Open Access

Knockdown of lncRNA JPX suppresses IL‑1β‑stimulated injury in chondrocytes through modulating an miR‑25‑3p/PPID axis

  • Authors:
    • Zhiyong Ren
    • Liguo Tang
    • Zhonghua Ding
    • Jun Song
    • Hailiang Zheng
    • Dongzhu Li
  • View Affiliations

  • Published online on: September 19, 2022     https://doi.org/10.3892/ol.2022.13508
  • Article Number: 388
  • Copyright: © Ren et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of this study was to investigate the potential mechanisms of long noncoding (lnc) RNA Just proximal to X‑inactive specific transcript (JPX) in interleukin (IL)‑1β‑stimulated chondrocytes. Human C28/I2 chondrocytes were treated with IL‑1β to simulate osteoarthritic (OA) injury. The expression levels of JPX, microRNA (miRNA/miR)‑25‑3p, and peptidylprolyl isomerase D (PPID) were measured using reverse transcription‑quantitative PCR or western blotting. The IL‑1β‑stimulated injury was assessed using a Cell Counting Kit‑8 assay, flow cytometry, and western blot analysis. The targeted relationship between miR‑25‑3p, JPX, and PPID was verified using a dual‑luciferase reporter and RNA immunoprecipitation (RIP) assays. The results showed that JPX expression was upregulated in OA patients and IL‑1β‑stimulated chondrocytes. JPX knockdown enhanced cell viability and suppressed apoptosis of IL‑1β‑stimulated chondrocytes. miR‑25‑3p inhibition rescued the inhibitory effect of JPX knockdown on IL‑1β‑stimulated injury. PPID overexpression eliminated the effects of JPX knockdown on IL‑1β‑stimulated chondrocytes. In conclusion, JPX knockdown increased cell viability and reduced apoptosis in IL‑1β‑stimulated chondrocytes, and this involved modulation of a miR‑25‑3p/PPID axis.

Introduction

Osteoarthritis (OA) is a complex chronic disease characterized by stiffness, arthralgia, and swelling, which is the primary cause of disability amongst the elderly (1,2). Chondrocytes are a type of cell in cartilage tissues that primarily function to generate and maintain the extracellular matrix (ECM) components (3). During OA, chondrocytes undergo multiple changes, such as in their secretory profiles and regarding their cell viability (4). Aberrant apoptosis and inflammatory responses in chondrocytes are related to cartilage degradation in OA (5,6). Therefore, probing the mechanism of chondrocyte dysfunction may be useful in understanding OA pathogenesis.

Long noncoding RNAs (lncRNAs) exert important roles in a variety of biological processes, including metabolism, immunity, differentiation, and apoptosis (7,8). A growing number of studies have shown that lncRNAs play vital effects in osteogenesis, chondrogenesis, and OA (9,10). More recently, the dysregulation of lncRNAs has been studied in OA (11,12). LncRNA ZNFX1 antisense RNA 1 was shown to inhibit apoptosis and matrix synthesis, and facilitate chondrocyte growth and migration in OA (13). In human chondrocytes, lncRNA urothelial cancer associated 1 increased matrix metallopeptidase 13 expression by suppressing microRNA (miRNA/miR)-204-5p (14). Reports on Just proximal to X-inactive specific transcript (JPX), a lncRNA, have primarily focused on its regulatory effects in various cancers, including hepatocellular carcinoma, colorectal cancer, lung cancer, oral squamous cell carcinoma, and myeloid malignancies (1519). Interestingly, Gál et al (20) found that JPX expression was upregulated in adult patients with allergic rhinitis compared with the adult control group. Chen et al (21) showed that JPX was upregulated in allergic rhinitis and knockdown of JPX improved the imbalance between Treg/Th17 observed in allergic rhinitis. Nevertheless, whether JPX participates in OA development and the underlying molecular mechanisms involved remain to be determined.

An increasing number of studies have demonstrated that miRNAs are involved in the regulation of cell viability, differentiation, inflammation, lipid metabolism, apoptosis, oncogenesis, and other core cellular activities (22). It has been shown that miRNAs may serve as novel therapeutic targets for OA (23). LncRNAs can act as competitive endogenous (ce)RNAs by functioning as miRNA sponges, leading to the suppression of miRNAs (24,25). For example, lncRNA plasmacytoma variant translocation 1 aggravated ECM degradation by inhibiting miR-140 expression in OA (26). LINC00461 enhanced chondrocyte proliferation and cell cycle progression by downregulating miR-30a-5p expression in OA (27). LncRNA HOX transcript antisense RNA (HOTAIR) knockdown promoted proliferation and inhibited ECM degradation of OA chondrocytes by increasing the activity of the miR-107/C-X-C motif chemokine ligand 12 axis (28). Kurowska et al (29) found that miR-25-3p expression was downregulated in patients with rheumatoid arthritis. Wang et al (30) found that miR-25-3p expression was downregulated in osteoarthritic cartilage compared with healthy cartilage. Moreover, miR-25-3p expression was decreased in TNF-α-induced rat chondrocytes, and miR-25-3p negatively regulated IGFBP7 to promote chondrocyte proliferation and reduce chondrocyte apoptosis in OA (31). However, whether lncRNA JPX regulates miR-25-3p in OA remains to be assessed.

