Back to Journals » OncoTargets and Therapy » Volume 12

lncRNA CASC9 regulates cell migration and invasion in hemangioma endothelial cells by targeting miR-125a-3p/Nrg1

Authors Li X, Chen B, Chi D, Zhang Y, Jiang W

Received 30 July 2018

Accepted for publication 4 September 2018

Published 7 January 2019 Volume 2019:12 Pages 423—432

DOI https://doi.org/10.2147/OTT.S181914

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Dr William C. Cho



Xianwei Li, Bo Chen, Decai Chi, Yingnan Zhang, Weiliang Jiang

Department of Vascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, People’s Republic of China

Background: Despite being one of the most common benign tumors, the prevalence and pathogenesis of hemangiomas (HAs) are poorly understood. We aimed to identify the biological role of the long non-coding RNA (lncRNA) CASC9 in the HA-derived endothelial cell (HDECs) phenotype as well as elucidate the mechanism involved.
Methods: The expression of CASC9 was identified by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). the effect of CASC9 on cell proliferation, migration and invasion of HDECs were examined by CCK8, wound healing, and transwell assay, respectively. Bioinformatics analysis and a luciferase reporter assay were utilized to investigated the mechanisms involved. The in vivo tumorigenesis capability of CASC9 on HA was also evaluated.
Results: The expression of CASC9 was significantly elevated in HA tissue compared to normal tissue. Down-regulation of CASC9 inhibited proliferation, migration, and invasion of HDECs. The translation of cyclinD1, N-cadherin, Twist, and MMP2 was also decreased by CASC9 knockdown treatment. Furthermore, CASC9 over-expression exerted the opposite effect of proliferation, migration, and invasion of HDECs. We also found that CASC9 interacts with miR-125a-3p/Nrg1 to regulate cellular functions. Interestingly, miR-125a-3p can reverse the effect of CASC9 on proliferation, migration, and invasion of HDECs. Together, the clinical data showed that CASC9 expression is negatively correlated with miR-125a-3p expression and positively correlated with Nrg1 expression. CASC9 also exerted anti-tumorigenesis capability in vivo.
Conclusion: Our study indicates that CASC9 accelerates cell growth and invasion of HDECs and provides new insights for the diagnosis and molecular therapy of HA.

Keywords: CASC9, cell migration, invasion, hemangioma, miR-125a-3p, Nrg1

Introduction

Hemangioma (HA) is the most common benign tumor in infants at present, and the etiology and pathogenesis of HAs are not completely clear.1 About 75%–80% of HA patients are females, and HA often occurs about at birth 2 weeks after the birth of a child.2 Severe HA causes a heavy physical and mental burden to patients and their family members, especially due to the disfigured skin lesions, which affect the function and life of the skin.2,3 Severe HA is difficult to treat even in today’s medical field. To effectively control tumor growth and reduce the incidence of complications, many scholars advocate early diagnosis and combined treatments.4 Further defining, the pathogenesis of HAs plays a crucial role in early diagnosis and effective combined treatment.

Current advances in the identification of noncoding RNAs and their interaction with their target genes have enhanced our awareness of HA pathogenesis. Wang et al5 indicated that miR-199a-5p expression was decreased in HA, which could inhibit proliferation and induce apoptosis through targeting HIF1A in HA cells. Cao et al6 suggested that the long noncoding RNA (lncRNA), SNHG16, is overexpressed in HA and that it promotes proliferation, migration, and invasion of HA endothelial cells through regulation of the miR-520d-3p/STAT3 axis. The lncRNA, CASC9, is reported to be an oncogene in many cancers, regulating proliferation, migration, and invasion of cells.7 We found that abnormal CASC9 expression was observed in HA tissues. However, the biological role and molecular mechanism of CASC9 in HA remain unclear.

In the present study, we aimed to explore the expression and effect of CASC9 in HA and HA cell activities. We found that CASC9 was overexpressed in HA tissues. We also found CASC9 could modulate proliferation, migration, and invasion of HA-derived endothelial cells (HDECs) in vivo and in vitro and elucidated the underlying molecular mechanism involved.

