00852 on the proliferation and invasion of ovarian cancer cells

As shown in Fig. 4a, miR-140-3p expression was significantly down-regulated in ovarian cancer tissues than adjacent normal tissues. Compared with oe-NC + mimic NC group, miR-140-3p expression in SKOV-3 cells was significantly up-regulated in oe-NC + miR-140-3p mimic group, and miR-140-3p expression was further down-regulated in oe-LINC00852 + miR-140-3p mimic group compared with oe-NC + miR-140-3p mimic group (Fig. 4b). Besides, the viability of SKOV-3 cells was significantly inhibited in oe-NC + miR-140-3p mimic group compared with oe-NC + mimic NC group, and the viability of SKOV-3 cells was further promoted in oe-LINC00852 + miR-140-3p mimic group compared with oe-NC + miR-140-3p mimic group (Fig. 4c). The colony formation ability of SKOV-3 cells was significantly inhibited in oe-NC + miR-140-3p mimic group than oe-NC + mimic NC group, and the colony formation ability of SKOV-3 cells was further promoted in oe-LINC00852 + miR-140-3p mimic group compared with oe-NC + miR-140-3p mimic group (Fig. 4d). The apoptosis of SKOV-3 cells was significantly facilitated in oe-NC + miR-140-3p mimic group compared with oe-NC + mimic NC group, and the apoptosis of SKOV-3 cells was further suppressed in oe-LINC00852 + miR-140-3p mimic group compared with oe-NC + miR-140-3p mimic group (Fig. 4e). The invasion of SKOV-3 cells was significantly inhibited in oe-NC + miR-140-3p mimic group compared with oe-NC + mimic NC group, and the invasion of SKOV-3 cells was further promoted in oe-LINC00852 + miR-140-3p mimic group compared with oe-NC + miR-140-3p mimic group (Fig. 4f). In addition, miR-140-3p mimic significantly down-regulated protein levels of invasion markers MMP-2, MMP-9, proliferation markers Ki67 and PCNA, and promoted protein levels of apoptosis markers cleaved caspase-3 and cleaved PARP in SKOV-3 cells, and oe-LINC00852 + miR-140-3p mimic further reversed protein levels of these invasion markers (Fig. 4g and h). According to gelatine zymography analyses, enzymatic forms of MMP-2 and MMP-9 were reduced in oe-NC + miR-140-3p mimic group than oe-NC + mimic NC group, and enzymatic forms of MMP-2 and MMP-9 were increased in oe-LINC00852 + miR-140-3p mimic group than oe-NC + miR-140-3p mimic group (Fig. 4i).

Fig. 4
figure 4

miR-140-3p overexpression reverses the promotion effect of LINC00852 on the proliferation and invasion of ovarian cancer cells. a miR-140-3p expression in ovarian cancer tissues and adjacent normal tissues was detected by qRT-PCR. N = 85. SKOV-3 cells were transfected with oe-NC + mimic NC, oe-NC + miR-140-3p mimic, and oe-LINC00852 + miR-140-3p mimic. b miR-140-3p expression in SKOV-3 cells was detected by qRT-PCR. c The viability of SKOV-3 cells was detected by CCK-8 assay. d The colony formation ability of SKOV-3 cells was detected by colony formation assay. e The apoptosis rate of SKOV-3 cells was observed by Hoechst 33342 staining. f The invasion rate of SKOV-3 cells was detected by Transwell assay. g-h Protein levels of MMP-2, MMP-9, Ki67, PCNA, cleaved caspase-3 and cleaved PARP in SKOV-3 cells were detected by Western blotting. I. Gelatine zymography analyses showed enzymatic forms of MMP-2 and MMP-9. *p < 0.05, oe-NC + mimic NC compared with oe-NC + miR-140-3p mimic, #p < 0.05, oe-NC + miR-140-3p mimic compared with oe-LINC00852 + miR-140-3p mimic

