Skip to main content

Long non-coding RNA KCNQ1OT1 promotes cell viability and migration as well as inhibiting degradation of CHON-001 cells by regulating miR-126-5p/TRPS1 axis

Abstract

Background

Osteoarthritis (OA) is defined as a degenerative disease. Pivotal roles of long non-coding RNA (lncRNAs) in OA are widely elucidated. Herein, we intend to explore the function and molecular mechanism of lncRNA KCNQ1OT1 in CHON-001 cells.

Methods

Relative expression of KCNQ1OT1, miR-126-5p and TRPS1 was determined by quantitative real-time polymerase chain reaction (qRT-PCR). Cell viability was examined by MTT assay. The migratory ability of chondrocytes was assessed by transwell assay. Western blot was used to determine relative protein expression of collagen II, MMP13 and TRPS1. Dual-luciferase reporter (DLR) assay was applied to test the target of lncRNA KCNQ1OT1 or miR-126-5p.

Results

Relative expression of KCNQ1OT1 and TRPS1 was reduced, whereas miR-126-5p was augmented in cartilage tissues of post-traumatic OA patients compared to those of subjects without post-traumatic OA. Increased KCNQ1OT1 or decreased miR-126-5p enhanced cell viability and migration, and repressed extracellular matrix (ECM) degradation in CHON-001 cells. MiR-126-5p was the downstream target of KCNQ1OT1, and it could directly target TRPS1. There was an inverse correlation between KCNQ1OT1 and miR-126-5p or between miR-126-5p and TRPS1. Meantime, there was a positive correlation between KCNQ1OT1 and TRPS1. The promoting impacts of KCNQ1OT1 on cell viability and migration as well as the suppressive impact of KCNQ1OT1 on ECM degradation were partially abolished by miR-126-5p overexpression or TRPS1 knockdown in CHON-001 cells.

Conclusions

Overexpression of KCNQ1OT1 attenuates the development of OA by sponging miR-126-5p to target TRPS1. Our findings may provide a possible therapeutic strategy for human OA in clinic.

Introduction

Osteoarthritis (OA) is a degenerative bone and joint disease, which is featured by destruction of joint matrix and the decrease of articular chondrocytes [1, 2]. Its clinical symptoms include swelling, stiffness and arthralgia [3]. As previously reported, OA can be caused by diverse factors, such as genetic predisposition, failure of nutrient supply, abnormal mechanical loading and trauma [1]. Currently, drug treatment, stem cell treatment, electromagnetic therapy and acupuncture therapy have been clinically applied for OA therapy [4,5,6,7]. However, the treatment effect is unsatisfactory. Thence, it is imperative to explore more effective targets for OA management.

Recent years, the crucial roles of long non-coding RNAs (lncRNAs) in OA have been gradually recognized [8,9,10]. For instance, MALAT1 and MFI2-AS1 are deeply involved in chondrocytes proliferation and extracellular matrix (ECM) degradation in OA [8, 9]. Similarly, TUG1 can facilitate ECM degradation of chondrocytes in OA [10]. Notably, lncRNA KCNQ1 overlapping transcript 1 (KCNQ1OT1), located in the KCNQ1 locus at 11p15.5 [11], has been reported to play important role in the proliferation and differentiation of chondrocytes in fracture or osteolysis [12, 13]. However, the role and mechanism of lncRNA KCNQ1OT1 in OA are not elucidated.

It is well known that lncRNAs participate in post-transcriptional regulation by binding to microRNAs (miRNAs) [14]. Ample evidence has revealed that miRNAs are critical regulators in the pathogenesis of OA [15], such as miR-335-5p [16] and miR-140 [17]. More importantly, miR-126 is also uncovered to possess a regulatory role in OA [18, 19]. For example, inhibition of miR-126 promotes the viability and migration of chondrocytes in a cell model of OA [18]. The viability of chondrocytes is enhanced by down-regulation of miR-126 [19]. In addition, miR-126 is revealed to be a target of lncRNA DNM3OS to regulate OA progression [19]. Nonetheless, the regulatory effect of lncRNA KCNQ1OT1 on miR-126-5p in OA has not been explored.

Tricho-rhino-phalangeal syndrome type I (TRPS1) is known as an oncogene to promote cell proliferation and metastasis in several types of human cancers, such as breast cancer [20], osteosarcoma [21], and colon cancer [22]. In recent years, the promoting role of TRPS1 in chondrocytes differentiation and proliferation has attracted increasing attention [23, 24]. Wuelling et al. found that loss of TRPS1 represses the differentiation of chondrocytes via Wnt5a [23]. Suemoto et al. demonstrated that overexpression of TRPS1 facilitates chondrocytes proliferation by reduction the level of Stat3 [24]. However, studies of TRPS1 effects on OA are limited.

Here, the roles of lncRNA KCNQ1OT1 and miR-126-5p were examined in CHON-001 cells. To further explore the regulatory mechanism of lncRNA KCNQ1OT1, the regulatory relation among KCNQ1OT1, miR-126-5p and TRPS1 was tested in CHON-001 cells. This investigation provided a new theoretical reference for OA treatment.

Materials and methods

Patients

The OA cartilage tissues were obtained from knees of post-traumatic OA (PTOA) patients (14 males and 9 females, an average age of 56.58 years) who underwent total knee arthroplasty. Normal cartilage tissues were collected from subjects without PTOA (8 males and 8 females, an average age of 51.66 years) who underwent the amputation. All samples were collected from our hospital between March 2017 and December 2019. The tissues samples were used to detect the expression levels of KCNQ1OT1, miR-126-5p, and TRPS1, as well as the correlations among them. The current study was approved by the ethics committees of our hospital, and all participants had signed written informed consents.

