scholarly journals Long Non-coding RNA PVT1 Promotes Chondrocyte Extracellular Matrix Degradation by Acting as a Sponge for miR-140 in IL-1β-stimulated Chondrocytes

Author(s):  
Nan Yao ◽  
Sha Peng ◽  
Huai Wu ◽  
Wengang Liu ◽  
Dake Cai ◽  
...  

Abstract Background: Osteoarthritis (OA) is a common degenerative joint disease, and chondrocyte extracellular matrix (ECM) degradation is one vital pathological feature of OA. Long noncoding RNA (lncRNA), a new kind of gene regulator, plays an important role in pathogenesis of many diseases like OA. Recent studies have confirmed that lncRNA plasmacytoma variant translocation 1 (PVT1) expression was up-regulated in OA patients; however, its effect on ECM degradation remained unknown. Methods: Cartilage tissue samples were obtained from 6 OA patients admitted by Guangdong Second Traditional Chinese Medicine Hospital. Chondrocytes were isolated and cultured from the collected cartilage tissue. Plasmid construction, RNA interference, Cell transfection, Fluorescence in situ hybridization (FISH), and Pull-down assay were carried out during the research.Results: In this study, PVT1 expression was significantly increased in chondrocytes stimulated by interleukin-1β (IL-1β). In addition, inhibition of PVT1 significantly down-regulated the increased expressions of ADAM metallopeptidase with thrombospondin type 1 motif-5 (ADAMTS-5) and matrix metalloproteinase-13 (MMP-13) induced by IL-1β. Further investigation revealed that PVT1 was an endogenous sponge RNA, which directly bound to miR-140 and inhibited miR-140 expression. Conclusion: To sum up, this study showed that PVT1 promoted expressions of ADAMTS-5 and MMP-13 as a competing endogenous RNA (ceRNA) of miR-140 in OA, which eventually led to aggravation of ECM degradation, thus providing a new and promising strategy for the treatment of OA.

Cartilage ◽  
2019 ◽  
pp. 194760351985575 ◽  
Author(s):  
Kang Chen ◽  
Hao Zhu ◽  
Min-Qian Zheng ◽  
Qi-Rong Dong

Background As a degenerative joint disease, osteoarthritis (OA) is characterized by articular cartilage degradation. Long noncoding RNAs (lncRNAs) act critical roles in the regulation of OA development, including affecting the proliferation, apoptosis, extracellular matrix (ECM) degradation, and inflammatory response of chondrocytes. The current study’s aim was to investigate the regulatory function and the underlying molecular mechanism of lncRNA MEG3 in ECM degradation of chondrocytes in OA. Methods In the current study, chondrocytes were induced by interleukin-1β (IL-1β) to simulate OA condition, and further assessed cell viability, lncRNA MEG3 and miR-93 expression levels. Overexpression or knockdown of lncRNA MEG3 in chondrocytes treated with IL-1β were performed to investigate the function of MEG3 in regulating cell proliferation, apoptosis and ECM degradation using EdU assay, flow cytometry, quantitative reverse transcription polymerase chain reaction (qRT-PCR), and Western blot. The interaction between MEG3 and miR-93 was assessed using qRT-PCR. Furthermore, overexpression of miR-93 was performed as recovery experiment to explore the functional mechanism of MEG3. Results MEG3 was significantly downregulated in chondrocytes treated with IL-1β, whereas miR-93 was upregulated concomitantly. Overexpression of MEG3 induced the proliferation, suppressed the apoptosis, and relieved the degradation of ECM in IL-1β-induced chondrocytes. By contrast, knockdown of MEG3 suppressed the proliferation, promoted the apoptosis, and aggravated ECM degradation in IL-1β induced chondrocytes. In addition, MEG3 was found to relieve the inhibitive expression of TGFBR2 as a competitive endogenous RNA (ceRNA) of miR-93, and then activated transforming growth factor-β (TGF-β) signaling pathway, regulated chondrocytes ECM degradation in IL-1β induced chondrocytes subsequently. Conclusion LncRNA MEG3 targeted miR-93/TGFBR2 axis, regulated the proliferation, apoptosis and ECM degradation of chondrocytes in OA.