The first peptidyl cis-trans prolyl isomerase (PPIase) was isolated in 1984 by Fischer et al (32). The inhibition of PPIase activity protects cells from apoptosis (33). Lebedev et al (34) reported that PPIF was involved in mitochondrial permeability transition. PPIF-mediated necrosis participates in the pathological process of myocardial and/or cerebral ischemia/reperfusion injury (35). Inhibition of PPIF increases cell viability of TNF-α treated osteoblast-like cells (36). However, as a member of the PPIase family, the potential regulatory role of peptidylprolyl isomerase D (PPID) has not been elucidated.

In the present study, we investigated the potential effects of lncRNA JPX in OA. The function of JPX in cell viability and apoptosis of chondrocytes, and the specific mechanism of modulating miR-25-3p activity were assessed.

Materials and methods

Specimen collection

A total of 20 OA patients and 16 non-OA patients (patients without symptoms of OA during total hip replacement) from Sunshine Union Hospital (Weifang, China) were collected in the present study. Patients provided signed consent to the collection of samples and the present study was approved by the Ethics Committee of Sunshine Union Hospital.

Cell culture and treatment

Human C28/I2 chondrocytes were obtained from BeNa Culture Collection. C28/I2 cells were cultured in DMEM/F12 (Thermo Fisher Scientific, Inc.) supplemented with 10% FBS and 1% penicillin-streptomycin solution (Thermo Fisher Scientific, Inc.) and maintained in a humidified incubator supplied with 5% CO2 at 37°C. To generate a model of OA, C28/I2 cells were treated with IL-1β recombinant protein (5, 10, and 20 ng/ml) for 24 h (37). C28/I2 cells without IL-1β treatment were used as the control.

Cell transfection

Small interfering (si)RNAs targeting JPX (50 nM; si-JPX#1, 5′-GTTGCAAGGCGTCCGAAGTAT-3′ and si-JPX#2, 5′-GTCCGAAGTATGAGTCCACTA-3′), and negative control (50 nM; si-NC, 5′-TTCTCCGAACGTGTCACGT-3′), miR-25-3p mimic (40 nM; sense, 5′-CAUUGCACUUGUCUCGGUCUGA-3′ and antisense, 5′-AGACCGAGACAAGUGCAAUGUU-3′) and mimic negative control (40 nM; mimic NC, sense 5′-UUCUCCGAACGUGUCACGUTT-3′ and antisense 5′-ACGUGACACGUUCGGAGAATT-3′), miR-25-3p inhibitor (40 nM; 5′-UCAGACCGAGACAAGUGCAAUG-3′) and inhibitor negative control (40 nM; inhibitor NC, 5′-CAGUACUUUUGUGUAGUACAA-3′) were purchased from Shanghai GenePharma, Co., Ltd. A PPID overexpression vector pcDNA™ 3.1/V5-HisB-PPID (pc-PPID, 75 ng) and an empty pcDNA3.1 vector (pc-NC, 75 ng) were obtained from Thermo Fisher Scientific, Inc. After addition of the transfection reagent [Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.)] and constructs, cells were cultured for 48 h, after which the subsequent experiments were performed.

Reverse transcription-quantitative (RT-q)PCR analysis

Total RNA was isolated from chondrocytes using TRIzol® reagent (Thermo Fisher Scientific, Inc.), and then converted to cDNA using a PrimeScript RT kit (Takara Bio, Inc.) or TaqMan™ MicroRNA Reverse Transcription Kit (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Subsequently, qPCR analysis was performed for miRNAs using a MiScript SYBR-Green PCR kit (Qiagen GmbH) or SYBR Premix Ex Taq™ Kit (Takara Bio, Inc.) on an ABI 7300HT system. The qPCR thermocycling conditions were as follows: Pre-denaturation at 95°C for 2 min; followed by 40 cycles of denaturation at 95°C for 15 sec, and annealing and extension at 60°C for 30 sec. GAPDH was used as the internal control for JPX and PPID, and U6 was used as the internal control for miR-25-3p. The expression levels of miR-25-3p and lncRNA JPX were quantified using the 2−ΔΔCq method (38). The sequences of the primers were: JPX forward, 5′-TTGCAAGGCGTCCGAAGTAT-3′ and reverse, 5′-AGGCGATCAGCGAGAAAGAA-3′; miR-25-3p forward, 5′-ACACTCCAGCTGGGCATTGCACTTGTCTCG-3′ and reverse, 5′-ACACTCCAGCTGGGCATTGCACTTGTCTCG-3′; PPID forward, 5′-GTGAAAAACCTGCTAAATTGTGCG-3′ and forward, 5′-ATCCGCATCCTCAGGGAAATC-3′; U6 forward, 5′-GGAACGATACAGAGAAGATTAGC-3′ and reverse, 5′-TGGAACGCTTCACGAATTTGCG-3′; GAPDH forward, 5′-GTCAACGGATTTGGTCTGTATT-3′ and reverse, 5′-AGTCTTCTGGGTGGCAGTGAT-3′.