Cell culture

A human chondrocyte cell line (CHON-001) was bought from American Type Culture Collection (ATCC; Manassas, Virginia, USA). CHON-001 cells were grown in Dulbecco’s Modified Eagle’s Medium (DMEM; Gibco, Waltham, MA, USA) supplemented with 0.1 mg/mL G-418 (Gibco) and 10% fetal bovine serum (FBS; Gibco), and maintained in an incubator with a temperature of 37 °C and 5% CO2.

Cell transfection

The pcDNA3.1-negative control (NC), pcDNA3.1-KCNQ1OT1 (containing full-length of KCNQ1OT1), short hairpin (sh)-NC, sh-KCNQ1OT1, mimics NC, miR-126-5p mimics, inhibitor NC, miR-126-5p inhibitor, sh-TRPS1 were bought from RiboBio (Guangzhou, China). Then these oligonucleotides and plasmids were transfected into CHON-001 cells through Lipofectamine3000 (Invitrogen, Carlsbad, CA, USA) for 48 h. Afterwards, the transfected CHON-001 cells were used to perform the functional experiments of KCNQ1OT1/miR-126-5p/TRPS1 axis in vitro.

Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR)

Total RNA from cartilage tissues and CHON-001 cells was extracted using TRIzol® reagent (Invitrogen), and RNA concentration was detected by NanoDropND-1000 spectrophotometer. After that, extracted RNA was converted to complementary DNA (cDNA) via a PrimeScript™ RT reagent kit (Takara, Beijing, China). The cDNA was used for performing qRT-PCR according to instructions of a SYBR Green PCR kit (Takara). The reaction procedures were shown as follows: 95 °C for 20 s, 35 cycles of 95 °C for 10 s, 60 °C for 30 s and 72 °C for 10 s. All primers (Table 1) were bought from Sangon Biotech (Shanghai, China). Finally, relative expression of KCNQ1OT1, miR-126-5p and TRPS1 was calculated via the 2-ΔΔCt method. U6 and GADPH were used as internal references.

Table 1 Primers for qRT-PCR

3-(4, 5-Dimethyl-2-Thiazolyl)-2, 5-Diphenyl-2-H-tetrazolium bromide (MTT) assay

CHON-001 cells (6 × 103/well) were seeded in 96-well plates, and then reacted with MTT (10 μL). After 4 h of reaction, and dimethyl sulfoxide (150 μL) were added into each well for dissolving formazan crystals. Cell viability was assessed by measuring the optical density (OD) at 450 nm using a microplate reader (BMG LABTECH, Durham, NC, USA).

Dual Luciferase Reporter (DLR) assay

The 3′-UTR sequence of KCNQ1OT1 or TRPS1 containing miR-126-5p binding sites was cloned into a pGL3 Basic Vector (Promega, Madison, WI) to construct KCNQ1OT1 wt or TRPS1 wt. KCNQ1OT1 mut or TRPS1 mut was obtained by mutating binding sites of miR-126-5p and then introduced into the pGL3 Basic Vector (Promega). Then CHON-001 cells were transfected with above plasmids along with mimics NC/miR-126-5p mimics. Following 48 h of transfection, Firefly and Renilla luciferase activity was examined through a dual-luciferase reporter assay system (Biotek, Winooski, VT, USA). Relative luciferase activity was defined as the ratio of Firefly luciferase activity and Renilla luciferase activity.

Western blot

Total proteins were isolated from CHON-001 cells using RIPA lysis (Beyotime, Shanghai, China), and the concentration of proteins was measured through a bicinchoninic acid kit (Pierce, Waltham, MA, USA). Next, protein samples were separated by sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) gel, followed by transferring onto polyvinylidene difluoride (PVDF) membranes (Millipore, Billerica, MA, USA). After being blocked with 5% skim milk, the membranes were incubated with the following primary antibodies overnight at 4 °C: anti-TRPS1 (1:1000, ab227867, Abcam), anti-collagen II (1:2000, ab188570, Abcam), anti-MMP13 (1:1000, ab51072, Abcam) and anti-α-tubulin (1:2000, ab52866, Abcam). After the membranes were washed with tris buffered saline Tween (TBST), the membranes were nurtured with the secondary antibody (1:10,000, ab205718, Abcam) at 37 °C for 2 h. Finally, the immune-reactivity was visualized using electrochemiluminescence (Thermo Fisher Scientific, Shanghai, China), and relative protein expression of MMP13, TRPS1 and collagen II over α-tubulin was quantified via Gel-pro Analyzer software (Media Cybernetics, Maryland, USA).

Transwell assay

For determining the migratory ability of CHON-001 cells, transwell assay was carried out with the 24-well transwell chamber (8-μm pore size; Corning, Cambridge, USA). CHON-001 cells (6 × 105 cells) in FBS-free medium were plated into the upper layer of the transwell chamber. Subsequently, the lower chamber was filled with 600 μL DMEM including 10% FBS. After incubation for 24 h, migratory cells on the lower chamber were fixed in 4% paraformaldehyde for 15 min and stained with 0.5% crystal violet for 10 min. The numbers of migratory cells were counted under a Leica microscope (Leica Microsystems, Wetzlar, Germany). The percentage of migration was calculated as the following formula: (the numbers of migratory cells in the treat group)/ (the numbers of migratory cells in the control group) × 100%.

Statistical analysis

Data were analyzed by the SPSS 22.0 software (IBM, Armonk, NY, USA). All results were displayed as the mean ± standard deviation. One-way ANOVA was utilized for multiple comparisons, followed by Tukey’s post hoc test. Student’s t-test was applied to compare differences between two groups. A value of P < 0.05 was regarded as statistically significant.