2021 ◽  
Vol 16 (1) ◽  
pp. 384-394
Author(s):  
Hu Han ◽  
Lijuan Liu

Abstract Osteoarthritis (OA) is a common chronic joint disease. This study aimed to explore the function of long noncoding RNA taurine-upregulated gene 1 (TUG1) in the progression and initiation of OA. Levels of TUG1, microRNA-320c (miR-320c) and fucosyltransferase 4 (FUT4) were examined via quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide and flow cytometry assays were used to detect cell viability and apoptosis, respectively. The expression of relative proteins was measured using Western blot. The interaction between miR-320c and TUG1 or FUT4 was confirmed utilizing dual-luciferase reporter and RNA immunoprecipitation assays. In this study, levels of TUG1 and FUT4 were distinctly upregulated, but miR-320c level significantly decreased in OA tissues and chondrocytes derived from OA tissues as well as in IL-1β-stimulated C28/I2 cells. Mechanically, TUG1 sponged miR-320c and miR-320c targeted FUT4. In addition, TUG1 knockdown accelerated cell proliferation and repressed apoptosis and extracellular matrix (ECM) degradation in IL-1β-induced C28/I2 cells, whereas these effects of TUG1 deletion were rescued by either miR-320c inhibitor or FUT4 upregulation. Meanwhile, TUG1 sponged miR-320c to regulate FUT4 expression in IL-1β-induced C28/I2 cells. Collectively, TUG1 modulated cell proliferation, apoptosis and ECM degradation in IL-1β-induced C28/I2 cells via the miR-320c/FUT4 axis, providing a new insight into the OA treatment.


2017 ◽  
Vol 43 (2) ◽  
pp. 602-610 ◽  
Author(s):  
Yu-Fei Li ◽  
Shu-Hua Li ◽  
Yong Liu ◽  
Ya-Tong Luo

Background/Aims: Osteoarthritis (OA) is a degenerative joint disease characterized by articular cartilage degradation. The degradation of the extracellular matrix (ECM) of chondrocyte is closely associated with the destruction of joints in OA patients. lncRNAs are non-coding segments of RNA that possess important regulatory functions at the cellular level and in a variety of pathophysiological processes. The present study was conducted to investigate whether lncRNA-CIR regulated the expression of MMP13 as a sponge of miR-27 in OA. Methods: Primary cultured chondrocytes were challenged by IL-1β and TNF-α to simulate OA conditions. qRT-PCR was performed to detect the miR-27, lncRNA-CIR, MMP13 mRNA expression levels. Western blot was applied to detect MMP13 protein expression. Soluble sGAG secretion/ formation was analysed by the dimethylmethylene blue (DMMB) assay. lncRNA-CIR overexpression or inhibition was performed using overexpression plasmid and small interfering RNAs (siRNAs), respectively. Results: lncRNA-CIR significantly up-regulated in OA patients, concomitantly down-regulated miR-27 and up-regulated MMP13. Bioinformatics analysis predicted miR-27 was the target of both lncRNA-CIR and MMP13. Overexpression of lncRNA-CIR significantly increased the expression of MMP13, while miR-27 remarkably suppressed the expression of MMP13, Accompanying with the increases of mRNA level, protein level and relative luciferase activity. Conclusion: The present findings indicated that lncRNA-CIR/miR-27/MMP13 axis involved in the degradation of the ECM of chondrocyte in OA.


2021 ◽  
Vol 16 (1) ◽  
Author(s):  
Jipeng Lu ◽  
Zhongxiong Wu ◽  
Ying Xiong

Abstract Background Osteoarthritis (OA) is a joint disease characterized via destruction of cartilage. Chondrocyte damage is associated with cartilage destruction during OA. Long noncoding RNAs (lncRNAs) are implicated in the regulation of chondrocyte damage in OA progression. This study aims to investigate the role and underlying mechanism of lncRNA homeobox antisense intergenic RNA (HOTAIR) in OA chondrocyte injury. Methods Twenty-three OA patients and healthy controls without OA were recruited. Chondrocytes were isolated from OA cartilage tissues. HOTAIR, microRNA-107 (miR-107) and C-X-C motif chemokine ligand 12 (CXCL12) levels were measured by quantitative real-time polymerase chain reaction and western blot. Cell proliferation, apoptosis and extracellular matrix (ECM) degradation were measured using cell counting kit-8, flow cytometry and western blot. The target interaction was explored by bioinformatics, luciferase reporter and RNA immunoprecipitation assays. Results HOTAIR expression was enhanced, and miR-107 level was reduced in OA cartilage samples. HOTAIR overexpression inhibited cell proliferation, but induced cell apoptosis and ECM degradation in chondrocytes. HOTAIR knockdown caused an opposite effect. MiR-107 was sponged and inhibited via HOTAIR, and knockdown of miR-107 mitigated the effect of HOTAIR silence on chondrocyte injury. CXCL12 was targeted by miR-107. CXCL12 overexpression attenuated the roles of miR-107 overexpression or HOTAIR knockdown in the proliferation, apoptosis and ECM degradation. CXCL12 expression was decreased by HOTAIR silence, and restored by knockdown of miR-107. Conclusion HOTAIR knockdown promoted chondrocyte proliferation, but inhibited cell apoptosis and ECM degradation in OA chondrocytes by regulating the miR-107/CXCL12 axis.