LINC00852 acts as a ceRNA to regulate AGTR1 expression by sponging miR-140-3p

According to the prediction of bioinformatics software (Starbase), AGTR1 was a target of miR-140-3p (Fig. 5a). Dual luciferase reporter gene assay showed that miR-140-3p mimic significantly decreased the luciferase activity of AGTR1-Wt, whereas miR-140-3p mimic did not change the luciferase activity of AGTR1-mut (Fig. 5b). Different transfection experiments showed that LINC00852 could negatively regulate miR-140-3p expression and positively regulate AGTR1 expression (Fig. 5c). Importantly, loss- and gain-of-function experiments showed that protein levels of phosphorylation MEK (p-MEK), p-ERK1/2 and p-STAT3 in SKOV-3 cells were significantly down-regulated in oe-LINC00852 + miR-140-3p mimic group than oe-LINC00852 + mimic-NC group (Fig. 5d). Protein levels of p-MEK, p-ERK1/2 and p-STAT3 in SKOV-3 cells were significantly up-regulated in sh-LINC00852 + miR-140-3p inhibitor group than sh-LINC00852 + inhibitor NC group (Fig. 5d). Protein levels of p-MEK, p-ERK1/2 and p-STAT3 in SKOV-3 cells were significantly down-regulated in miR-140-3p inhibitor+sh-AGTR1 group than miR-140-3p inhibitor+sh-NC group (Fig. 5d). Moreover, we found AGTR1 expression level was significantly up-regulated in ovarian cancer cells (SKOV-3, OV-90 and CAOV3) than normal human ovarian epithelial cells IOSE80 (Fig. 5e). In in vivo model, tumor volume was significantly decreased in sh-AGTR1 group than sh-NC group (Fig. 5f and g). Besides, tumor weight was significantly reduced in sh-AGTR1 group than sh-NC group (Fig. 5h). These findings indicated that LINC00852 sponged miR-140-3p to promote AGTR1 expression, thereby facilitating the activation of MEK/ERK/STAT3 pathway.

Fig. 5
figure 5

LINC00852 acts as a ceRNA to regulate AGTR1 expression by sponging miR-140-3p. a Bioinformatics software (Starbase) predicted that AGTR1 was a target of miR-140-3p. b The regulation of miR-140-3p on AGTR1 was confirmed by dual luciferase reporter gene assay. *p < 0.05, compared with mimic NC. c LINC00852, miR-140-3p and AGTR1 expressions were detected in different groups of SKOV-3 cells by qRT-PCR. *p < 0.05, compared with oe-LINC00852 + mimic NC. d Protein levels of MEK, p-MEK, ERK1/2, p-ERK1/2, STAT3 and p-STAT3 were detected in different groups of SKOV-3 cells by Western blotting. *p < 0.05, compared with oe-LINC00852 + mimic NC. e AGTR1 expression level was detected in normal human ovarian epithelial cells IOSE80 and ovarian cancer cells (A2780, SKOV-3, OV-90 and CAOV3). *p < 0.05, compared with IOSE80. f-g In in vivo model, tumor volume was detected in sh-AGTR1 group and sh-NC group. h Tumor weight was measured in sh-AGTR1 group and sh-NC group. *p < 0.05, compared with sh-NC

LINC00852 knockdown inhibits the growth and invasion of ovarian cancer and AGTR1/MEK/ERK/STAT3 pathway in vivo

As shown in Fig. 6a and b, tumor volume was significantly decreased in sh-LINC00852-1 group than sh-NC group, whereas there was no significant difference between control and sh-NC groups. Besides, tumor weight was significantly inhibited in sh-LINC00852-1 group than sh-NC group, whereas there was no significant difference between control and sh-NC groups (Fig. 6c). In addition, there was no significant difference in the body weight of mice among control, sh-NC and sh-LINC00852-1 groups (Fig. 6d). LINC00852 expression in tumor tissues was significantly down-regulated in sh-LINC00852-1 group than sh-NC group, whereas there was no significant difference between control and sh-NC groups, and miR-140-3p expression in tumor tissues was significantly up-regulated in sh-LINC00852-1 group than sh-NC group, whereas there was no significant difference between control and sh-NC groups (Fig. 6e). Moreover, protein levels of AGTR1, p-MEK, p-ERK1/2 and p-STAT3 in tumor tissues were significantly down-regulated in sh-LINC00852-1 group than sh-NC group (Fig. 6f). IHC staining showed the expressions of invasion marker MMP-2, MMP-9, proliferation markers Ki67 and PCNA were inhibited in sh-LINC00852-1 group than sh-NC group (Fig. 6g). Lung metastasis assay showed that sh-LINC00852-1 treatment attenuated lung metastasis of SKOV-3 cells compared with sh-NC group, whereas there was no significant difference between control and sh-NC groups (Fig. 6h and i).