Materials and methods

Tissue samples

Normal skin (n=43), involuting-phase HA tumor tissues (n=43), and proliferating-phase HA tumor tissues (n=43) were obtained from patients diagnosed with HA in the Second Affiliated Hospital of Harbin Medical University (Harbin, People’s Republic of China). Every patient provided written informed consent prior to this study. Among the patients with HAs, the HA tissues originated from the infants and children, of which there were 12 males and 31 females, and their age ranged from 3 months to 15 years (average age 24 months). The experimental protocol was approved by the Institutional Human Experiment and Ethic Committee of the Second Affiliated Hospital of Harbin Medical University, and the study was performed in accordance with the Declaration of Helsinki.

Cell culture and transfection

The primary HDECs used in these experiments were purchased from the Institute of Biochemistry and Cell Biology (Shanghai, People’s Republic of China). Cells were cultured in Dulbecco’s Modified Eagle’s Medium supplemented with 10% heat-inactivated fetal bovine serum, 100 U/mL of penicillin, and 100 μg/mL of streptomycin placed in a humidified atmosphere containing 5% CO2 at 37°C.

For cell transfection, miR-125a-39 mimics, siRNA-targeted CASC9, short hairpin RNA (shRNA)-targeted CASC9 (shLINC0129), and relative controls were obtained from GenePharma Co., Ltd. (Shanghai, People’s Republic of China). The transfection was performed by using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) in 5×106 cells at a final concentration of 50 nM. Subsequent assays were performed at 48 hours after transfection. To overexpress CASC9, HDECs were transfected with pCDNA3-CASC9 using Lipofectamine® 2000. After transfection for 24 hours, expression of CASC9 was validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR).

RT-qPCR

Total RNA was extracted from HA tissues and normal skin tissue using the TRIzol reagent (Thermo Fisher Scientific, Inc.) according to the manufacturer’s instructions. For miRNA analysis, RT-qPCR was performed using the TaqMan MicroRNA Reverse Transcription kit and TaqMan Universal PCR Master Mix (Applied Biosystems, Thermo Fisher Scientific, Inc.) according to the manufacturer’s instructions with the corresponding primers. Universal miRNA RT-qPCR primer was forward 5′-AACGAGACGACGACAGAC-3′; reverse 5′-GCAAATTCGTGAAGCGTTCCATA-3′ for U6; and forward 5′-GGGACAGGTGAGGTTCTTG-3′; reverse 5′-GAGTTGGAGGTTGCGTTAGA-3′ for miR-125a-3p. For mRNA analysis, RT-qPCR was performed using the TaqMan High-Capacity cDNA Reverse Transcription Kit and TaqMan Fast PCR Master Mix (Applied Biosystems; Thermo Fisher Scientific, Inc.) according to the manufacturer’s instructions with corresponding primers. GAPDH was used as an internal control to normalize Nrg1. The RT-qPCR protocol included an initial denaturation step (95°C for 5 minutes) and 40 cycles of denaturation (95°C for 10 seconds), annealing (60°C for 20 seconds), and extension (72°C for 10 seconds). The relative expression levels were calculated using 2−ΔΔCq method, as previously described. The primer sequences were as follows: CASC9 (forward, 5′-AGCAGCAAATGTGTCCATC-3′ and reverse, 5′-CAGACAGCAGCAAAGCAAT-3′), Nrg1 (forward, 5′-TGAATGGAGGGGAGTGCT-3′ and reverse, 5′-CAGGCAGAGACAGAAAGGG-3′), and GAPDH (forward, 5′-TCCTCTGACTTCAACAGCGACAC-3′ and reverse, 5′-CACCCTGTTGCTGTAGCCAAATTC-3′).

Cell proliferation assay

Cell proliferation was determined using the Cell Counting Kit-8 (CCK-8) assay. Briefly, cells were seeded into 96-well plates at 1×104 cells/well and cultured overnight. After transfection for 48 hours, 10 μL of CCK-8 reagent (Dojindo Molecular Technologies, Dojindo, Japan) was added to the wells and continuously cultured for 2 hours at 37°C. Absorbance at a wavelength of 450 nm was read on an automated microplate reader (BioTek Instruments, Winooski, VT, USA).

