Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
Front Immunol ; 12: 781185, 2021.
Article in English | MEDLINE | ID: mdl-34956209

ABSTRACT

Osteoarthritis (OA) is a serious joint inflammation that leads to cartilage degeneration and joint dysfunction. Mesenchymal stem cells (MSCs) are used as a cell-based therapy that showed promising results in promoting cartilage repair. However, recent studies and clinical trials explored unsatisfied outcomes because of slow chondrogenic differentiation and increased calcification without clear reasons. Here, we report that the overexpression of indoleamine 2,3 dioxygenase 1 (IDO1) in the synovial fluid of OA patients impairs chondrogenic differentiation of MSCs in the joint of the OA mice model. The effect of MSCs mixed with IDO1 inhibitor on the cartilage regeneration was tested compared to MSCs mixed with IDO1 in the OA animal model. Further, the mechanism exploring the effect of IDO1 on chondrogenic differentiation was investigated. Subsequently, miRNA transcriptome sequencing was performed for MSCs cocultured with IDO1, and then TargetScan was used to verify the target of miR-122-5p in the SF-MSCs. Interestingly, we found that MSCs mixed with IDO1 inhibitor showed a significant performance to promote cartilage regeneration in the OA animal model, while MSCs mixed with IDO1 failed to stimulate cartilage regeneration. Importantly, the overexpression of IDO1 showed significant inhibition to Sox9 and Collagen type II (COL2A1) through activating the expression of ß-catenin, since inhibiting of IDO1 significantly promoted chondrogenic signaling of MSCs (Sox9, COL2A1, Aggrecan). Further, miRNA transcriptome sequencing of SF-MSCs that treated with IDO1 showed significant downregulation of miR-122-5p which perfectly targets Wnt1. The expression of Wnt1 was noticed high when IDO1 was overexpressed. In summary, our results suggest that IDO1 overexpression in the synovial fluid of OA patients impairs chondrogenic differentiation of MSCs and cartilage regeneration through downregulation of miR-122-5p that activates the Wnt1/ß-catenin pathway.


Subject(s)
Chondrogenesis/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Osteoarthritis, Knee/pathology , Animals , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Cartilage, Articular/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Chondrogenesis/drug effects , Female , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mesenchymal Stem Cells/drug effects , Mice , MicroRNAs/metabolism , Middle Aged , Osteoarthritis, Knee/enzymology , Rats , Rats, Wistar , Regeneration/drug effects , Regeneration/physiology , Synovial Fluid/enzymology
2.
Int Immunopharmacol ; 90: 106846, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33168412

ABSTRACT

Accumulating evidence has highlighted the remarkable role of long noncoding RNAs (lncRNAs) in the pathogenesis of various diseases including osteoarthritis (OA). Since current treatment available for OA has limited efficacy, it is urgent to elucidate the pathogenesis of OA. Therefore, we aimed at elucidating the specific regulatory role of LINC00671 in OA progression. Differentially expressed lncRNAs were initially screened using the OA profile. LINC00671, ONECUT2, and Smurf2 expression in OA cartilage tissues were determined, while their interaction was verified by RNA-pull down assay, ChIP, and dual-luciferase reporter gene assay. After chondrocytes were transfected with shRNA and overexpressed plasmids, the proliferation and apoptosis were determined. Meanwhile, extracellular matrix (ECM)-related proteins were detected by Western blot analysis. Establishment of the OA model was performed by surgical destabilization of the medial meniscus (DMM) surgery in mice. Upregulation of LINC00671, ONECUT2, and Smurf2 expression were detected in OA cartilage. LINC00671 was bound to ONECUT2 and ONECUT2 was conjugated to Smurf2. Overexpression of LINC00671 resulted in inhibited chondrocytes proliferation, enhanced apoptosis, and ECM degradation, which was readily reversed by silencing ONECUT2 or Smurf2. Furthermore, LINC00671 induced GSK-3ß ubiquitination and upregulated ß-catenin expression through Smurf2. In vivo experiment revealed that silencing of LINC00671 or GSK-3ß activator resulted in alleviated ECM degradation and ameliorated OA progression. Collectively, these data demonstrated that LINC00671 exacerbates OA progression through GSK-3ß ubiquitination by upregulating ONECUT2-mediated Smurf2.


Subject(s)
Chondrocytes/enzymology , Extracellular Matrix/enzymology , Homeodomain Proteins/metabolism , Knee Joint/enzymology , Osteoarthritis, Knee/enzymology , RNA, Long Noncoding/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Adult , Aged , Animals , Apoptosis , Case-Control Studies , Cell Proliferation , Cells, Cultured , Chondrocytes/pathology , Databases, Genetic , Disease Models, Animal , Disease Progression , Extracellular Matrix/pathology , Female , Gene Expression Regulation , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Homeodomain Proteins/genetics , Humans , Knee Joint/pathology , Male , Mice, Inbred C57BL , Middle Aged , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , RNA, Long Noncoding/genetics , Signal Transduction , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
3.
Int J Mol Sci ; 21(18)2020 Sep 22.
Article in English | MEDLINE | ID: mdl-32971951