Western blot assay

Total protein was extracted from chondrocytes using RIPA lysis buffer containing protease inhibitors. The concentration of proteins was measured using a BCA kit (Beyotime Institute of Biotechnology). Then, 50 µg protein lysate was loaded per lane on a 10% SDS-gel, resolved using SDS-PAGE, and transferred onto a PVDF membrane. After blocking with 5% fat-free milk for 1 h, the PVDF membranes were incubated with the following primary antibodies: Anti-PPID antibody (cat. no. ab3562; 1:1,000; Abcam), anti-Bax antibody (cat. no. ab32503; 1:1,000; Abcam), anti-Bcl-2 (cat. no. ab32124; 1:1,000; Abcam), anti-cleaved (c)-caspase-9 (cat. no. 20750; 1:1,000; Cell Signaling Technology, Inc.) and GAPDH (cat. no. ab9485; 1:1,000; Abcam) at 4°C overnight. Subsequently, the membranes were incubated with goat anti-rabbit IgG H&L (HRP) secondary antibody (cat. no. ab205718; 1:10,000; Abcam) at room temperature for 1 h. The protein bands were visualized using the BeyoECL Plus kit (Beyotime Institute of Biotechnology) and were semi-quantified with ImageJ version 1.52v (National Institutes of Health).

Cell viability assay

Cell viability was analyzed using a Cell Counting Kit (CCK)-8 assay (Beyotime Institute of Biotechnology) according to the manufacturer's protocol. After 48 h of transfection, cells were plated to a 96-well plate and cultured at 37°C, and incubated until they had adhered. Then, the CCK-8 reagent was added, and cells were further incubated for 4 h. Finally, the absorbance was measured using a microplate reader at 450 nm.

Apoptosis analysis

Apoptosis was measured using an Annexin V-FITC Apoptosis Detection Kit (Beyotime Institute of Biotechnology). After 48 h of transfection, cells were stained with Annexin V-FITC and PI for 15 min at 25°C in the dark. Cell apoptosis was scanned using a BD FACSCalibur™ flow cytometer (BD Biosciences) and analyzed using FlowJo 10.0.6 software (FlowJo, LLC).

Dual-luciferase reporter assay

The binding sites between miR-25-3p and JPX or the 3′UTR PPID were predicted using starBase (https://starbase.sysu.edu.cn/). Cells were plated in a 24-well plate. When cell confluence reached 80%, the WT (or Mut) pmirGLO luciferase reporter gene vector of JPX (or PPID 3′UTR) and miR-25-3p mimic (or mimic NC) were co-transfected into cells with Lipofectamine 2000. After 48 h of transfection, the luciferase activity was measured using a dual luciferase assay kit according to the manufacturer's protocol (Promega Corporation) and normalized to Renilla luciferase activity.

RNA immunoprecipitation (RIP) assay

RIP assays were performed to verify the targeted relationship between miR-25-3p, JPX, and PPID using the RIP Kit (Millipore Sigma) according to the manufacturer's protocol. Cells were lysed using the lysis buffer and incubated with magnetic beads pre-coated with Ago2 antibody (cat. no. ab186733; 1:1,000; Abcam) for 6 h at 4°C. IgG (cat. no. SAB5600195; 1:1,000; MilliporeSigma) was used as the control. Subsequently, beads were washed with RNA binding buffer, and the levels of JPX, miR-25-3p, and PPID were detected by RT-qPCR.

starBase database analysis

The starBase database is a widely-used open-source platform for studying ncRNA interactions from CLIP-seq, degradome-seq, and RNA-RNA interactome data (39,40). Here, starBase was used to predict the binding sites between lncRNA JPX, miR-25-3p, and PPID.

Statistical analysis

All data are presented as the mean ± SD. All experiments were performed in triplicate. A one-way ANOVA followed by a Tukey's post hoc test was used to analyze the differences between multiple groups. A Student's t-test was used to analyze the differences between two groups. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of lncRNA JPX on cell viability and apoptosis of IL-1β-treated chondrocytes

The RT-qPCR results showed that JPX was notably upregulated in OA cartilage tissues compared with the NC cartilage tissues (Fig. 1A). JPX expression was significantly increased in chondrocytes after treatment with 10 and 20 ng/ml IL-1β (Fig. 1B). Thus, 10 ng/ml IL-1β was used for the follow-up experiments. To probe the effect of JPX on chondrocytes, JPX siRNAs were transfected into chondrocytes. Transfection of JPX siRNAs markedly decreased JPX expression levels, especially si-JPX#1 (Fig. 1C). JPX knockdown significantly decreased JPX expression in IL-1β-treated chondrocytes (Fig. 1D). Thus, the effect of JPX knockdown on IL-1β-treated chondrocytes was assessed. IL-1β treatment reduced cell viability and increased cell apoptosis of chondrocytes (Fig. 1E-G); JPX knockdown significantly enhanced cell viability and inhibited cell apoptosis of IL-1β-stimulated chondrocytes (Fig. 1E-G). Additionally, the protein expression levels of Bcl-2, Bax, and c-caspase-9 were determined using western blotting. Bcl-2 expression levels in the IL-1β group were significantly decreased whereas the expression levels of Bax and c-caspase-9 were significantly increased (Fig. 1H and I). JPX knockdown increased Bcl-2 expression levels whilst decreasing the expression levels of Bax and c-caspase-9 in the IL-1β-stimulated chondrocytes (Fig. 1H and I).