Colony formation assay

At 48 hours posttransfection, the cells were digested and resuspended into single-cell status. Then, 1,000 cells from each treatment were seeded into a 6-well plate coated with 0.4% agar medium. After culture for 14 days at 37°C, cells were fixed with 4% paraformaldehyde and stained with 0.1% crystal violet for 10 minutes. After washing with phosphate-buffered saline, colonies were observed and counted using an inverted microscope (Olympus CKX41; Olympus Corporation, Tokyo, Japan).

Cell migration

Cell migration was evaluated using a wound healing assay. In brief, at 48 hours after transfection, cells were cultured in 6-well plates (5×104 cells/well). Once cells were 90%–95% confluent, the monolayer was scratched using a sterile plastic micropipette tip, and then cells were cultured under standard conditions for 24 hours. Following several washes, recovery of the wound was observed and imaged using an X71 inverted microscope (Olympus Corporation).

Cell invasion

A transwell invasion assay was performed to determine cell invasion. Transfected cells (1×105) were seeded into the upper chamber of Matrigel-coated inserts with serum-free medium. Medium with 10% fetal bovine serum was added to the lower chamber as a chemo-attractant. The cells were allowed to invade for 48 hours at 37°C with 5% CO2. Then, cells that invaded to the lower surface of the filter were fixed in 70% ethanol for 30 minutes and stained with 0.1% crystal violet for 10 minutes at 25°C. The number of cells that migrated to the lower side was counted in five randomly selected fields under an X71 inverted microscope.

Bioinformatics analysis

The target miRNAs of CASC9 were predicted with computational algorithms, including starBase (http://starbase.sysu.edu.cn/agoClipRNA.php?source=lncRNA) and miRDB (http://mirdb.org/miRDB/index.html). miR-125a-3p ranked the top and was predicted as a target of CASC9. To find genes targeted by miR-125a-3p, we used the online software programs TargetScan (http://www.targetscan.org/) and miRDB (http://mirdb.org/miRDB/index.html). From the list of target genes obtained, we extracted all genes that could contribute to hemangioma progression. The 3′-UTR of the Nrg1 gene was predicted to have miR-125a-3p binding sites.

Luciferase activity assay

Wild-type CASC9 and mutant CASC9 were individually inserted into pmirGLO reporter vectors (Promega, Madison, WI, USA). HDECs were cotransfected with miR-125a-3p mimics and wild-type CASC9 or mutant CASC9 by Lipofectamine 2000 (Invitrogen). Relative luciferase activity was measured on a dual-luciferase reporter assay system (Promega) at 48 hours posttransfection. Data were expressed as the ratio of Renilla luciferase activity to firefly luciferase activity. Luciferase reporter assays to validate the direct binding of miR-125a-3p to Nrg1 3′-UTR were performed as described.

Western blot analysis

Proteins were extracted from cells using radioimmunoprecipitation assay buffer (Sigma-Aldrich, St Louis, MO, USA), and protein concentration was quantified using the BCA Protein Assay Kit (Beyotime Biotechnology, Haimen, People’s Republic of China). Equal amounts of proteins (50 μg) from each sample were separated by 10% sodium dodecyl sulfate–polyacrylamide gel and then transferred to a polyvinylidene fluoride membrane (Millipore, Boston, MA, USA). The membrane was blocked by 5% skimmed milk for 1 hour at 37°C. The target proteins were probed with primary anti-Nrg1 (ab180808, 1:1,000), anti-cyclin D1 (ab134175, 1:1,000), anti-N-cadherin (ab18203, 1:800), anti-Twist (ab49254, 1:1,000), anti-MMP2 (ab37150; 1:1,200), and anti-GAPDH (ab9485, 1:2,500) antibodies and incubated at 4°C overnight. Thereafter, the membrane was incubated with horseradish peroxidase-conjugated secondary antibodies (1:2,000; Beyotime Biotechnology) at 25°C for 1 hour. Immunoreactive protein bands were visualized using the enhanced chemiluminescence Western blot kit (Thermo Fisher Scientific). Protein band intensity was quantified using Image-Pro Plus 6.0 software (Media Cybernetics, Inc., Rockville, MD, USA). Relative protein expression was obtained by normalization against GAPDH.