Fig. 6
figure 6

LINC00852 knockdown inhibits ovarian cancer and AGTR1/MEK/ERK/STAT3 pathway in vivo. SKOV-3 cells transfected with sh-LINC00852-1 or sh-NC were subcutaneously injected into nude mice. Tumor tissues were collected at day 42. a-b Tumor volume was suppressed in sh-LINC00852-1 group than sh-NC group, whereas there was no significant differences between control and sh-NC groups. c Tumor weight was inhibited in sh-LINC00852-1 group than sh-NC group. d Body weight of mice in control, sh-NC, sh-LINC00852-1 groups at 1, 2, 3, 4, 5 and 6 weeks. e LINC00852 and miR-140-3p expression in tumor tissues of sh-LINC00852-1 group, control and sh-NC group were detected by qRT-PCR. f Protein levels of AGTR1, MEK, p-MEK, ERK1/2, p-ERK1/2, STAT3 and p-STAT3 in tumor tissues of sh-LINC00852-1 group and sh-NC group were detected by Western blotting. g The expressions of invasion marker MMP-2, MMP-9, Ki67 and PCNA were detected by IHC staining. h-i The average number of lung metastasis tumors per mouse in control, sh-NC and sh-LINC00852 groups . *p < 0.05, compared with control group

Discussion

LINC00852, also known as nasopharyngeal carcinoma related gene NAG73, C3orf42, ghrelin opposite strand/antisense RNA (GHRLOS), and GHRL-AS2, can be highly expressed in cancer tissues and cancer cells, such as gastric cancer and thyroid cancer [34, 35]. The highly expressed lncRNAs usually act as oncogenes to promote the progression of cancer, and lowly expressed lncRNAs usually act as tumor suppressers to suppress the progression of cancer [36, 37]. A report by Seim et al. has illustrated that GHRLOS (LINC00852) expression was highly expressed in human cerebellum, foetal brain, whole brain, thymus, thyroid, testis, uterus and ovary [35]. However, whether LINC00852 acts as oncogene or tumor suppressing gene in ovarian cancer is not clear. So, it is meaningful for identifying the role of LINC00852 in the regulation of ovarian cancer progression. In this study, we found that LINC00852 was highly expressed in ovarian cancer tissues and ovarian cancer cells. Therefore, we first identified that LINC00852 was up-regulated in ovarian cancer, which might play vital roles in the progression of ovarian cancer.

The dysregulation of lncRNAs is involved in modulating the proliferation, migration, invasion and metastasis of cancers [38]. As a new modulatory RNA molecule, GHRLOS (LINC00852) was up-regulated in gastric cancer tissues, and high expression of GHRLOS predicted poor overall survival in patients of gastric cancer [34]. LINC00852 was up-regulated in lung adenocarcinoma spinal metastases and lung adenocarcinoma cells, and LINC00852 overexpression promoted the proliferation and inhibited the apoptosis of lung adenocarcinoma cells in vitro [11]. GHRLOS (LINC00852) was lowly expressed in colorectal cancer tissues, and decreased expression of GHRLOS was correlated with distant metastasis, lymph node metastasis and poor histological tumor grade [39]. In addition, high expression of GHRLOS promoted the proliferation and migration of breast cancer cells in vitro, and GHRLOS overexpression facilitated the orthotopic xenograft growth in vivo [40]. These studies indicate LINC00852 might exert important functions in regulating the progression of a variety of cancers. In this study, loss-of-function assays illustrated that LINC00852 knockdown decreased cell viability, inhibited the colony formation and invasion of ovarian cancer cells, and promoted the apoptosis of ovarian cancer cells in vitro. These results first illustrated that LINC00852 functioned as an oncogene in ovarian cancer for promoting the proliferation and invasion of ovarian cancer cells.