Results

LncRNA KCNQ1OT1 promotes cell viability and migration, and represses ECM degradation in CHON-001 cells

In order to probe the function of lncRNA KCNQ1OT1 in OA, the expression of KCNQ1OT1 was firstly determined in PTOA patients. We found that KCNQ1OT1 expression was lower in the PTOA group than that in the control group (P < 0.001, Fig. 1A). Subsequently, lncRNA KCNQ1OT1 was overexpressed or silenced in CHON-001 cells. As expected, KCNQ1OT1 expression was evidently diminished by addition of sh-KCNQ1OT1, and distinctly boosted by addition of pcDNA3.1-KCNQ1OT1 in CHON-001 cells (all P < 0.001, Fig. 1B). Then functional experiments were performed. Results from MTT and transwell assays displayed that the viability (P < 0.001, Fig. 1C) and migratory ability of CHON-001 cells (P < 0.01, Fig. 1D) were enhanced when KCNQ1OT1 was overexpressed, whereas silencing of KCNQ1OT1 resulted in opposite results. Moreover, data from western blot indicated that up-regulation of KCNQ1OT1 elevated relative protein expression of the ECM protein (collagen II) (P < 0.01, Fig. 1E), whereas reduced relative protein expression of the cartilage-degrading enzyme (MMP13) in CHON-001 cells (P < 0.001, Fig. 1E), thereby repressing the ECM degradation of CHON-001 cells. In contrast to KCNQ1OT1 up-regulation, KCNQ1OT1 down-regulation made opposite effects on the protein expression of collagen II and MMP13 in CHON-001 cells.

Fig. 1
figure 1

LncRNA KCNQ1OT1 promotes cell viability and migration, and suppresses ECM degradation in CHON-001 cells. a Relative expression of KCNQ1OT1 was detected by qRT-PCR in cartilage tissues of PTOA patients and normal cartilage tissues. ***P < 0.001, vs. Control. b Relative expression of KCNQ1OT1 was determined by qRT-PCR in CHON-001 cells. ***P < 0.001, vs. sh-NC. ###P < 0.001, vs. pcDNA3.1-NC. c The cell viability was determined by MTT assay in CHON-001 cells. ***P < 0.001, vs. sh-NC. ###P < 0.001, vs. pcDNA3.1-NC. d Relative migration was assessed by transwell assay in CHON-001 cells. **P < 0.01, vs. sh-NC. ##P < 0.01, vs. pcDNA3.1-NC. e Relative protein expression of collagen II and MMP13 was determined by western blot in CHON-001 cells. **P < 0.01, ***P < 0.001, vs. sh-NC. ##P < 0.01, ###P < 0.001, vs. pcDNA3.1-NC

LncRNA KCNQ1OT1 can directly bind to miR-126-5p

For exploring the downstream mechanism by which KCNQ1OT1 affected CHON-001 cells, we searched for targeting miRNAs of KCNQ1OT1 by starbase. Interestingly, we observed some complementary bases between miR-126-5p and KCNQ1OT1 (Fig. 2A). Meantime, DLR assay was used to verify the interaction of miR-126-5p and KCNQ1OT1. As shown in Fig. 2B, addition of miR-126-5p mimics markedly decreased relative luciferase activity of KCNQ1OT1 wt compared to addition of mimics NC (P < 0.01), while it failed to affect relative luciferase activity of KCNQ1OT1 mut in CHON-001 cells, confirming the direct binding between miR-126-5p and KCNQ1OT1 (Fig. 2B). Moreover, we discovered that relative expression of miR-126-5p was greatly boosted by knockdown of KCNQ1OT1 but reduced by overexpression of KCNQ1OT1 in CHON-001 cells (all P < 0.001, Fig. 2C). MiR-126-5p expression was higher in the PTOA group relative to that in the control group (P < 0.001, Fig. 2D). There was an inverse correlation between KCNQ1OT1 expression and miR-126-5p expression in cartilage tissues of PTOA patients (P = 0.0171, R2 = 0.2422, Fig. 2E).

Fig. 2
figure 2

LncRNA KCNQ1OT1 can directly bind to miR-126-5p. a The binding sequences between KCNQ1OT1 and miR-126-5p were predicted by starbase. b The interaction between KCNQ1OT1 and miR-126-5p in CHON-001 cells was verified by dual-luciferase reporter (DLR) assay. **P < 0.01, vs. mimics NC. c Relative expression of miR-126-5p was detected by qRT-PCR in CHON-001 cells. ***P < 0.001, vs. sh-NC. ###P < 0.001, vs. pcDNA3.1-NC. d Relative expression of miR-126-5p was detected by qRT-PCR in cartilage tissues of PTOA patients and normal cartilage tissues. ***P < 0.001, vs. Control. e The correlation between KCNQ1OT1 and miR-126-5p was analyzed by Pearson’s correlation analysis in cartilage tissues of PTOA patients. P = 0.0171. R2 = 0.2422

Decreased expression of miR-126-5p enhances the viability and migratory ability but represses ECM degradation in CHON-001 cells

Afterwards, to examine whether miR-126-5p made impacts on CHON-001 cells, CHON-001 cells were transfected with miR-126-5p mimics, miR-126-5p inhibitor and corresponding controls. As manifested in Fig. 3A, miR-126-5p mimics caused notable up-regulation of miR-126-5p, and miR-126-5p inhibitor led to distinct down-regulation of miR-126-5p in CHON-001 cells (all P < 0.001). Functionally, it was found that the cell viability (P < 0.001, Fig. 3B) and migratory ability (P < 0.01, Fig. 3C) were enhanced by miR-126-5p inhibitor, and reduced by miR-126-5p mimics in CHON-001 cells. Relative protein expression of collagen II was decreased by miR-126-5p mimics but elevated by miR-126-5p inhibitor in CHON-001 cells (all P < 0.001, Fig. 3D). Meantime, relative protein expression of MMP13 was boosted by miR-126-5p mimics (P < 0. 01) but reduced by miR-126-5p inhibitor (P < 0. 001) in CHON-001 cells (Fig. 3D).