In vivo xenograft experiments

Male BALB/c nude mice (6-week-old; n=4) were purchased from Beijing HFK Bioscience Co. Ltd. (Beijing, People’s Republic of China) and maintained under pathogen-free conditions after receipt of approval by the Committee of the Second Affiliated Hospital of Harbin Medical University. For tumor propagation analysis, 1×107 HDECs were subcutaneously injected into BALB/c nude mice. Tumor volume was calculated using the formula, volume = πab2/6 (a, tumor length; b, tumor width), at indicative time points. Tumor weight was measured at week 3 postinjection. Animal experiments were performed in accordance with relevant guidelines and regulations of the Animal Care and Use Committees at the Second Affiliated Hospital of Harbin Medical University.

Statistical analysis

Data are presented as mean ± SD. Data were statistically analyzed by Student’s t-test or one-way analysis of variance using SPSS version 19.0 (IBM Corporation, Armonk, NY, USA). Correlation between CASC9 and miR-125a-3p or Nrg1 expression in clinical specimens was analyzed using Pearson’s correlation analysis. A P-value of <0.05 was considered statistically significant.

Results

CASC9 was overexpressed in HA tissues and HDECs

To evaluate the biological role of CASC9 in HAs, the expressions of CASC9 in three different types of tissues were first examined by RT-PCR. The result suggested that the levels CASC9 expression were obviously higher in proliferating-phase HA tissues than that in involuting-phase HA tissues and normal tissues (P<0.01, Figure 1A). The CASC9 expression was increased significantly in proliferating-phase HA tissues compared with that in involuting-phase HA tissues (P<0.05, Figure 1A). HDECs then were transfected with siCASC9, CASC9, or a negative control (NC) plasmid. The transfection efficiency was detected by RT-PCR (P<0.001, Figure 1B and C).

Figure 1 The expressions of CASC9 in HAs patients.
Notes: (A) The expressions of CASC9 in normal skin tissues (n=43), involuting-phase HAs (n=43), and proliferating-phase HAs (n=43) were detected by RT-PCR analysis. (B, C) HDECs then were transfected with siCASC9, or CASC9, or NC plasmid. The transfection efficiency was detected by RT-PCR. Data are presented as the mean ± SD. *P<0.05, **P<0.01, and ***P<0.001.
Abbreviations: HA, hemangioma; HDEC, HA-derived endothelial cell; NC, negative control; RT-PCR, reverse transcription-polymerase chain reaction.

CASC9 regulated proliferation, invasion, and migration of HDECs

To identify the effect of CASC9 on the cellular phenotype of HDECs, cell proliferation, migration, and invasion were assessed. It was shown that siCASC9 significantly inhibited proliferation and colony formation of HDECs (P<0.01, Figure 2A and C). Remarkably, overexpression of CASC9 could promote proliferation and colony forming capability compared with NC treatment (P<0.001, Figure 2B and C). Cell migration and invasion were then detected by a wound healing and transwell assay. The results suggested that downregulation of CASC9 significantly inhibited cell migration and invasion, whereas overexpression of CASC9 exerted the opposite effect (P<0.001, Figure 2D and E). The expressions of several oncogenic proteins were also measured by Western blot analysis, and the results revealed that siCASC9 could significantly inhibit the expression of cyclin D1, N-cadherin, Twist, and MMP2 (P<0.01, Figure 2F). Upregulation of CASC9 obviously increased the cyclin D1, N-cadherin, Twist, and MMP2 expression (P<0.001, Figure 2F). These results indicated that CASC9 exhibits a tumorigenic role in HAs progression.

Figure 2 Downregulation of CASC9 inhibited cell proliferation, migration, and invasion of angiosarcoma cells.
Notes: (A, B) The proliferation of HDECs transfected with siCASC9 or CASC9 plasmid was detected by CCK-8. (C) Cell cloning ability of HDECs transfected with siCASC9 or CASC9 plasmid was detected by cell cloning formation assay. (D) The migration of HDECs transfected with siCASC9 or CASC9 plasmid was determined by wound healing assay. (E) The invasion of HDECs transfected with siCASC9 or CASC9 plasmid was examined by transwell assay. (F) The expressions of cyclin D1, N-cadherin, Twist, and MMP2 in HDECs transfected with siCASC9 or CASC9 plasmid were detected by Western blot analysis. GAPDH was used as a loading control. Data are presented as the mean ± SD. **P<0.01, ***P<0.001.
Abbreviations: CCK-8, Cell Counting Kit-8; HA, hemangioma; HDEC, HA-derived endothelial cell; NC, negative control.