miR-25-3p is a target of JPX in chondrocytes

starBase analysis predicted that JPX could target miR-25-3p (Fig. 2A). A dual-luciferase reporter assay and RIP assay were used to confirm the targeting relationship between JPX and miR-25-3p. miR-25-3p mimics decreased luciferase activity in the JPX-WT group (Fig. 2B). The results of RIP revealed that JPX and miR-25-3p were significantly enriched using anti-Ago2 (Fig. 2C). JPX knockdown significantly increased the expression levels of miR-25-3p (Fig. 2D). The results of RT-qPCR showed that miR-25-3p expression in OA patients was notably decreased compared with that in healthy individuals (Fig. 2E). The correlation analysis revealed that miR-25-3p levels in OA patients were negatively related with JPX levels (Fig. 2F). Additionally, miR-25-3p expression levels were significantly decreased by IL-1β (Fig. 2G). Together, these results suggested that lncRNA JPX could target miR-25-3p in chondrocytes.

Inhibition of miR-25-3p abrogates the effects of JPX knockdown on IL-1β-stimulated chondrocytes

Next, whether JPX regulated miR-25-3p to affect the activity and apoptosis of IL-1β-stimulated chondrocytes was assessed. miR-25-3p inhibitor notably reversed the JPX knockdown-induced increase in miR-25-3p expression (Fig. 3A). CCK-8 results showed that JPX knockdown increased cell viability of IL-1β-stimulated chondrocytes (Fig. 3B). JPX knockdown notably reduced cell apoptosis in the IL-1β-stimulated chondrocytes (Fig. 3C and D). However, cell viability was decreased and apoptosis was increased in the IL-1β+si-JPX#1+miR-25-3p inhibitor group compared with the IL-1β+si-JPX#1+inhibitor NC group (Fig. 3B-D). Western blotting results showed that JPX knockdown significantly increased Bcl-2 levels and decreased the levels of Bax and c-caspase-9 (Fig. 3E and F); the effect of JPX knockdown was abrogated by the miR-25-3p inhibitor (Fig. 3E and F). Together, these findings demonstrated that inhibition of miR-25-3p abrogated the effects of JPX knockdown on IL-1β-stimulated injury in chondrocytes.

PPID is a target of miR-25-3p in chondrocytes

starBase analysis predicted that PPID was a target of miR-25-3p (Fig. 4A). The results of the dual-luciferase reporter assay and RIP analysis confirmed the targeted relationship between miR-25-3p and PPID (Fig. 4B and C). The results of RT-qPCR showed that PPID expression in OA patients was significantly higher than that in the healthy individuals (Fig. 4D). The correlation analysis showed that the PPID levels in OA patients were negatively associated with miR-25-3p and positively related to JPX levels (Fig. 4E and F). Next, the effect of miR-25-3p and JPX on PPID expression was assessed. miR-25-3p inhibitor significantly decreased miR-25-3p expression levels (Fig. 4G). Additionally, IL-1β treatment significantly increased PPID expression in chondrocytes (Fig. 4I and K). The protein expression levels of PPID were significantly increased by transfection of the miR-25-3p inhibitor whereas JPX knockdown significantly decreased PPID expression irrespective of IL-1β treatment (Fig. 4H and J). These results suggested that PPID is a target of miR-25-3p and it can be regulated by lncRNA JPX in chondrocytes.

Figure 4.

PPID is a target of miR-25-3p in chondrocytes. (A) starBase was used to predict the potential binding sites between miR-25-3p and the 3′UTR of PPID mRNA. (B and C) The targeted relationship between miR-25-3p and PPID was confirmed using a dual-luciferase reporter assay and RIP assay. **P<0.01. (D) PPID mRNA expression in OA and NC cartilage tissues were measured using RT-qPCR. (E) Correlation analysis between miR-25-3p and PPID in the OA cartilage tissues. (F) Correlation analysis between JPX and PPID in the OA cartilage tissues. (G) Expression of miR-25-3p in IL-1β-stimulated chondrocytes after transfection with the miR-318-3p inhibitor was measured using RT-qPCR. (H) PPID protein expression in chondrocytes after transfection with the miR-25-3p inhibitor. **P<0.01 vs. Control group. (I) PPID protein expression in chondrocytes after treatment with IL-1β and/or transfection with the miR-25-3p inhibitor was measured using western blotting. **P<0.01 vs. Control group; ##P<0.01 vs. IL-1β+inhibitor NC group. (J) PPID protein expression in chondrocytes after transfection with JPX siRNA was detected using western blotting. **P<0.01 vs. Control group. (K) PPID protein expression PPID in chondrocytes after treatment with IL-1β and/or transfection with JPX siRNA was detected by western blotting. **P<0.01 vs. Control group; ##P<0.01 vs. IL-1β+si-NC group. PPID, Peptidyl cis-trans prolyl isomerase D; miR, microRNA; UTR, untranslated region; RIP, RNA immunoprecipitation; OA, osteoarthritic; NC, negative control; siRNA, small interfering RNA.