Fig. 3
figure 3

Decreased expression of miR-126-5p enhances the viability and migratory ability of chondrocytes, and represses ECM degradation in CHON-001 cells. a Relative expression of miR-126-5p was determined by qRT-PCR in CHON-001 cells. ***P < 0.001, vs. mimics NC. ###P < 0.001, vs. inhibitor NC. b The cell viability was determined by MTT assay in CHON-001 cells. ***P < 0.001, vs. mimics NC. ###P < 0.001, vs. inhibitor NC. c Relative migration was assessed by transwell assay in CHON-001 cells. **P < 0.01, vs. mimics NC. ##P < 0.01, vs. inhibitor NC. d Relative protein expression of collagen II and MMP13 was determined by western blot in CHON-001 cells. **P < 0.01, ***P < 0.001, vs. mimics NC. ###P < 0.001, vs. inhibitor NC

TRPS1 was targeted by miR-126-5p

After that, target genes of miR-126-5p were predicted with starbase. It was uncovered that TRPS1 possessed binding regions of miR-126-5p (Fig. 4A). Meanwhile, the result of DLR displayed that relative luciferase activity of CHON-001 cells was evidently reduced after co-transfection of miR-126-5p mimics and TRPS1 wt (P < 0.01), while showed no significant change after co-transfection of miR-126-5p mimics and TRPS1 mut (Fig. 4B). Besides, we found that the protein expression of TRPS1 was decreased by miR-126-5p up-regulation and increased by miR-126-5p down-regulation in CHON-001 cells (all P < 0.001, Fig. 4C). The mRNA expression of TRPS1 was declined in the PTOA group compared to the control group (P < 0.001, Fig. 4D). TRPS1 was inversely correlated with miR-126-5p (P = 0.0037, R2 = 0.336, Fig. 4E) and positively correlated with KCNQ1OT1 (P < 0.0001, R2 = 0.6136, Fig. 4F) in cartilage tissues of PTOA patients.

Fig. 4
figure 4

TRPS1 was targeted by miR-126-5p. a The binding sequences between TRPS1 and miR-126-5p were predicted by starbase. b The interaction between TRPS1 and miR-126-5p in CHON-001 cells was verified by dual-luciferase reporter (DLR) assay. **P < 0.01, vs. mimics NC. c Relative protein expression of TRPS1 was detected by western blot in CHON-001 cells. ***P < 0.001, vs. mimics NC. ###P < 0.001, vs. inhibitor NC. d Relative expression of TRPS1 was detected by qRT-PCR in cartilage tissues of PTOA patients and normal cartilage tissues. ***P < 0.001, vs. Control. e The correlation between TRPS1 and miR-126-5p was analyzed by Pearson’s correlation analysis in cartilage tissues of PTOA patients. P = 0.0037. R2 = 0.336. f The correlation between KCNQ1OT1 and TRPS1 was analyzed by Pearson’s correlation analysis in cartilage tissues of PTOA patients. P < 0.0001. R2 = 0.6136

LncRNA KCNQ1OT1 enhances the viability and migration but suppresses degradation of CHON-001 cells via sponging miR-126-5p to target TRPS1

Finally, we explored the interrelation among KCNQ1OT1, miR-126-5p and TRPS1 in CHON-001 cells. Firstly, TRPS1 knockdown was achieved by sh-TRPS1 transfection. As expected, the mRNA expression of TRPS1 was remarkably reduced by sh-TRPS1 transfection in CHON-001 cells (P < 0.01, Fig. 5A). Then, the regulatory relation of KCNQ1OT1, miR-126-5p and TRPS1 was tested. We found that TRPS1 expression was elevated by pcDNA3.1-KCNQ1OT1, and the elevation effect of pcDNA3.1-KCNQ1OT1 on TRPS1 expression was reversed by miR-126-5p mimics in CHON-001 cells (all P < 0.001, Fig. 5B). Next, rescue assays were implemented. It was proved that KCNQ1OT1 overexpression strengthened the viability and migratory ability of CHON-001 cells (all P < 0.001, Fig. 5C-D), and these enhanced effects were partially attenuated by miR-126-5p overexpression or TRPS1 knockdown in CHON-001 cells (all P < 0.01, Fig. 5C-D). Additionally, KCNQ1OT1 overexpression elevated the protein expression of collagen II but reduced the protein expression of MMP13 in CHON-001 cells (all P < 0.001, Fig. 5E), which was reversed by miR-126-5p overexpression or TRPS1 knockdown (all P < 0.01, Fig. 5E).