2020 ◽  
Author(s):  
Qinghui Ji ◽  
Xiaofeng Qiao ◽  
Yongxiang Liu ◽  
Dawei Wang ◽  
Jinglong Yan

Abstract Objective: To investigate the expression of long chain non-coding RNA GAS5 in osteoarthritis(OA) and the effect on autophagy and apoptosis of chondrocytes. Method: OA rat model was constructed and the expressions of GAS5 in articular cartilage tissues at 4 weeks(early OA) and 12 weeks(late OA) after modeling were detected. The chondrocytes of OA rat were isolated, cultured and transfected with si-GAS5 to silence the GAS5. Then the change of apoptosis and autophagy levels of OA chondrocytes were detected. The microRNA binding to GAS5 and the competitive target gene were analyzed and verified. Results: The expression of GAS5 in cartilage tissue of OA rats was higher than that of control, which was higher in late OA than that in early OA. After silencing the GAS5, the autophagy ability of OA chondrocytes was increased and the apoptosis rate was decreased. GAS5 could bind with miR-144 competitively and regulate mTOR positively. The mTOR inhibitor GDC-0349 could reverse the inhibition of GAS5 on cell autophagy but could not reverse the promotion on apoptosis. Conclusion: GAS5 expression was high and increased with the progression of OA. GAS5 could inhibit the autophagy and promote the apoptosis of OA chondrocytes, and the effect on autophagy may be related to the regulation on mTOR.


Author(s):  
Sean K Bedingfield ◽  
Fang Yu ◽  
Danielle D. Liu ◽  
Meredith A. Jackson ◽  
Lauren E. Himmel ◽  
...  

AbstractOsteoarthritis (OA) is a debilitating and prevalent chronic disease, but there are no approved disease modifying OA drugs (DMOADs), only pharmaceuticals for pain management. OA progression, particularly for post-traumatic osteoarthritis (PTOA), is associated with inflammation and enzymatic degradation of the extracellular matrix. In particular, Matrix Metalloproteinase 13 (MMP13) breaks down collagen type 2 (CII), a key structural component of cartilage extracellular matrix, and consequently, matrix degradation fragments perpetuate inflammation and a degenerative cycle that leads to progressive joint pathology. Here, we tested targeted delivery of endosome-escaping, MMP13 RNA interference (RNAi) nanoparticles (NPs) as a DMOAD. The new targeting approach pursued here deviates from the convention of targeting specific cell types (e.g., through cell surface receptors) and instead leverages a monoclonal antibody (mAbCII) that targets extracellular CII that becomes uniquely accessible at early OA focal defects. Targeted mAbCII-siNPs create an in situ NP depot for retention and potent activity within OA joints. The mAbCII-siNPs loaded with MMP13 siRNA (mAbCII-siNP/siMMP13) potently suppressed MMP13 expression (95% silencing) in TNFα-stimulated chondrocytes in vitro, and the targeted mAbCII-siNPs had higher binding to trypsin-damaged porcine cartilage than untargeted control NPs. In an acute mechanical injury mouse model of PTOA, mAbCII-siNP/siMMP13 achieved 80% reduction in MMP13 expression (p = 0.00231), whereas a non-targeted control achieved only 55% silencing. In a more severe, PTOA model, weekly mAbCII-siNP/siMMP13 long-term treatment provided significant protection of cartilage integrity (0.45+/− .3 vs 1.6+/−.5 on the OARSI scale; p=0.0166), and overall joint structure (1.3+/−.6 vs 2.8+/−.2 on the Degenerative Joint Disease scale; p<0.05). Intra-articular mAbCII-siNPs better protected articular cartilage (OARSI score) relative to either single or weekly treatment with the clinical gold stand steroid treatment methylprednisolone. Finally, multiplexed gene expression analysis of 254 inflammation-related genes showed that MMP13 inhibition suppressed clusters of genes associated with tissue restructuring, angiogenesis (associated with synovial inflammation and thickening), innate immune response, and proteolysis. This work establishes the new concept of targeting unique local extracellular matrix signatures to sustain retention and increase delivery efficacy of biologics with intracellular activity and also validates the promise of MMP13 RNAi as a DMOAD in a clinically-relevant therapeutic context. Abstract Figure:PTOA targeted delivery of MMP13 siRNA to block disease progressionThe top left schematic illustrates the progression (left to right) from healthy knee joint, through inflammation induction following traumatic injury, to cartilage loss and degenerative joint disease (including synovial response). Degradation of cartilage enhances inflammation, inducing a degenerative cycle (middle right). The bottom of the graphic illustrates the concept of the matrix targeted nanocarriers for enhanced retention and activity of MMP13 siRNA at sites of cartilage injury.