Studies have identified that the expression of miR-140 can be reduced by lncRNAs in several cancer cells [41, 42]. By competitively binding with miR-140, a variety of lncRNAs, such as lncRNA XIST, lncRNA MALAT1 and lncRNA H19, can promote cancer progression via facilitating the proliferation, migration and invasion of cancer cells in vitro and metastasis in vivo [42]. For example, lncRNA PGM5-AS1 directly bound with miR-140 in osteosarcoma cells to modulate downstream FBN1 pathway, thereby affecting invasion, migration and tumorigenesis of osteosarcoma [43]. LncRNA SNHG20 directly bound with miR-140 in laryngeal squamous cell carcinoma cells, and SNHG20 knockdown decreased the proliferation and suppressed the malignant progression of laryngeal squamous cell carcinoma [44]. In this study, we confirmed the subcellular localization of LINC00852 in the cytoplasm. Dual luciferase reporter gene assay together with RIP assay and RNA pull down assay showed that miR-140-3p was a downstream molecule of LINC00852, and LINC00852 acted as a sponge for miR-140-3p in ovarian cancer cells. We further found that miR-140-3p was down-regulated in ovarian cancer tissues. LINC00852 significantly reduced the cell viability, inhibited the colony formation and invasion of ovarian cancer cells, and promoted the apoptosis of ovarian cancer cells in vitro by sponging miR-140-3p. To the best of our knowledge, this study firstly confirmed that the expression and biological functions of LINC00852 in ovarian cancer, and identified that LINC00852 knockdown increased miR-140-3p expression to suppress the proliferation and invasion of ovarian cancer cells.

A lot of target genes of miR-140-3p, including RRM2, PD-L1, MAPK and BRD9, can be remarkably suppressed by miR-140-3p in cancer cells through binding to the 3′-untranslated regions (3′ UTRs) [13, 45]. It has been reported that AGTR1 expression is related with the growth, metastasis and poor prognosis in breast cancer, colorectal cancer and gastric cancer [46]. Moreover, AGTR1 has been verified to promote the colony formation, migration and metastasis of ovarian cancer cells [24]. MEK/ERK/STAT3 pathway has been found to be involved in regulating the proliferation, invasion and migration of ovarian cancer cells and hepatocellular carcinoma cells [47]. In addition, AGTR1 has been identified to be the upstream molecule of MEK/ERK/STAT3 pathway in regulating the growth and metastasis of prostate cancer [48]. In this study, we found that AGTR1 was the target gene modulated by miR-140-3p. The introduction of AGTR1 knockdown reversed the promotion effect of miR-140-3p inhibitor on the activation of MEK/ERK/STAT3 pathway in ovarian cancer cells, indicating the loss of AGTR1 expression can result in the inhibition of ovarian cancer progression.

The metastasis of ovarian cancer is a main factor that contributes to the recurrence and poor prognosis of ovarian cancer [49]. More and more in vivo experiments have verified that lncRNAs could affect the metastasis of tumors to regulate the progression of ovarian cancer [50, 51]. In this study, we found that the loss of LINC00852 significantly reduced tumor volume and tumor weight in a SKOV-3 xenograft mouse model. Besides, the loss of LINC00852 significantly increased miR-140-3p expression, reduced AGTR1 expression and inhibited the activation of MEK/ERK/STAT3 pathway in tumor tissues from SKOV-3 xenograft mice. Moreover, IHC staining showed the invasion marker MMP-2, MMP-9, Ki67 and PCNA were down-regulated by the loss of LINC00852, suggesting LINC00852 could promote the invasion of ovarian cancer in vivo. We will focus on the effects of high/low LINC00852 and miR-140-3p expressions on the prognosis of ovarian cancer patients in further researches.

Conclusion

In summary, this study found that LINC00852 was up-regulated in ovarian cancer and acted as a tumor promoter to facilitate ovarian cancer cell proliferation and invasion. Besides, LINC00852 was found to promote AGTR1 expression by sponging miR-140-3p. These results indicate that LINC00852/miR-140-3p/AGTR1 pathway exerts important function in ovarian cancer and possess a potential function in the treatment of ovarian cancer. However, we only initially verified the regulatory mechanism of LINC00852/miR-140-3p/AGTR1 pathway in ovarian cancer. Due to the usage of cervical cancer cell lines with high expression level of LINC00852 in this study, more researches are needed to verify whether low LINC00852 expressing cell lines or tumor can follow the conclusions of these results.

Availability of data and materials

The data used to support the findings of this study are available from the corresponding author upon request.

Abbreviations

qRT-PCR:

Quantitative real-time PCR

lncRNAs:

Long non-coding RNAs

sh:

Short hairpin

NC:

Negative control

RIP:

RNA immunoprecipitation

miR-140-3p:

MicroRNA-140-3p

AGTR1:

Angiotensin II Receptor Type 1

IHC:

Immunohistochemistry

3′ UTRs:

3′-untranslated regions

References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70(1):7–30. https://doi.org/10.3322/caac.21590.