Fig. 5
figure 5

LncRNA KCNQ1OT1 enhances the cell viability and migration but suppresses ECM degradation via sponging miR-126-5p to target TRPS1 in CHON-001 cells. a Relative expression of TRPS1 in CHON-001 cells was detected by qRT-PCR after transfection of sh-NC and sh-TRPS1. **P < 0.01, vs. sh-NC. b Relative expression of TRPS1 was determined by qRT-PCR in CHON-001 cells. ***P < 0.001, vs. pcDNA3.1-NC. ###P < 0.001, vs. pcDNA3.1-KCNQ1OT1. c The cell viability was determined by MTT assay in CHON-001 cells. ***P < 0.001, vs. pcDNA3.1-NC. ##P < 0.01, vs. pcDNA3.1-KCNQ1OT1. d Relative migration was assessed by transwell assay in CHON-001 cells. ***P < 0.001, vs. pcDNA3.1-NC. ##P < 0.01, vs. pcDNA3.1-KCNQ1OT1. e Relative protein expression of collagen II and MMP13 was determined by western blot in CHON-001 cells. ***P < 0.001, vs. pcDNA3.1-NC. ##P < 0.01, vs. pcDNA3.1-KCNQ1OT1

Discussion

OA is the most common joint disease with high morbidity in the middle-aged and elderly population [25, 26]. Numerous lncRNAs have been confirmed to be downregulated in OA [27,28,29], such as lncRNA ATB [27], lncRNA PACER [28], and lncRNA ANCR [29]. In line with the previous studies, a decreased expression of lncRNA KCNQ1OT1 was also observed in cartilage tissues of PTOA patients compared to that of subjects without PTOA. The results implied that lncRNA KCNQ1OT1 may participate in the progression of OA. Given that loss of chondrocytes is the main cause of OA [30], numerous reports have focused on the impacts of lncRNAs on chondrocytes [8, 9, 31]. For instance, knockdown of lncRNA-p21 strengths cell viability and repress apoptosis in OA chondrocytes [31]. LncRNA MALAT1 promotes cell proliferation and inhibit ECM degradation in IL-1β-induced chondrocytes [8]. Silencing of lncRNA MFI2-AS1 elevates chondrocytes viability but represses ECM degradation [9]. In this study, we discovered that KCNQ1OT1 overexpression promoted the viability and migration as well as inhibition of ECM degradation in CHON-001 cells. Similarly, a recent study conducted by Gu et al. has indicated that KCNQ1OT1 facilitates the proliferation and suppress apoptosis of articular chondrocytes, thus accelerating fracture healing [12]. Therefore, we speculated that KCNQ1OT1 may be a favourable factor to prevent the development of OA due to its important role in promoting the viability and migration of chondrocytes.

Emerging evidence has shown that miR-162 is implicated in OA [32]. Yu et al. have indicated that miR-126 is highly expressed in OA tissues, whereas decreased miR-126 strengthens chondrocyte viability and migration [18]. Interestingly, Ai et al. also found an increased miR-126 in CHON-001 cells, and the cell proliferation can be promoted by suppression of miR-126 [19]. Similarly, we revealed that miR-126-5p was up-regulated in cartilage tissues of PTOA patients, suggesting that miR-126-5p may be involved in the development of OA. Then, our functional experiments on miR-126-5p demonstrated that inhibition of miR-126-5p enhanced the viability and migratory ability of CHON-001 cells, and repressed ECM degradation in CHON-001 cells. Based on these results, we speculated that miR-126-5p may be a pathogenic miRNA existed in OA progression. On the other hand, miR-126-5p is revealed to act as the downstream targets of varied lncRNAs, such as lncRNA CRNDE [33], lncRNA HOTAIR [34], and lncRNA TMPO-AS1 [35]. Here, miR-126-5p was also identified as the target of and negatively regulated by lncRNA KCNQ1OT1. Therefore, we speculated that KCNQ1OT1 may promote the viability and migration of CHON-001 cells by targeting miR-126-5p. The rescue experiments confirmed that miR-126-5p mimics reversed the promoting effects of KCNQ1OT1 on cell viability and migration as well as the suppressive effect of KCNQ1OT1 on ECM degradation in CHON-001 cells. These results verified our speculation.

Numerous studies have uncovered the vital role of TRPS1 in cellular processes of chondrocytes [24, 36]. Gai et al. have stated that TRPS1 can regulate the proliferation and apoptosis of chondrocytes [36]. Suemoto et al. have unveiled that overexpression of TRPS1 facilitates chondrocytes proliferation by reduction the level of Stat3 [24]. In the present study, we found that TRPS1 expression was reduced in cartilage tissues, suggesting that TRPS1 may exhibited an anti-osteoarthritis OA progression. It is noticed that TRPS1, an atypical member of the GATA transcriptional factor family, is initially identified as a transcriptional repressor [37]. Indeed, it was previously uncovered to be targeted by varied miRNAs, including miR-26b-5p [38], and miR-221 [39]. Additionally, convincing evidence has demonstrated that complex interactions between miRNAs and their target genes are essential in regulating of homeostatic pathways in OA [40]. Inspired by this perception, bioinformatics analysis was implemented to predict target genes of miR-126-5p. We found that TRPS1 was a direct target gene of miR-126-5p. Meanwhile, we observed the negative correlation between TRPS1 and miR-126-5p in cartilage tissues of PTOA patients. Taken together, we speculated that decreased miR-126-5p enhanced the viability and migratory ability as well as repressing ECM degradation by targeting TRPS1 in CHON-001 cells. More importantly, we discovered that TRPS1 was positively correlated with KCNQ1OT1 in cartilage tissues of PTOA patients. The promoting effects of KCNQ1OT1 on cell viability and migration as well as the inhibitory effect of KCNQ1OT1 on ECM degradation were reversed by TRPS1 knockdown in CHON-001 cells. Therefore, we believed that lncRNA KCNQ1OT1 could repress OA progression via mediating miR-126-5p/TRPS1 axis in vitro. In addition, there may be some other important genes or pathways related to this axis in several types of human diseases, such as TRPS1-SOX9 in hereditary hypertrichoses or chondrocytes differentiation [41], KCNQ1OT1-PI3K/AKT pathway in cardiomyocytes injury [42] or colorectal cancer [43], miR-126-5p-PI3K/AKT pathway in ovarian cancer [44], cervical cancer [45], and gastric cancer [46]. We speculated that these important genes or pathways may be also involved in regulation of OA pathogenesis via interaction of KCNQ1OT1/miR-126-5p/TRPS1 axis. This is a research direction in our future studies.