CASC9 interacted with miR-125a-3p

To identify the mechanism by which CASC9 regulates proliferation, migration, and invasion of HDECs, the target miRNAs of CASC9 were predicted with computational algorithms, including TargetScan (http://targetscan.org/vert_71/) and miRDB (http://mirdb.org/miRDB/index.html). miR-125a-3p was predicted as a target of CASC9, and the predicted miR-125a-3p interaction site in CASC9 is illustrated in Figure 3A. To determine the interaction between CASC9 and miR-125a-3p, a dual-luciferase reporter assay was performed. The results suggested that overexpression of miR-125a-3p significantly decreased the luciferase activity of the reporter containing wild-type CASC9 in HDECs (P<0.001), while the activity of the reporter with the mutated miR-125a-3p binding site was not repressed (Figure 3B). The expression of miR-125a-3p in HDECs transfected with siCASC9 or CASC9 was also examined by RT-PCR, and the expression of miR-125a-3p was significantly increased in siCASC9-treated HDECs and notably decreased in CASC9 overexpression HDECs (P<0.001, Figure 3C). Furthermore, Pearson’s correlation analysis indicated that miR-125a-3p expression was negatively correlated with CASC9 expression in HA tissues (P<0.001, Figure 3D).

Figure 3 CASC9 interacted with 125a-3p.
Notes: (A) miR-125a-3p binding sites in CASC9 were predicted by bioinformatics analysis. (B) Luciferase reporter assays were performed using HDECs cells cotransfected with the miR-125a-3p mimics and CASC9-wt or CASC9-mut reporter plasmid. (C) Analysis of miR-125a-3p expression levels in HDECs cells transfected with CASC9 overexpressing plasmid or siCASC9 was performed by RT-PCR. (D) Pearson’s correlation analysis was used to determine the correlations between the levels of CASC9 and miR-125a-3p in HAs (n=43). Data are presented as the mean ± SD. ***P<0.001.
Abbreviations: HA, hemangioma; HDEC, HA-derived endothelial cell; Mut, mutation; RT-PCR, reverse transcription-polymerase chain reaction; wt, wild type.

miR-125a-3p reversed the effect of siCASC9 on HDECs

To further investigate whether the regulation of proliferation, migration, and invasion of HDECs by CASC9 was through regulation of miR-125a-3p, HDECs were transfected with CASC9 and miR-125a-3p mimics. Cell viability, migration, and invasion were analyzed using CCK-8, cell cloning formation, and wound healing and transwell assays, respectively. CASC9 overexpression significantly suppressed proliferation, migration, and invasion in HDECs, whereas miR-125a-3p mimics reversed the promoting effect of CASC9 on proliferation (P<0.001), migration, and invasion (P<0.001, Figure 4AD). In addition, upregulation of CASC9 also increased the expressions of cyclin D1, N-cadherin, Twist, and MMP2 (P<0.001), whereas miR-125a-3p significantly suppressed the increased expressions induced by CASC9 (P<0.001, Figure 4E). The results demonstrated that CASC9 exerts its oncogenic role by interacting with miR-125a-3p.

Figure 4 CASC9 regulated cell proliferation, migration, and invasion through targeting miR-125a-3p.
Notes: Cell proliferation (A), cell cloning formation (B), cell migration (C), and cell invasion (D) of HDECs transfected with NC, or CASC9, or CASC9+ miR-125a-3p mimics was identified by CCK-8, cell cloning formation, wound healing, and transwell assay, respectively. (E) The expressions of cyclin D1, N-cadherin, Twist, and MMP2 in HDECs transfected with NC, or CASC9, or CASC9+ miR-125a-3p mimics were detected by Western blot analysis. GAPDH was served as a loading control. Data are presented as the mean ± SD. ***P<0.001.
Abbreviations: CCK-8, Cell Counting Kit-8; HA, hemangioma; HDEC, HA-derived endothelial cell; NC, negative control.