PPID overexpression abrogates the effect of JPX knockdown in IL-1β-stimulated chondrocytes

To confirm whether JPX affected cartilage damage via regulation of PPID, JPX siRNA and PPID overexpression plasmid were transfected into chondrocytes. PPID was notably increased in the pc-PPID group compared with the pc-NC group (Fig. 5A). In the IL-1β-stimulated chondrocytes, JPX knockdown markedly decreased PPID expression compared with the IL-1β+si-NC group. Compared with the IL-1β+si-JPX#1+pc-NC group, the expression levels of PPID were significantly enhanced in the IL-1β+si-JPX#1+pc-PPID group (Fig. 5B). CCK-8 analysis showed that JPX knockdown increased cell viability in IL1β-stimulated chondrocytes, and the increase in cell viability following JPX knockdown was eliminated by PPID overexpression (Fig. 5C). As shown in Fig. 5D and E, JPX knockdown significantly decreased apoptosis of IL-1β-stimulated chondrocytes. Additionally, the inhibitory effect of JPX knockdown on apoptosis was eliminated by overexpression of PPID. JPX knockdown markedly increased Bcl-2 expression and reduced expression of Bax and c-caspase-9 in IL-1β-stimulated chondrocytes, and the effects of JPX knockdown on Bcl-2, Bax, and c-caspase-9 were eliminated by PPID overexpression (Fig. 5F and G). These results indicated that JPX knockdown may reduce cell injury in IL-1β-stimulated chondrocytes by regulating PPID expression.

Discussion

lncRNAs are associated with the function and inflammatory response of chondrocytes in OA (41,42). JPX has been reported to play an important role in myeloid malignancies (19). However, the underlying mechanism of JPX in OA remains unclear. In this study, C28/I2 cells were used to explore the effect of JPX on IL-1β-stimulated injury. The results showed that JPX knockdown enhanced cell viability and reduced apoptosis of IL-1β-stimulated chondrocytes. Moreover, the results showed that JPX was associated with the miR-25-3p/PPID axis in OA.

The cartilage damage is a characteristic and defining feature of OA (5,43). IL-1β has been reported to participate in OA progression (44). Therefore, chondrocyte C28/I2 cells were treated with IL-1β to establish an in vivo OA model. The results showed that IL-1β treatment reduced the viability of C28/I2 cells. Apoptosis is an important process related to cell viability (45). Activation of apoptosis-related proteins, such as Bcl-2, Bax, and caspase-3, are reliable markers of cell apoptosis (46,47). In this study, it was shown that IL-1β treatment facilitated apoptosis, which was observed as an increase in Bcl-2 levels, and a decrease in the levels of Bax and cleaved-caspase-9 in C28/I2 cells. Additionally, it was found that JPX expression was upregulated in OA patients and IL-1β-stimulated chondrocytes, which indicated that JPX may be associated with OA pathogenesis. The experiments confirmed that JPX knockdown suppressed the cell injury stimulated by IL-1β in chondrocytes, highlighting the therapeutic potential of JPX suppression on the progression of OA.

It has been found that JPX can competitively bind to various miRNAs as a ceRNA, such as miR-33a-5p (17), miR-155-5p (48), and miR-944 (18). In intervertebral disc degeneration, JPX upregulates HIF-1α expression by inhibiting miR-18a-5p in nucleus pulposus cells (49). In the present study, it was confirmed that JPX could bind to miR-25-3p as a ceRNA in C28/I2 cells. Additionally, miR-25-3p expression was downregulated in OA tissues and IL-1β-stimulated chondrocytes. miR-25-3p inhibitor reduced cell viability, promoted apoptosis in chondrocytes, and reversed the effects of JPX knockdown in chondrocytes. Li et al (50) found that miR-25-3p has anti-apoptotic effects on cultured primary neurons. Suppression of miR-25-3p reduced cell proliferation in a mouse model of polycystic kidney disease (51). The results of the present study are in agreement with the previous studies; the protective effects of JPX knockdown on chondrocytes were achieved by abrogating the effects of miR-25-3p.

As a member of the PPIase family, PPID knockdown protected HaCaT keratinocytes from death following UVA irradiation (52). In the present study, PPID was a target gene of miR-25-3p. PPID expression was upregulated in OA tissues and IL-1β-stimulated chondrocytes. Additionally, PPID expression was regulated by JPX, and JPX levels were positively correlated with PPID. Next, whether JPX could regulate PPID to affect IL-1β-stimulated chondrocytes was assessed. The data showed that PPID overexpression promoted apoptosis in chondrocytes and increased the expression of Bax and c-caspase-9. Moreover, PPID overexpression partly eliminated the influence of JPX knockdown on chondrocytes. These findings also resulted in accelerating the effect of PPID on OA progression, which was observed as the reversal of the protective effect of JPX knockdown in IL-1β-stimulated injury. Together, JPX can affect OA progression via actively modulating PPID through competitively sponging miR-25-3p.