Furthermore, there are also some other limitations in this study: i) lncRNA KCNQ1OT1 may be related with paternal allele, and the differences on KCNQ1OT1 expression in female and male OA patients are needed to be studied; ii), CHON-001 cells combined with cytokine stimulation and osmotic pressure to establish a vitro model for OA or use of the isolated chondrocytes from OA patients may be more rigorous and significant; iii) this study is focused on a cellular level, and in vivo experiments should be performed.

Conclusions

In summary, lncRNA KCNQ1OT1 was decreased in the PTOA group compared to the control group. LncRNA KCNQ1OT1 acted as a sponge of miR-126-5p, and TRPS1 was the downstream target of miR-126-5p. Overall, lncRNA KCNQ1OT1 repressed OA progression via binding to miR-126-5p and indirectly regulating TRPS1 in vitro. Hence, our study provided insights into the development of a novel target for treating OA.

Availability of data and materials

All data generated or analysed during this study are included in this published article.

Abbreviations

OA:

Osteoarthritis

LncRNAs:

Long non-coding RNA

QRT-PCR:

Quantitative real-time polymerase chain reaction

DLR:

Dual-luciferase reporter

ECM:

Extracellular matrix

KCNQ1OT1:

LncRNA KCNQ1 overlapping transcript 1

MiRNAs:

microRNAs

TRPS1:

Tricho-rhino-phalangeal syndrome type I

PVDF:

Polyvinylidene difluoride

TBST:

Tris buffered saline Tween

References

  1. Pereira D, Ramos E, Branco J. Osteoarthritis. Acta Medica Port. 2015;28(1):99–106. https://doi.org/10.20344/amp.5477.

    Article  Google Scholar 

  2. Liu C, Wang B, Xiao L, Li Y, Xu L, Zhao Z, et al. Protective effects of the pericellular matrix of chondrocyte on articular cartilage against the development of osteoarthritis. Histol Histopathol. 2018;33(8):757–64. https://doi.org/10.14670/HH-11-967.

    Article  CAS  PubMed  Google Scholar 

  3. Suri P, Morgenroth DC, Hunter DJ. Epidemiology of osteoarthritis and associated comorbidities. PM R. 2012;4(5 Suppl):S10–9. https://doi.org/10.1016/j.pmrj.2012.01.007.

    Article  PubMed  Google Scholar 

  4. Wollheim FA. Current pharmacological treatment of osteoarthritis. Drugs. 1996;52(Suppl 3):27–38. https://doi.org/10.2165/00003495-199600523-00006.

    Article  CAS  PubMed  Google Scholar 

  5. Richmond J, Hunter D, Irrgang J, Jones MH, Snyder-Mackler L, van Durme D, et al. American Academy of Orthopaedic surgeons clinical practice guideline on the treatment of osteoarthritis (OA) of the knee. J Bone Joint Surg Am. 2010;92(4):990–3. https://doi.org/10.2106/JBJS.I.00982.

    Article  PubMed  Google Scholar 

  6. Brosseau L, Wells G, Marchand S, Gaboury I, Stokes B, Morin M, et al. Randomized controlled trial on low level laser therapy (LLLT) in the treatment of osteoarthritis (OA) of the hand. Lasers Surg Med. 2005;36(3):210–9. https://doi.org/10.1002/lsm.20137.

    Article  PubMed  Google Scholar 

  7. Hermann W, Lambova S, Muller-Ladner U. Current treatment options for osteoarthritis. Curr Rheumatol Rev. 2018;14(2):108–16. https://doi.org/10.2174/1573397113666170829155149.

    Article  CAS  PubMed  Google Scholar 

  8. Zhang Y, Wang F, Chen G, He R, Yang L. LncRNA MALAT1 promotes osteoarthritis by modulating miR-150-5p/AKT3 axis. Cell Biosci. 2019;9(1):54. https://doi.org/10.1186/s13578-019-0302-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Luo X, Wang J, Wei X, Wang S, Wang A. Knockdown of lncRNA MFI2-AS1 inhibits lipopolysaccharide-induced osteoarthritis progression by miR-130a-3p/TCF4. Life Sci. 2020;240:117019. https://doi.org/10.1016/j.lfs.2019.117019.

    Article  CAS  PubMed  Google Scholar 

  10. Tang LP, Ding JB, Liu ZH, Zhou GJ. LncRNA TUG1 promotes osteoarthritis-induced degradation of chondrocyte extracellular matrix via miR-195/MMP-13 axis. Eur Rev Med Pharmacol Sci. 2018;22(24):8574–81. https://doi.org/10.26355/eurrev_201812_16620.

    Article  PubMed  Google Scholar 

  11. Kanduri C. Kcnq1ot1: a chromatin regulatory RNA. Semin Cell Dev Biol. 2011;22(4):343–50. https://doi.org/10.1016/j.semcdb.2011.02.020.

    Article  CAS  PubMed  Google Scholar 

  12. Gu H, Li Z, Lv XF, Zhao AB, Zhu MY, Zhang Y. LncRNA KCNQ1OT1 delayed fracture healing through the Wnt/beta-catenin pathway. Eur Rev Med Pharmacol Sci. 2019;23(11):4575–83. https://doi.org/10.26355/eurrev_201906_18034.