Nrg1 was a target of miR-125a-3p

To identify the molecular mechanism behind CASC9 regulation of proliferation, migration, and invasion of HDECs through miR-125a-3p, TargetScan was used to predict miR-125a-3p targets (http://targetscan.org/vert_71/). Nrg1 was predicted as a target of miR-125a-3p, and the predicted miR-125a-3p interaction site in CASC9 is illustrated in Figure 5A. To determine the interaction between miR-125a-3p and Nrg1, a dual-luciferase reporter assay was performed. Remarkably, we found that miR-125a-3p mimics decreased the luciferase activity of the reporter containing wild-type Nrg1 in HDECs (P<0.001), while it failed to repress the reporter with a mutated miR-125a-3p binding site (Figure 5B). Protein expression of Nrg1 in HDECs transfected with miR-125a-3p mimics or an NC was examined by Western blot, and Nrg1 levels were reduced in HDECs treated with miR-125a-3p mimics (P<0.001, Figure 5C). Moreover, Pearson’s correlation analysis indicated that Nrg1 expression was positively correlated with CASC9 expression in HA tissues (P<0.001, Figure 5D). Finally, downregulation of CASC9 significantly inhibited Nrg1 expression in HDECs (P<0.001), while overexpression of CASC9 had the opposite effect (P<0.001, Figure 5E). The results showed that CASC9 regulates cell proliferation, migration, and invasion via interacting with the miR-125a-3p/Nrg1 axis.

Figure 5 Nrg1 was a target of miR-125a-3p.
Notes: (A) miR-125a-3p binding sites in Nrg1 were predicted by bioinformatics analysis. (B) Luciferase reporter assays were performed using HDECs cells cotransfected with the miR-125a-3p mimics and Nrg1-wt or Nrg1-mut reporter plasmid. (C) Analysis of Nrg1 expression levels in HDECs cells transfected with miR-125a-3p mimics or NC was performed by Western blot. GAPDH served as a loading control. (D) Pearson’s correlation analysis was used to determine the correlations between the levels of CASC9 and Nrg1 in HAs (n=43). (E) Analysis of Nrg1 expression levels in HDECs cells transfected with CASC9 overexpressing plasmid or siCASC9 was performed Western blot. GAPDH served as a loading control. Data are presented as the mean ± SD. ***P<0.001.
Abbreviations: HA, hemangioma; HDEC, HA-derived endothelial cell; Mut, mutation; NC, negative control; wt, wild type.

Downregulation of CASC9 inhibited tumor growth in vivo

The in vivo carcinogenic effect of CASC9 on HAs was evaluated via a xenograft mouse model. HDECs transfected with shNC or shCASC9 were implanted subcutaneously into nude mice. Then, the tumor growth was detected every 3 days. The results suggested that CASC9 knockdown significantly delayed tumor growth in vivo (P<0.01, Figure 6A). At 3 weeks postimplantation, the nude mice were sacrificed, and tumors were harvested and weighed. CASC9 knockdown significantly decreased the tumor size and weight (P<0.01, Figure 6B and C). These results reveal that CASC9 can promote HAs progression in vivo.

Figure 6 Downregulation of CASC9 inhibited cell growth in vivo.
Notes: (A) Tumor growth curves were established by detecting tumor volume every 3 for 21 days after injection. (B, C) Tumor weights isolated from nude mice in each treatment group were determined on day 21 after injection. Data are presented as the mean ± SD. **P<0.01, ***P<0.001.
Abbreviation: NC, negative control.

Discussion

Previous studies reported that the lncRNA CASC9 functions as an elicitor in various kinds of human tumors, such as esophageal squamous cell cancer,8 hepatocellular carcinoma,7 and lung adenocarcinoma.9 CASC9 also correlates with various biological activities of tumor cells, especially cell proliferation, invasion, and migration.10 In this study, we aimed to deeply explore the impact of CASC9 on the cellular physiology of HDECs and elucidate the underlying molecular mechanism. We first found that CASC9 was overexpressed in HA tumor tissues. These results indicated that CASC9 might act as an oncogene in HAs.