    Article  PubMed  Google Scholar 

  2. Rottmann M, Burges A, Mahner S, Anthuber C, Beck T, Grab D, et al. Cancer of the ovary, fallopian tube, and peritoneum: a population-based comparison of the prognostic factors and outcomes. J Cancer Res Clin Oncol. 2017;143(9):1833–44. https://doi.org/10.1007/s00432-017-2422-6.

    Article  PubMed  Google Scholar 

  3. Doubeni CA, Doubeni AR, Myers AE. Diagnosis and Management of Ovarian Cancer. Am Fam Physician. 2016;93(11):937–44.

    PubMed  Google Scholar 

  4. Li S, Lv M, Qiu S, Meng J, Liu W, Zuo J, et al. NF-κB p65 promotes ovarian cancer cell proliferation and migration via regulating mortalin. J Cell Mol Med. 2019;23(6):4338–48. https://doi.org/10.1111/jcmm.14325.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hoskins PJ, Gotlieb WH. Missed therapeutic and prevention opportunities in women with BRCA-mutated epithelial ovarian cancer and their families due to low referral rates for genetic counseling and BRCA testing: a review of the literature. CA Cancer J Clin. 2017;67(6):493–506. https://doi.org/10.3322/caac.21408.

    Article  PubMed  Google Scholar 

  6. Eisenhauer EA. Real-world evidence in the treatment of ovarian cancer. Ann Oncol. 2017;28:viii61–5.

    Article  CAS  PubMed  Google Scholar 

  7. Pignata S, Cecere SC, Du Bois A, Harter P, Heitz F. Treatment of recurrent ovarian cancer. Ann Oncol. 2017;28:viii51–6.

    Article  CAS  PubMed  Google Scholar 

  8. Jayson GC, Kohn EC, Kitchener HC, Ledermann JA. Ovarian cancer. Lancet. 2014;384(9951):1376–88. https://doi.org/10.1016/S0140-6736(13)62146-7.

    Article  PubMed  Google Scholar 

  9. Liang H, Yu T, Han Y, Jiang H, Wang C, You T, et al. LncRNA PTAR promotes EMT and invasion-metastasis in serous ovarian cancer by competitively binding miR-101-3p to regulate ZEB1 expression. Mol Cancer. 2018;17(1):119. https://doi.org/10.1186/s12943-018-0870-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Wu D-D, Chen X, Sun K-X, Wang L-L, Chen S, Zhao Y. Role of the lncRNA ABHD11-AS (1) in the tumorigenesis and progression of epithelial ovarian cancer through targeted regulation of RhoC. Mol Cancer. 2017;16(1):138. https://doi.org/10.1186/s12943-017-0709-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Liu P, Wang H, Liang Y, Hu A, Xing R, Jiang L, et al. LINC00852 promotes lung adenocarcinoma spinal metastasis by targeting S100A9. J Cancer. 2018;9(22):4139–49. https://doi.org/10.7150/jca.26897.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Li Q, Wang X, Jiang N, Xie X, Liu N, Liu J, et al. Exosome-transmitted linc00852 associated with receptor tyrosine kinase AXL dysregulates the proliferation and invasion of osteosarcoma. Cancer Med. 2020;9(17):6354–66. https://doi.org/10.1002/cam4.3303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Jiang W, Li T, Wang J, Jiao R, Shi X, Huang X, et al. miR-140-3p suppresses cell growth and induces apoptosis in colorectal cancer by targeting PD-L1. OncoTargets Ther. 2019;12:10275–85. https://doi.org/10.2147/OTT.S226465.