    Article  CAS  PubMed  Google Scholar 

  13. Gao X, Ge J, Li W, Zhou W, Xu L. LncRNA KCNQ1OT1 promotes osteogenic differentiation to relieve osteolysis via Wnt/beta-catenin activation. Cell Biosci. 2018;8(1):19. https://doi.org/10.1186/s13578-018-0216-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Paraskevopoulou MD, Hatzigeorgiou AG. Analyzing MiRNA-LncRNA interactions. Methods Mol Biol. 2016;1402:271–86. https://doi.org/10.1007/978-1-4939-3378-5_21.

    Article  CAS  PubMed  Google Scholar 

  15. Swingler TE, Niu L, Smith P, et al. The function of microRNAs in cartilage and osteoarthritis. Clin Exp Rheumatol. 2019;37(Suppl 120(5)):40–7.

    PubMed  Google Scholar 

  16. Zhong G, Long H, Ma S, et al. miRNA-335-5p relieves chondrocyte inflammation by activating autophagy in osteoarthritis. Life Sci. 2019;226:164–72. https://doi.org/10.1016/j.lfs.2019.03.071.

    Article  CAS  PubMed  Google Scholar 

  17. Si HB, Zeng Y, Liu SY, Zhou ZK, Chen YN, Cheng JQ, et al. Intra-articular injection of microRNA-140 (miRNA-140) alleviates osteoarthritis (OA) progression by modulating extracellular matrix (ECM) homeostasis in rats. Osteoarthr Cartil. 2017;25(10):1698–707. https://doi.org/10.1016/j.joca.2017.06.002.

    Article  Google Scholar 

  18. Yu CD, Miao WH, Zhang YY, Zou MJ, Yan XF. Inhibition of miR-126 protects chondrocytes from IL-1beta induced inflammation via upregulation of Bcl-2. Bone Joint Res. 2018;7(6):414–21. https://doi.org/10.1302/2046-3758.76.BJR-2017-0138.R1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ai D, Yu F. LncRNA DNM3OS promotes proliferation and inhibits apoptosis through modulating IGF1 expression by sponging MiR-126 in CHON-001 cells. Diagn Pathol. 2019;14(1):106. https://doi.org/10.1186/s13000-019-0877-2.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Wang H, Huang Y, Yang Y. LncRNA PVT1 regulates TRPS1 expression in breast Cancer by sponging miR-543. Cancer Manag Res. 2020;12:7993–8004. https://doi.org/10.2147/CMAR.S263383.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhang J, Ma X, Zhou R, Zhou Y. TRPS1 and YAP1 regulate cell proliferation and drug resistance of osteosarcoma via competitively binding to the target of circTADA2A - miR-129-5p. Onco Targets Ther. 2020;13:12397–407. https://doi.org/10.2147/OTT.S276953.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hong J, Sun J, Huang T. Increased expression of TRPS1 affects tumor progression and correlates with patients' prognosis of colon cancer. Biomed Res Int. 2013;2013:454085.

    PubMed  PubMed Central  Google Scholar 

  23. Wuelling M, Schneider S, Schrother VA, et al. Wnt5a is a transcriptional target of Gli3 and Trps1 at the onset of chondrocyte hypertrophy. Dev Biol. 2020;457(1):104–18. https://doi.org/10.1016/j.ydbio.2019.09.012.

    Article  CAS  PubMed  Google Scholar 

  24. Suemoto H, Muragaki Y, Nishioka K, Sato M, Ooshima A, Itoh S, et al. Trps1 regulates proliferation and apoptosis of chondrocytes through Stat3 signaling. Dev Biol. 2007;312(2):572–81. https://doi.org/10.1016/j.ydbio.2007.10.001.

    Article  CAS  PubMed  Google Scholar 

  25. Mandl LA. Osteoarthritis year in review 2018: clinical. Osteoarthr Cartil. 2019;27(3):359–64. https://doi.org/10.1016/j.joca.2018.11.001.

    Article  CAS  Google Scholar 

  26. Guan VX, Mobasheri A, Probst YC. A systematic review of osteoarthritis prevention and management with dietary phytochemicals from foods. Maturitas. 2019;122:35–43. https://doi.org/10.1016/j.maturitas.2019.01.005.

    Article  CAS  PubMed  Google Scholar 

  27. Dang X, Lian L, Wu D. The diagnostic value and pathogenetic role of lncRNA-ATB in patients with osteoarthritis. Cell Mol Biol Lett. 2018;23(1):55. https://doi.org/10.1186/s11658-018-0118-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Jiang M, Liu J, Luo T, et al. LncRNA PACER is down-regulated in osteoarthritis and regulates chondrocyte apoptosis and lncRNA HOTAIR expression. Biosci Rep. 2019;39(6):BSR20190404. https://doi.org/10.1042/BSR20190404.

  29. Li Q, Zhang Z, Guo S, Tang G, Lu W, Qi X. LncRNA ANCR is positively correlated with transforming growth factor-beta1 in patients with osteoarthritis. J Cell Biochem. 2019;120(9):14226–32. https://doi.org/10.1002/jcb.28881.

    Article  CAS  PubMed  Google Scholar 

  30. Hosseinzadeh A, Kamrava SK, Joghataei MT, Darabi R, Shakeri-Zadeh A, Shahriari M, et al. Apoptosis signaling pathways in osteoarthritis and possible protective role of melatonin. J Pineal Res. 2016;61(4):411–25. https://doi.org/10.1111/jpi.12362.

    Article  CAS  PubMed  Google Scholar 

  31. Tang L, Ding J, Zhou G, Liu Z. LncRNAp21 promotes chondrocyte apoptosis in osteoarthritis by acting as a sponge for miR451. Mol Med Rep. 2018;18(6):5295–301. https://doi.org/10.3892/mmr.2018.9506.