The present study has some limitations. The role of JPX in OA was only explored in vitro, thus in vivo experiments should be performed to confirm the results in future studies. The effect of JPX on inflammatory response, oxidative stress, and other aspects associated with OA development/progression need further study. Finally, the number of patients included in the present study was low, thus the results should be confirmed in a larger cohort.

In conclusion, the results of the present study showed that lncRNA JPX increased the cell viability of chondrocytes and suppressed apoptosis in OA by modulating a miR-25-3p/PPID axis, thereby reducing the cell damage in OA. These findings highlight the JPX/miR-25-3p/PPID axis as a potentially novel therapeutic target in OA.

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

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

Authors' contributions

ZR designed the study. LT and ZD performed the experiments. JS, HZ and DL analyzed the data. ZR wrote the manuscript. ZR, LT and ZD confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of the Sunshine Union Hospital. All patients provided written informed consent.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

lncRNAs

long noncoding RNAs

OA

osteoarthritis

PPID

peptidylprolyl isomerase D

mir/miRNA

microRNA

CCK-8

Cell Counting Kit-8

RIP

RNA immunoprecipitation

ECM

extracellular matrix

JPX

Just proximal to X-inactive specific transcript

ceRNA

competitive endogenous RNA

PPIase

peptidyl cis-trans prolyl isomerase

TNF-α

tumor necrosis factor-α

References

1 

Sacitharan PK: Ageing and osteoarthritis. Subcell Biochem. 91:123–159. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Zhi L, Zhao J, Zhao H, Qing Z, Liu H and Ma J: Downregulation of LncRNA OIP5-AS1 induced by IL-1β aggravates osteoarthritis via regulating miR-29b-3p/PGRN. Cartilage. 3 (2_suppl):1345S–1355S. 2020.PubMed/NCBI

3 

Bolduc JA, Collins JA and Loeser RF: Reactive oxygen species, aging and articular cartilage homeostasis. Free Radic Biol Med. 132:73–82. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Charlier E, Deroyer C, Ciregia F, Malaise O, Neuville S, Plener Z, Malaise M and de Seny D: Chondrocyte dedifferentiation and osteoarthritis (OA). Biochem Pharmacol. 165:49–65. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Pap T and Korb-Pap A: Cartilage damage in osteoarthritis and rheumatoid arthritis-two unequal siblings. Nat Rev Rheumatol. 11:606–615. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Theocharis AD, Manou D and Karamanos NK: The extracellular matrix as a multitasking player in disease. FEBS J. 286:2830–2869. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Quinn JJ and Chang HY: Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 17:47–62. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Cao L, Wang Y, Wang Q and Huang J: LncRNA FOXD2-AS1 regulates chondrocyte proliferation in osteoarthritis by acting as a sponge of miR-206 to modulate CCND1 expression. Biomed Pharmacother. 106:1220–1226. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Wawrzyniak O, Zarębska Ż, Rolle K and Gotz-Więckowska A: Circular and long non-coding RNAs and their role in ophthalmologic diseases. Acta Biochim Pol. 65:497–508. 2018.PubMed/NCBI

10 

Wei JW, Huang K, Yang C and Kang CS: Non-coding RNAs as regulators in epigenetics (Review). Oncol Rep. 37:3–9. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Zhang L, Zhang P, Sun X, Zhou L and Zhao J: Long non-coding RNA DANCR regulates proliferation and apoptosis of chondrocytes in osteoarthritis via miR-216a-5p-JAK2-STAT3 axis. Biosci Rep. 38:BSR201812282018. View Article : Google Scholar : PubMed/NCBI

12 

Mao G, Kang Y, Lin R, Hu S and Zhang Z, Li H, Liao W and Zhang Z: Long Non-coding RNA HOTTIP promotes CCL3 expression and induces cartilage degradation by sponging miR-455-3p. Front Cell Dev Biol. 7:1612019. View Article : Google Scholar : PubMed/NCBI

13 

Ye D, Jian W, Feng J and Liao X: Role of long noncoding RNA ZFAS1 in proliferation, apoptosis and migration of chondrocytes in osteoarthritis. Biomed Pharmacother. 104:825–831. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Wang G, Bu X, Zhang Y, Zhao X, Kong Y, Ma L, Niu S, Wu B and Meng C: LncRNA-UCA1 enhances MMP-13 expression by inhibiting miR-204-5p in human chondrocytes. Oncotarget. 8:91281–91290. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Ma X, Yuan T, Yang C, Wang Z, Zang Y, Wu L and Zhuang L: X-inactive-specific transcript of peripheral blood cells is regulated by exosomal Jpx and acts as a biomarker for female patients with hepatocellular carcinoma. Ther Adv Med Oncol. 9:665–677. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Khajehdehi M, Khalaj-Kondori M and Hosseinpour Feizi MA: Expression profiling of cancer-related long non-coding RNAs revealed upregulation and biomarker potential of HAR1B and JPX in colorectal cancer. Mol Biol Rep. 49:6075–6084. 2022. View Article : Google Scholar : PubMed/NCBI