    Article  CAS  Google Scholar 

  14. Zhou Y, Wang B, Wang Y, Chen G, Lian Q, Wang H. miR-140-3p inhibits breast cancer proliferation and migration by directly regulating the expression of tripartite motif 28. Oncol Lett. 2019;17(4):3835–41. https://doi.org/10.3892/ol.2019.10038.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chen M-S, Lin C-H, Huang L-Y, Qiu X-M. CircRNA SMARCC1 sponges MiR-140-3p to regulate cell progression in colorectal cancer. Cancer Manag Res. 2020;12:4899–910. https://doi.org/10.2147/CMAR.S254185.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Salem O, Erdem N, Jung J, Münstermann E, Wörner A, Wilhelm H, et al. The highly expressed 5'isomiR of hsa-miR-140-3p contributes to the tumor-suppressive effects of miR-140 by reducing breast cancer proliferation and migration. BMC Genomics. 2016;17(1):566. https://doi.org/10.1186/s12864-016-2869-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Miles GD, Seiler M, Rodriguez L, Rajagopal G, Bhanot G. Identifying microRNA/mRNA dysregulations in ovarian cancer. BMC Res Notes. 2012;5(1):164. https://doi.org/10.1186/1756-0500-5-164.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Cooper HA, Scalia R, Rizzo V, Eguchi S. Angiotensin II- and Alzheimer-type cardiovascular aging. Circ Res. 2018;123(6):651–3. https://doi.org/10.1161/CIRCRESAHA.118.313477.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yap RWK, Shidoji Y, Yap WS, Masaki M. Association and interaction effect of AGTR1 and AGTR2 gene polymorphisms with dietary pattern on metabolic risk factors of cardiovascular disease in Malaysian adults. Nutrients. 2017;9(8):853. https://doi.org/10.3390/nu9080853.

    Article  CAS  PubMed Central  Google Scholar 

  20. Arrieta O, Guevara P, Escobar E, García-Navarrete R, Pineda B, Sotelo J. Blockage of angiotensin II type I receptor decreases the synthesis of growth factors and induces apoptosis in C6 cultured cells and C6 rat glioma. Br J Cancer. 2005;92(7):1247–52. https://doi.org/10.1038/sj.bjc.6602483.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Oh E, Kim JY, Cho Y, An H, Lee N, Jo H, et al. Overexpression of angiotensin II type 1 receptor in breast cancer cells induces epithelial–mesenchymal transition and promotes tumor growth and angiogenesis. Biochim Biophys Acta (BBA) – Mol Cell Res. 2016;1863(6):1071–81. https://doi.org/10.1016/j.bbamcr.2016.03.010.

    Article  CAS  Google Scholar 

  22. Ni L, Feng Y, Wan H, Ma Q, Fan L, Qian Y, et al. Angiotensin-(1-7) inhibits the migration and invasion of A549 human lung adenocarcinoma cells through inactivation of the PI3K/Akt and MAPK signaling pathways. Oncol Rep. 2012;27(3):783–90. https://doi.org/10.3892/or.2011.1554.

    Article  CAS  PubMed  Google Scholar 

  23. Chauhan VP, Martin JD, Liu H, Lacorre DA, Jain SR, Kozin SV, et al. Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels. Nat Commun. 2013;4(1):2516. https://doi.org/10.1038/ncomms3516.

    Article  CAS  PubMed  Google Scholar 

  24. Zhang Q, Yu S, Lam MMT, Poon TCW, Sun L, Jiao Y, et al. Angiotensin II promotes ovarian cancer spheroid formation and metastasis by upregulation of lipid desaturation and suppression of endoplasmic reticulum stress. J Exp Clin Cancer Res. 2019;38(1):116. https://doi.org/10.1186/s13046-019-1127-x.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Xie J, Xu Y, Huang X, Chen Y, Fu J, Xi M, et al. Berberine-induced apoptosis in human breast cancer cells is mediated by reactive oxygen species generation and mitochondrial-related apoptotic pathway. Tumor Biol. 2015;36(2):1279–88. https://doi.org/10.1007/s13277-014-2754-7.

    Article  CAS  Google Scholar 

  26. Milovic V, Teller IC, Murphy GM, Caspary WF, Stein J. Deoxycholic acid stimulates migration in colon cancer cells. Eur J Gastroenterol Hepatol. 2001;13(8):945–9. https://doi.org/10.1097/00042737-200108000-00012.

    Article  CAS  PubMed  Google Scholar 

  27. Ren J, Ding L, Zhang D, Shi G, Xu Q, Shen S, et al. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018;8(14):3932–48. https://doi.org/10.7150/thno.25541.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Liao D, Zhong L, Duan T, Zhang R-H, Wang X, Wang G, et al. Aspirin suppresses the growth and metastasis of osteosarcoma through the NF-κB pathway. Clin Cancer Res. 2015;21(23):5349–59. https://doi.org/10.1158/1078-0432.CCR-15-0198.