2006 ◽  
Vol 58 (2) ◽  
pp. 300-322 ◽  
Author(s):  
D NESIC ◽  
R WHITESIDE ◽  
M BRITTBERG ◽  
D WENDT ◽  
I MARTIN ◽  
...  

2020 ◽  
Author(s):  
Hongdan Wang ◽  
Cunying Cui ◽  
Yanan Li ◽  
Yuanyuan Liu ◽  
Taibing Fan ◽  
...  

ABSTRACTTetralogy of Fallot (TOF) is still the most common and complicated cyanotic congenital heart defect of all congenital heart diseases with a 10% incidence. Surgery repair is often necessary in infancy. The etiology of TOF is complex and genetic and epigenetic mechanisms such as chromosomal abnormalities, gene mutations, nucleic acid modifications, non-coding RNA, and circular RNA(circRNA) play an important role in its occurrence. RNA not only plays an auxiliary role of genetic information carrier, but also plays a more important role in various regulatory functions. There are few studies on the action mechanism of non-coding RNA. Aim to gain more in-depth knowledge of TOF, we collected tissue samples of the right ventricular outflow tract of 5 TOF children with no other intracardiac and extracardiac malformations and 5 normal fetuses. We systematically analyzed the specific long non-coding RNA (lncRNA), microRNA(miRNA), circRNA and messenger RNA(mRNA) profiles of TOF. To our knowledge, there are no reports of genome-wide study of transcriptome in TOF and we first obtained meaningful differentially expressed lncRNAs, miRNAs, circle RNA and mRNAs.


2021 ◽  
Author(s):  
Jian Hao ◽  
Yun Zhou ◽  
Weimin Yu ◽  
Hui Li ◽  
Dandan He

Abstract Background: LncRNA have been increasingly shown that plays pivotal roles in the development of various diseases, including renal fibrosis. Nevertheless, the pathological function of Long non-coding RNA SNHG10 (SNHG10) in the renal fibrosis remains obscure.Methods: We detected the expression levels of SNHG10 in the tissue samples and cell lines via RT-qPCR analysis. The functions of SNHG10 on the progression of renal fibrosis were examined by CCK-8, EdU, dual luciferase reporter and immunofluorescence analyses.Results: In the present study, SNHG10, production of extracellular matrix (ECM), including α-SMA and Fibronectin levels were significantly increased in HK-2 cells after TGF-β stimulation. Ectopic of SNHG10 inhibited cell proliferation and inhibits theα-SMA and Fibronectin expression of TGF-β1-induced HK-2 cells. In addition, bioinformatics analysis and dual luciferase reporter assay indicated that miR-378b was a target gene of SNHG10. Mechanistically, miR-378b overexpression abolished the repressive effects of SNHG10 on TGF-β1-induced HK-2 cells.Conclusion: SNHG10 plays an anti-fibrotic effect through suppression of miR-378b expression in renal fibrosis, which provides a promising therapeutic target for the treatment of renal fibrosis.


2020 ◽  
Vol 107 (2) ◽  
pp. 231-242
Author(s):  
Sara Asadi ◽  
Parvin Farzanegi ◽  
Mohammad Ali Azarbayjani

AbstractPurposeKnee osteoarthritis (OA) is a common type of degenerative joint disease which decreases the quality of life. Sex-determining region Y box 9 (SOX9) and hypoxia-inducible factor-1 (HIF1) are considered as the key regulators of OA. We investigated the effect of combined therapies with mesenchymal stem cells (MSCs), ozone (O3) and exercise training on SOX9 and HIF1 expression in the cartilage of rats with knee OA.MethodsKnee OA was induced by surgical method. OA rats were divided into model, MSCs, ozone, exercise, MSCs + ozone, MSCs + exercise, ozone + exercise and MSCs + ozone + exercise groups. Rats in the MSCs group received intraarticular injection of 1 × 106 cells/kg. Rats in the ozone group received O3 at the concentration of 20 μg/mL, once weekly for 3 weeks. Rats in the exercise group were trained on rodent treadmill three times per week. 48 hours after the programs, cartilage tissues were isolated and the expression of SOX9 and HIF1 was determined using Real-Time PCR.ResultsSignificant differences were found in the expression of SOX9 and HIF1 between groups (P < 0.0001). Although combined therapies with exercise, MSCs and O3 significantly increased the expression of SOX9 and HIF1 in the cartilage tissue of rats with knee OA, combination of exercise with O3 was significantly more effective compared to the other combined therapies (P < 0.001).ConclusionsCombined therapy with exercise, MSCs and O3 significantly increased the expression of SOX9 and HIF1 genes in the cartilage of rats with knee OA; however, exercise + O3 was significantly more effective.


Sign in / Sign up

Export Citation Format

Share Document