17 

Pan J, Fang S, Tian H, Zhou C, Zhao X, Tian H, He J, Shen W, Meng X, Jin X and Gong Z: lncRNA JPX/miR-33a-5p/Twist1 axis regulates tumorigenesis and metastasis of lung cancer by activating Wnt/β-catenin signaling. Mol Cancer. 19:92020. View Article : Google Scholar : PubMed/NCBI

18 

Yao Y, Chen S, Lu N, Yin Y and Liu Z: LncRNA JPX overexpressed in oral squamous cell carcinoma drives malignancy via miR-944/CDH2 axis. Oral Dis. 27:924–933. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Zimta AA, Tomuleasa C, Sahnoune I, Calin GA and Berindan-Neagoe I: Long Non-coding RNAs in myeloid malignancies. Front Oncol. 9:10482019. View Article : Google Scholar : PubMed/NCBI

20 

Gál Z, Gézsi A, Semsei ÁF, Nagy A, Sultész M, Csoma Z, Tamási L, Gálffy G and Szalai C: Investigation of circulating lncRNAs as potential biomarkers in chronic respiratory diseases. J Transl Med. 18:4222020. View Article : Google Scholar : PubMed/NCBI

21 

Chen Z, Ke X, Wang X, Kang H and Hong S: LncRNA JPX contributes to Treg/Th17 imbalance in allergic rhinitis via targeting the miR-378g/CCL5 axis. Immunopharmacol Immunotoxicol. 44:519–524. 2022. View Article : Google Scholar : PubMed/NCBI

22 

Mellis D and Caporali A: MicroRNA-based therapeutics in cardiovascular disease: Screening and delivery to the target. Biochem Soc Trans. 46:11–21. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Swingler TE, Niu L, Smith P, Paddy P, Le L, Barter MJ, Young DA and Clark IM: The function of microRNAs in cartilage and osteoarthritis. Clin Exp Rheumatol. 37 (Suppl):S40–S47. 2019.PubMed/NCBI

24 

Chen K, Zhu H, Zheng MQ and Dong QR: LncRNA MEG3 inhibits the degradation of the extracellular matrix of chondrocytes in osteoarthritis via targeting miR-93/TGFBR2 axis. Cartilage. 13 (2_Suppl):1274S–1284S. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Zhang P, Sun J, Liang C, Gu B, Xu Y, Lu H, Cao B and Xu H: lncRNA IGHCγ1 Acts as a ceRNA to regulate macrophage inflammation via the miR-6891-3p/TLR4 axis in osteoarthritis. Mediators Inflamm. 2020:97430372020. View Article : Google Scholar : PubMed/NCBI

26 

Yao N, Peng S, Wu H, Liu W, Cai D and Huang D: Long noncoding RNA PVT1 promotes chondrocyte extracellular matrix degradation by acting as a sponge for miR-140 in IL-1β-stimulated chondrocytes. J Orthop Surg Res. 17:2182022. View Article : Google Scholar : PubMed/NCBI

27 

Zhang Y, Ma L, Wang C, Wang L, Guo Y and Wang G: Long noncoding RNA LINC00461 induced osteoarthritis progression by inhibiting miR-30a-5p. Aging. 12:4111–4123. 2020. View Article : Google Scholar : PubMed/NCBI

28 

Lu J, Wu Z and Xiong Y: Knockdown of long noncoding RNA HOTAIR inhibits osteoarthritis chondrocyte injury by miR-107/CXCL12 axis. J Orthop Surg Res. 16:4102021. View Article : Google Scholar : PubMed/NCBI

29 

Kurowska W, Kuca-Warnawin E, Radzikowska A, Jakubaszek M, Maślińska M, Kwiatkowska B and Maśliński W: Monocyte-related biomarkers of rheumatoid arthritis development in undifferentiated arthritis patients-a pilot study. Reumatologia. 56:10–16. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Wang X, Ning Y, Zhou B, Yang L, Wang Y and Guo X: Integrated bioinformatics analysis of the osteoarthritis-associated microRNA expression signature. Mol Med Rep. 17:1833–1838. 2018.PubMed/NCBI

31 

He X and Deng L: Potential of miR-25-3p in protection of chondrocytes: Emphasis on osteoarthritis. Folia Histochem Cytobiol. 59:30–39. 2021. View Article : Google Scholar : PubMed/NCBI

32 

Fischer G, Berger E and Bang H: Kinetic beta-deuterium isotope effects suggest a covalent mechanism for the protein folding enzyme peptidylprolyl cis/trans-isomerase. FEBS Lett. 250:267–270. 1989. View Article : Google Scholar : PubMed/NCBI

33 

Galat A: Peptidylprolyl cis/trans isomerases (immunophilins): Biological diversity-targets-functions. Curr Top Med Chem. 3:1315–1347. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Lebedev I, Nemajerova A, Foda ZH, Kornaj M, Tong M, Moll UM and Seeliger MA: A novel in vitro CypD-Mediated p53 aggregation assay suggests a model for mitochondrial permeability transition by chaperone systems. J Mol Biol. 428:4154–4167. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Lu LQ, Tian J, Luo XJ and Peng J: Targeting the pathways of regulated necrosis: A potential strategy for alleviation of cardio-cerebrovascular injury. Cell Mol Life Sci. 78:63–78. 2021. View Article : Google Scholar : PubMed/NCBI