    Article  CAS  PubMed  Google Scholar 

  29. Wu X-S, Wang F, Li H-F, Hu Y-P, Jiang L, Zhang F, et al. LncRNA-PAGBC acts as a microRNA sponge and promotes gallbladder tumorigenesis. EMBO Rep. 2017;18(10):1837–53. https://doi.org/10.15252/embr.201744147.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Huang L, Zhou Y, Cao X-P, Lin J-X, Zhang L, Huang S-T, et al. KPNA2 promotes migration and invasion in epithelial ovarian cancer cells by inducing epithelial-mesenchymal transition via Akt/GSK-3&#946;/Snail activation. J Cancer. 2018;9:157–65. https://doi.org/10.7150/jca.20879.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wu S, Donigan A, Platsoucas CD, Jung W, Soprano DR, Soprano KJ. All-trans-retinoic acid blocks cell cycle progression of human ovarian adenocarcinoma cells at late G1. Exp Cell Res. 1997;232(2):277–86. https://doi.org/10.1006/excr.1997.3495.

    Article  CAS  PubMed  Google Scholar 

  32. Liu J, Guo Q, Chen B, Yu Y, Lu H, Li Y-Y. Cathepsin B and its interacting proteins, bikunin and TSRC1, correlate with TNF-induced apoptosis of ovarian cancer cells OV-90. FEBS Lett. 2006;580(1):245–50. https://doi.org/10.1016/j.febslet.2005.12.005.

    Article  CAS  PubMed  Google Scholar 

  33. Ricciardelli C, Lokman NA, Cheruvu S, Tan IA, Ween MP, Pyragius CE, et al. Transketolase is upregulated in metastatic peritoneal implants and promotes ovarian cancer cell proliferation. Clin Exp Metastasis. 2015;32(5):441–55. https://doi.org/10.1007/s10585-015-9718-1.

    Article  CAS  PubMed  Google Scholar 

  34. Wu X, Wu Y, Ye B, Wu F, Wang P. High expression of ghrelin and obestatin prepropeptide in tumor tissues predicted adverse overall survival in gastric carcinoma patients. Medicine. 2020;99(26):e20635. https://doi.org/10.1097/MD.0000000000020635.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Seim I, Carter SL, Herington AC, Chopin LK. Complex organisation and structure of the ghrelin antisense strand gene GHRLOS, a candidate non-coding RNA gene. BMC Mol Biol. 2008;9(1):95. https://doi.org/10.1186/1471-2199-9-95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Wu W, Gao H, Li X, Zhu Y, Peng S, Yu J, et al. LncRNA TPT1-AS1 promotes tumorigenesis and metastasis in epithelial ovarian cancer by inducing TPT1 expression. Cancer Sci. 2019;110(5):1587–98. https://doi.org/10.1111/cas.14009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yan H, Li H, Li P, Li X, Lin J, Zhu L, et al. Long noncoding RNA MLK7-AS1 promotes ovarian cancer cells progression by modulating miR-375/YAP1 axis. J Exp Clin Cancer Res. 2018;37(1):237. https://doi.org/10.1186/s13046-018-0910-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Shang A, Wang W, Gu C, Chen C, Zeng B, Yang Y, et al. Long non-coding RNA HOTTIP enhances IL-6 expression to potentiate immune escape of ovarian cancer cells by upregulating the expression of PD-L1 in neutrophils. J Exp Clin Cancer Res. 2019;38(1):411. https://doi.org/10.1186/s13046-019-1394-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wu S, Liu J, Wang X, Li M, Chen Z, Tang Y. Aberrant expression of the long non-coding RNA GHRLOS and its prognostic significance in patients with colorectal cancer. J Cancer. 2017;8(19):4040–7. https://doi.org/10.7150/jca.21304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Thomas PB, Seim I, Jeffery PL, Gahete MD, Maugham M, Crisp GJ, et al. The long non-coding RNA GHSROS facilitates breast cancer cell migration and orthotopic xenograft tumour growth. Int J Oncol. 2019;55(6):1223–36. https://doi.org/10.3892/ijo.2019.4891.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Liang S, Gong X, Zhang G, Huang G, Lu Y, Li Y. The lncRNA XIST interacts with miR-140/miR-124/iASPP axis to promote pancreatic carcinoma growth. Oncotarget. 2017;8(69):113701–18. https://doi.org/10.18632/oncotarget.22555.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Hao T, Wang Z, Yang J, Zhang Y, Shang Y, Sun J. MALAT1 knockdown inhibits prostate cancer progression by regulating miR-140/BIRC6 axis. Biomed Pharmacother. 2020;123:109666. https://doi.org/10.1016/j.biopha.2019.109666.