Emerging studies have indicated that lncRNAs regulate proliferation, migration, and invasion of cancer cells.11,12 Zhou et al9 indicated that CASC9 promoted the proliferation, migration, and invasion of lung adenocarcinoma cells. In the current study, HDECs were transfected with siCASC9 or a CASC9 overexpression plasmid. The findings suggest that siCASC9 can suppress cell viability, migration, and invasion of HDECs, while the upregulation of CASC9 shows a simulative effect on cellular activities. The expression of oncogenic proteins, including cyclin D1, N-cadherin, Twist, and MMP2, was significantly decreased by downregulation of CASC9 and promoted by its overexpression. These results demonstrate that CASC9 can affect the cellular physiology of HDECs through regulating oncogenic proteins.

miRNAs have been considered as a group of independent regulators in various cancers. Many studies indicate that miRNAs are the target genes of lncRNAs to participate in tumorigenesis. They modulate numerous biological activities in human tumors via interacting with lncRNAs or target mRNAs. The target miRNA of CASC9 was also predicted and verified, and miR-125a-3p was shown to interact with CASC9 in HDECs. Previous studies suggested that miR-125a-3p acts as a tumor suppressor, and that its expression is decreased in non-small-cell lung cancer,13 gastric cancer,14 prostate cancer,15 breast cancer,16 and colon cancer.17 Upregulation of miR-125a-3p inhibits proliferation, migration, and invasion in tumor cells.18 In our study, we found that CASC9 overexpression effectively promoted cell proliferation, migration, and invasion, an effect that could be reversed by treatment with miR-125a-3p mimics in HDECs. Furthermore, the clinical data showed that CASC9 expression was negatively correlated with miR-125a-3p expression. To deeply investigate the mechanism by which CASC9 exerted its tumorigenesis role in HAs, the target of miR-125a-3p was also predicted. NRG1 was confirmed as a target of miR-125a-3p, which is recognized as an oncogene in gastric cancer,19 colon cancer,20 lung cancer,21 and papillary thyroid cancer.22 Yin et al23 reported that miR-125a-3p affected cell apoptosis and invasion through regulating Nrg1 in human glioma cells. Our study also demonstrated that CASC9 expression was positively correlated with miR-125a-3p expression in HAs. Finally, the in vivo effect of CASC9 downregulation on cell growth was examined. These findings suggested that CASC9 promoted HDECs proliferation, migration, and invasion through interacting with the miR-125a-3p/Nrg1 axis.

Conclusion

Our study demonstrated that CASC9 overexpression in HAs can modulate cell proliferation, migration, and invasion by regulating miR-125a-3p/Nrg1. CASC9 promotes progression of HAs, a finding that might provide novel insights into HA diagnosis and molecular therapy.

Author contributions

Xianwei Li and Bo Chen conceived and designed the study. Literature search and data extraction were performed by Decai Chi and Yingnan Zhang. Xianwei Li and Decai Chi analyzed the data. Weiliang Jiang drafted the manuscript. All authors contributed to data analysis, drafting and revising the article, gave final approval of the version to be published, and agree to be accountable for all aspects of the work.

Disclosure

The authors report no conflicts of interest in this work.


References

1.

Malakan Rad E, Radmehr H, Vasei M, Rahimi Rastgoo B. Giant congenital right atrial epithelioid-capillary hemangioma with prolonged QT interval: Case report and practical surgical treatment strategy for primary cardiac tumors in children based on 25-year review of 299 cases. Echocardiography. 2018;35(9):1471–1481.

2.

Chibbaro S, Cebula H, Ganau M, et al. Multidisciplinary management of an intra-sellar cavernous hemangioma: Case report and review of the literature. J Clin Neurosci. 2018;52(52):135–138.

3.

Aka KE, Apollinaire Horo G, Fomba M, et al. A rare case of important and recurrent abnormal uterine bleeding in a post partum woman caused by cavernous hemangioma: a case report and review of literature. Pan Afr Med J. 2017;28:130.

4.

Chen YL, Hu XD, Xu NJ, Jiang WY, Ma WH. Surgical treatment of compressive spinal hemangioma: A case series of three patients and literature review. Orthopade. 2018;47(3):221–227.

5.

Wang Y, Dai YX, Wang SQ, et al. miR-199a-5p inhibits proliferation and induces apoptosis in hemangioma cells through targeting HIF1A. Int J Immunopathol Pharmacol. 2018;31(31):1832658285.

6.

Cao X, Xu J, Yue D. LncRNA-SNHG16 predicts poor prognosis and promotes tumor proliferation through epigenetically silencing p21 in bladder cancer. Cancer Gene Ther. 2018;25(1–2):10–17.