36 

Gan X, Zhang L, Liu B, Zhu Z, He Y, Chen J, Zhu J and Yu H: CypD-mPTP axis regulates mitochondrial functions contributing to osteogenic dysfunction of MC3T3-E1 cells in inflammation. J Physiol Biochem. 74:395–402. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Huang Z, Lan J and Gao X: Feprazone Mitigates IL-1β-Induced cellular senescence in chondrocytes. ACS Omega. 6:9442–9448. 2021. View Article : Google Scholar : PubMed/NCBI

38 

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

39 

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

40 

Yang JH, Li JH, Shao P, Zhou H, Chen YQ and Qu LH: starBase: A database for exploring microRNA-mRNA interaction maps from Argonaute CLIP-Seq and Degradome-Seq data. Nucleic Acids Res. 39:(Database Issue). D202–D209. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Pearson MJ and Jones SW: Review: Long Noncoding RNAs in the regulation of inflammatory pathways in rheumatoid arthritis and osteoarthritis. Arthritis Rheumatol. 68:2575–2583. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Cen X, Huang XQ, Sun WT, Liu Q and Liu J: Long noncoding RNAs: A new regulatory code in osteoarthritis. Am J Transl Res. 9:4747–4755. 2017.PubMed/NCBI

43 

Brooks P: Inflammation as an important feature of osteoarthritis. Bull World Health Organ. 81:689–690. 2003.PubMed/NCBI

44 

Yu T, Qu J, Wang Y and Jin H: Ligustrazine protects chondrocyte against IL-1β induced injury by regulation of SOX9/NF-κB signaling pathway. J Cell Biochem. 119:7419–7430. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Zuniga MC, Raghuraman G and Zhou W: Physiologic levels of resistin induce a shift from proliferation to apoptosis in macrophage and VSMC co-culture. Surgery. 163:906–911. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Yao C, Cao X, Fu Z, Tian J, Dong W, Xu J, An K, Zhai L and Yu J: Boschniakia rossica polysaccharide triggers laryngeal carcinoma cell apoptosis by regulating expression of Bcl-2, Caspase-3, and P53. Med Sci Monit. 23:2059–2064. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Kim JA, Kim JC, Min JS, Kang I, Oh J and Ahn JK: HSV-1 ICP27 induces apoptosis by promoting Bax translocation to mitochondria through interacting with 14-3-3θ. BMB Rep. 50:257–262. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Lin XQ, Huang ZM, Chen X, Wu F and Wu W: XIST Induced by JPX suppresses hepatocellular carcinoma by sponging miR-155-5p. Yonsei Med J. 59:816–826. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Yang H, Wang G, Liu J, Lin M, Chen J, Fang Y, Li Y, Cai W and Zhan D: LncRNA JPX regulates proliferation and apoptosis of nucleus pulposus cells by targeting the miR-18a-5p/HIF-1α/Hippo-YAP pathway. Biochem Biophys Res Commun. 566:16–23. 2021. View Article : Google Scholar : PubMed/NCBI

50 

Li R, Wen Y, Wu B, He M, Zhang P, Zhang Q and Chen Y: MicroRNA-25-3p suppresses epileptiform discharges through inhibiting oxidative stress and apoptosis via targeting OXSR1 in neurons. Biochem Biophys Res Commun. 523:859–866. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Liu G, Kang X, Guo P, Shang Y, Du R, Wang X, Chen L, Yue R and Kong F: miR-25-3p promotes proliferation and inhibits autophagy of renal cells in polycystic kidney mice by regulating ATG14-Beclin 1. Ren Fail. 42:333–342. 2020. View Article : Google Scholar : PubMed/NCBI

52 

Jandova J, Janda J and Sligh JE: Cyclophilin 40 alters UVA-induced apoptosis and mitochondrial ROS generation in keratinocytes. Exp Cell Res. 319:750–760. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2022
Volume 24 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ren Z, Tang L, Ding Z, Song J, Zheng H and Li D: Knockdown of lncRNA JPX suppresses IL‑1β‑stimulated injury in chondrocytes through modulating an miR‑25‑3p/PPID axis. Oncol Lett 24: 388, 2022
APA
Ren, Z., Tang, L., Ding, Z., Song, J., Zheng, H., & Li, D. (2022). Knockdown of lncRNA JPX suppresses IL‑1β‑stimulated injury in chondrocytes through modulating an miR‑25‑3p/PPID axis. Oncology Letters, 24, 388. https://doi.org/10.3892/ol.2022.13508
MLA
Ren, Z., Tang, L., Ding, Z., Song, J., Zheng, H., Li, D."Knockdown of lncRNA JPX suppresses IL‑1β‑stimulated injury in chondrocytes through modulating an miR‑25‑3p/PPID axis". Oncology Letters 24.5 (2022): 388.
Chicago
Ren, Z., Tang, L., Ding, Z., Song, J., Zheng, H., Li, D."Knockdown of lncRNA JPX suppresses IL‑1β‑stimulated injury in chondrocytes through modulating an miR‑25‑3p/PPID axis". Oncology Letters 24, no. 5 (2022): 388. https://doi.org/10.3892/ol.2022.13508