    Article  CAS  PubMed  Google Scholar 

  43. Liu W, Liu P, Gao H, Wang X, Yan M. Long non-coding RNA PGM5-AS1 promotes epithelial-mesenchymal transition, invasion and metastasis of osteosarcoma cells by impairing miR-140-5p-mediated FBN1 inhibition. Mol Oncol. 2020;14(10):2660–77. https://doi.org/10.1002/1878-0261.12711.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Li Y, Xu J, Guo YN, Yang BB. LncRNA SNHG20 promotes the development of laryngeal squamous cell carcinoma by regulating miR-140. Eur Rev Med Pharmacol Sci. 2019;23(8):3401–9. https://doi.org/10.26355/eurrev_201904_17704.

    Article  CAS  PubMed  Google Scholar 

  45. Zhang Q-Y, Men C-J, Ding X-W. Upregulation of microRNA-140-3p inhibits epithelial-mesenchymal transition, invasion, and metastasis of hepatocellular carcinoma through inactivation of the MAPK signaling pathway by targeting GRN. J Cell Biochem. 2019;120(9):14885–98. https://doi.org/10.1002/jcb.28750.

    Article  CAS  PubMed  Google Scholar 

  46. Okazaki M, Fushida S, Harada S, Tsukada T, Kinoshita J, Oyama K, et al. The angiotensin II type 1 receptor blocker candesartan suppresses proliferation and fibrosis in gastric cancer. Cancer Lett. 2014;355(1):46–53. https://doi.org/10.1016/j.canlet.2014.09.019.

    Article  CAS  PubMed  Google Scholar 

  47. Wang C-X, Xiong H-F, Wang S, Wang J, Nie X, Guo Q, et al. Overexpression of TEM8 promotes ovarian cancer progression via Rac1/Cdc42/JNK and MEK/ERK/STAT3 signaling pathways. Am J Transl Res. 2020;12(7):3557–76.

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhang D, Liu H, Yang B, Hu J, Cheng Y. L-securinine inhibits cell growth and metastasis of human androgen-independent prostate cancer DU145 cells via regulating mitochondrial and AGTR1/MEK/ERK/STAT3/PAX2 apoptotic pathways. Biosci Rep. 2019;39(5):BSR20190469. https://doi.org/10.1042/BSR20190469.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gharpure KM, Pradeep S, Sans M, Rupaimoole R, Ivan C, Wu SY, et al. FABP4 as a key determinant of metastatic potential of ovarian cancer. Nat Commun. 2018;9:2923.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Guo L, Chen J, Liu D, Liu L. OIP5-AS1/miR-137/ZNF217 Axis promotes malignant behaviors in epithelial ovarian cancer. Cancer Manag Res. 2020;12:6707–17. https://doi.org/10.2147/CMAR.S237726.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ge J, Han T, Shan L, Na J, Li Y, Wang J. Long non-coding RNA THOR promotes ovarian Cancer cells progression via IL-6/STAT3 pathway. J Ovarian Res. 2020;13(1):72. https://doi.org/10.1186/s13048-020-00672-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not Applicable.

Funding

This study was supported by the Natural Science Foundation of Liaoning Province, China (No.20170540570). The fund supported experimentation, data analysis, and writing of this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

JZ designed the study, LW1, JJ&JZ performed the experiment, ZQ wrote the manuscript, LW2&ZQ acquired and analyzed the data, ZQ&YJ reviewed the manuscript. PM provided financial and technical support and revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jing-ru Zhang or Peng Mu.

Ethics declarations

Ethics approval and consent to participate

Written informed consent was obtained from all ovarian cancer patients. All clinical experiments were approved by the institutional ethical review boards of Cancer Hospital of China Medical University. All animal experiments were approved by the Ethics Committee of Cancer Hospital of China Medical University. The number of the ethical approval is SYXK (Liao)2017-0004.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qiao, Zw., Jiang, Y., Wang, L. et al. LINC00852 promotes the proliferation and invasion of ovarian cancer cells by competitively binding with miR-140-3p to regulate AGTR1 expression. BMC Cancer 21, 1004 (2021). https://doi.org/10.1186/s12885-021-08730-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-021-08730-7

Keywords