7.

Klingenberg M, Groß M, Goyal A, et al. The lncRNA CASC9 and RNA binding protein HNRNPL form a complex and co-regulate genes linked to AKT signaling. Hepatology. 2018 Epub 2018 May 23.

8.

Gao GD, Liu XY, Lin Y, Liu HF, Zhang GJ. LncRNA CASC9 promotes tumorigenesis by affecting EMT and predicts poor prognosis in esophageal squamous cell cancer. Eur Rev Med Pharmacol Sci. 2018;22(2):422–429.

9.

Zhou J, Xiao H, Yang X, et al. Long noncoding RNA CASC9.5 promotes the proliferation and metastasis of lung adenocarcinoma. Sci Rep. 2018;8(1):37.

10.

Liang Y, Chen X, Wu Y, et al. LncRNA CASC9 promotes esophageal squamous cell carcinoma metastasis through upregulating LAMC2 expression by interacting with the CREB-binding protein. Cell Death Differentiation. Epub 2018 Mar 6.

11.

Sadek KM, Lebda MA, Nasr NE, Nasr SM, El-Sayed Y. Role of lncRNAs as prognostic markers of hepatic cancer and potential therapeutic targeting by S-adenosylmethionine via inhibiting PI3K/Akt signaling pathways. Environ Sci Pollut Res Int. 2018;25(20):20057–20070.

12.

Zhao J, Fu Y, Wu J, Li J, Huang G, Qin L. The Diverse Mechanisms of miRNAs and lncRNAs in the Maintenance of Liver Cancer Stem Cells. Biomed Res Int. 2018;2018:8686027.

13.

Jiang L, Huang Q, Zhang S, et al. Hsa-miR-125a-3p and hsa-miR-125a-5p are downregulated in non-small cell lung cancer and have inverse effects on invasion and migration of lung cancer cells. BMC Cancer. 2010;10(10):318.

14.

Hashiguchi Y, Nishida N, Mimori K, et al. Down-regulation of miR-125a-3p in human gastric cancer and its clinicopathological significance. Int J Oncol. 2012;40(5):1477–1482.

15.

Ninio-Many L, Grossman H, Levi M, et al. MicroRNA miR-125a-3p modulates molecular pathway of motility and migration in prostate cancer cells. Oncoscience. 2014;1(4):250–261.

16.

Xu X, Lv YG, Yan CY, Yi J, Ling R. Enforced expression of hsa-miR-125a-3p in breast cancer cells potentiates docetaxel sensitivity via modulation of BRCA1 signaling. Biochem Biophys Res Commun. 2016;479(4):893–900.

17.

Wang J, Yan F, Zhao Q, et al. Circulating exosomal miR-125a-3p as a novel biomarker for early-stage colon cancer. Sci Rep. 2017;7(1):4150.

18.

Liang L, Gao C, Li Y, et al. miR-125a-3p/FUT5-FUT6 axis mediates colorectal cancer cell proliferation, migration, invasion and pathological angiogenesis via PI3K-Akt pathway. Cell Death Dis. 2017;8(8):e2968.

19.

Yun S, Koh J, Nam SK, et al. Clinical significance of overexpression of NRG1 and its receptors, HER3 and HER4, in gastric cancer patients. Gastric Cancer. 2018;21(2):225–236.

20.

Stahler A, Heinemann V, Neumann J, et al. Prevalence and influence on outcome of HER2/neu, HER3 and NRG1 expression in patients with metastatic colorectal cancer. Anticancer Drugs. 2017;28(7):717–722.

21.

Xia D, Le LP, Iafrate AJ, Lennerz J. KIF13B-NRG1 Gene Fusion and KRAS Amplification in a Case of Natural Progression of Lung Cancer. Int J Surg Pathol. 2017;25(3):238–240.

22.

Zhang TT, Qu N, Sun GH, et al. NRG1 regulates redox homeostasis via NRF2 in papillary thyroid cancer. Int J Oncol. 2018;53(2):685–693.

23.

Yin F, Zhang JN, Wang SW, et al. MiR-125a-3p regulates glioma apoptosis and invasion by regulating Nrg1. PLoS One. 2015;10(1):e116759.

Creative Commons License © 2019 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.