    Article  CAS  PubMed  Google Scholar 

  32. Fang Y, Shen C, Zhang H, et al. Effect of MiR-126 on cartilage regeneration in osteoarthritis rabbits through MAPK signaling pathway. Minerva Med. 2019.

  33. Liu C, Hou J, Shan F, Wang L, Lu H, Ren T. Long non-coding RNA CRNDE promotes colorectal carcinoma cell progression and paclitaxel resistance by regulating miR-126-5p/ATAD2 Axis. Onco Targets Ther. 2020;13:4931–42. https://doi.org/10.2147/OTT.S237580.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Liu L, Cui S, Wan T, Li X, Tian W, Zhang R, et al. Long non-coding RNA HOTAIR acts as a competing endogenous RNA to promote glioma progression by sponging miR-126-5p. J Cell Physiol. 2018;233(9):6822–31. https://doi.org/10.1002/jcp.26432.

    Article  CAS  PubMed  Google Scholar 

  35. Hu Y, Zhang Y, Ding M, et al. LncRNA TMPO-AS1/miR-126-5p/BRCC3 axis accelerates gastric cancer progression and angiogenesis via activating PI3K/Akt/mTOR pathway. J Gastroenterol Hepatol. 2020. Online ahead of print.

  36. Gai Z, Gui T, Muragaki Y. The function of TRPS1 in the development and differentiation of bone, kidney, and hair follicles. Histol Histopathol. 2011;26(7):915–21. https://doi.org/10.14670/HH-26.915.

    Article  CAS  PubMed  Google Scholar 

  37. Momeni P, Glockner G, Schmidt O, et al. Mutations in a new gene, encoding a zinc-finger protein, cause tricho-rhino-phalangeal syndrome type I. Nat Genet. 2000;24(1):71–4. https://doi.org/10.1038/71717.

    Article  CAS  PubMed  Google Scholar 

  38. Wilke CM, Hess J, Klymenko SV, Chumak VV, Zakhartseva LM, Bakhanova EV, et al. Expression of miRNA-26b-5p and its target TRPS1 is associated with radiation exposure in post-Chernobyl breast cancer. Int J Cancer. 2018;142(3):573–83. https://doi.org/10.1002/ijc.31072.

    Article  CAS  PubMed  Google Scholar 

  39. Penolazzi L, Lambertini E, Scussel Bergamin L, et al. Reciprocal Regulation of TRPS1 and miR-221 in Intervertebral Disc Cells. Cells. 2019;8(10):1170. https://doi.org/10.3390/cells8101170.

  40. Oliviero A, Della Porta G, Peretti GM, Maffulli N. MicroRNA in osteoarthritis: physiopathology, diagnosis and therapeutic challenge. Br Med Bull. 2019;130(1):137–47. https://doi.org/10.1093/bmb/ldz015.

    Article  CAS  PubMed  Google Scholar 

  41. Tan Z, Niu B, Tsang KY, Melhado IG, Ohba S, He X, et al. Synergistic co-regulation and competition by a SOX9-GLI-FOXA phasic transcriptional network coordinate chondrocyte differentiation transitions. PLoS Genet. 2018;14(4):e1007346. https://doi.org/10.1371/journal.pgen.1007346.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pan F, Xu X, Zhan Z, et al. 6-Gingerol protects cardiomyocytes against hypoxia-induced injury by regulating the KCNQ1OT1/miR-340-5p/ PI3K/AKT pathway. Panminerva Med. 2020. Online ahead of print.

  43. Duan Q, Cai L, Zheng K, Cui C, Huang R, Zheng Z, et al. lncRNA KCNQ1OT1 knockdown inhibits colorectal cancer cell proliferation, migration and invasiveness via the PI3K/AKT pathway. Oncol Lett. 2020;20(1):601–10. https://doi.org/10.3892/ol.2020.11619.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Bi X, Lv X, Liu D, et al. METTL3-mediated maturation of miR-126-5p promotes ovarian cancer progression via PTEN-mediated PI3K/Akt/mTOR pathway. Cancer Gene Ther. 2020;28(3-4):335–49.

  45. Shi WJ, Liu H, Ge YF, Wu D, Tan YJ, Shen YC, et al. LINC00673 exerts oncogenic function in cervical cancer by negatively regulating miR-126-5p expression and activates PTEN/PI3K/AKT signaling pathway. Cytokine. 2020;136:155286. https://doi.org/10.1016/j.cyto.2020.155286.

    Article  CAS  PubMed  Google Scholar 

  46. Hu Y, Zhang Y, Ding M, et al. Long noncoding RNA TMPO-AS1/miR-126-5p/BRCC3 axis accelerates gastric cancer progression and angiogenesis via activating PI3K/Akt/mTOR pathway. J Gastroenterol Hepatol. 2020. Online ahead of print.

Download references

Acknowledgements

Not applicable.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

All authors made substantial contributions to conception and design, acquisition of data, or analysis and interpretation of data; took part in drafting the article or revising it critically for important intellectual content; gave final approval of the version to be published; and agree to be accountable for all aspects of the work.

Corresponding author

Correspondence to Xiangwei Liu.

Ethics declarations

Ethics approval and consent to participate

The current study was approved by the ethics committees of Chifeng Municipal Hospital, and all participants had signed written informed consents.

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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, B., Liu, X. Long non-coding RNA KCNQ1OT1 promotes cell viability and migration as well as inhibiting degradation of CHON-001 cells by regulating miR-126-5p/TRPS1 axis. Adv Rheumatol 61, 31 (2021). https://doi.org/10.1186/s42358-021-00187-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s42358-021-00187-3

Keywords