ABSTRACT

Knee osteoarthritis (OA) is a condition mainly characterized by cartilage degradation. Currently, no effective treatment exists to slow down the progression of OA-related cartilage damage. Selective COX-2 inhibitors may, next to their pain killing properties, act chondroprotective in vivo. To determine whether the route of administration is important for the efficacy of the chondroprotective properties of selective COX-2 inhibitors, a systematic review was performed according to the PRISMA guidelines. Studies investigating OA-related cartilage damage of selective COX-2 inhibitors in vivo were included. Nine of the fourteen preclinical studies demonstrated chondroprotective effects of selective COX-2 inhibitors using systemic administration. Five clinical studies were included and, although in general non-randomized, failed to demonstrate chondroprotective actions of oral selective COX-2 inhibitors. All of the four preclinical studies using bolus intra-articular injections demonstrated chondroprotective actions, while one of the three preclinical studies using a slow release system demonstrated chondroprotective actions. Despite the limited evidence in clinical studies that have used the oral administration route, there seems to be a preclinical basis for considering selective COX-2 inhibitors as disease modifying osteoarthritis drugs when used intra-articularly. Intra-articularly injected selective COX-2 inhibitors may hold the potential to provide chondroprotective effects in vivo in clinical studies.


Subject(s)
Chondrocytes , Cyclooxygenase 2 Inhibitors/therapeutic use , Cyclooxygenase 2/metabolism , Cytoprotection/drug effects , Osteoarthritis, Knee , Animals , Chondrocytes/enzymology , Chondrocytes/pathology , Humans , Osteoarthritis, Knee/drug therapy , Osteoarthritis, Knee/enzymology , Osteoarthritis, Knee/pathology
4.
Biomed Pharmacother ; 129: 110471, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32768958

ABSTRACT

Huoxuezhitong capsule (HXZT, activating blood circulation and relieving pain capsule), has been applied for osteoarthritis since 1974. It consists of Angelica sinensis (Oliv.) Diels, Panax notoginseng (Burkill) F. H. Chen ex C. H., Boswellia sacra, Borneol, Eupolyphaga sinensis Walker, Pyritum. However, the direct effects of HXZT on osteoarthritis and the underlying mechanisms were poorly understood. In this study, we aimed to explore the analgesia effect of HXZT on MIA-induced osteoarthritis rat and the underlying mechanisms. The analgesia and anti-inflammatory effect of HXZT on osteoarthritis in vivo were tested by the arthritis model rats induced by monosodium iodoacetate (MIA).. Mechanistic studies confirmed that HXZT could inhibit the activation of NF-κB and down-regulate the mRNA expression of related inflammatory factors in LPS-induced RAW264.7 and ATDC5 cells. Furtherly, in LPS-induced RAW264.7 cells, HXZT could suppress NF-κB via inhibiting PI3K/Akt pathway. Taken together, HXZT capsule could ameliorate MIA-induced osteoarthritis of rats through suppressing PI3K/ Akt/ NF-κB pathway.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Experimental/prevention & control , Drugs, Chinese Herbal/pharmacology , Knee Joint/drug effects , NF-kappa B/metabolism , Osteoarthritis, Knee/prevention & control , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Capsules , Cytokines/metabolism , Inflammation Mediators/metabolism , Iodoacetic Acid , Knee Joint/enzymology , Knee Joint/pathology , Male , Mice , Osteoarthritis, Knee/chemically induced , Osteoarthritis, Knee/enzymology , Osteoarthritis, Knee/pathology , Phosphorylation , RAW 264.7 Cells , Rats, Sprague-Dawley , Signal Transduction
5.
Biomed Pharmacother ; 128: 110349, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32521454

ABSTRACT

Long non-coding RNAs (lncRNAs) are largely involved in the development of osteoarthritis (OA), a chronic and degenerative joint disease. The objective of this paper is to research the functional role and molecular mechanism of lncRNA X inactive specific transcript (XIST) in OA. The levels of XIST, microRNA-149-5p (miR-149-5p), and DNA methyltransferase 3A (DNMT3A) were measured. Cell viability and apoptosis rate were determined. Associated protein levels were examined through Western blot. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were implemented for confirming the target relation. And the role of XIST on OA in vivo was investigated by a rat model. XIST was expressed at a high level in OA cartilage tissues and IL-1ß-treated chondrocytes. XIST knockdown promoted cell viability but restrained cell apoptosis and extracellular matrix (ECM) protein degradation in IL-1ß-treated chondrocytes. XIST directly targeted miR-149-5p and miR-149-5p down-regulation restored si-XIST-mediated pro-proliferative and anti-apoptotic or ECM degradative effects. DNMT3A was a target gene of miR-149-5p and DNMT3A overexpression ameliorated miR-149-5p-induced promotion of cell viability but repression of apoptosis and ECM degradation. Knockdown of XIST reduced DNMT3A level by motivating miR-149-5p expression. The inhibitory influence of XIST down-regulation on OA evolvement was also achieved by miR-149-5p/DNMT3A axis in vivo. In a word, knockdown of XIST can repress the development of OA by miR-149-5p/DNMT3A axis. This study discovers the XIST/miR-149-5p/DNMT3A axis in regulating OA evolution, which is beneficial for understanding the molecular pathomechanism and can lay a good foundation for targeted therapy of OA treatment.


Subject(s)
Chondrocytes/enzymology , DNA (Cytosine-5-)-Methyltransferases/metabolism , MicroRNAs/metabolism , Osteoarthritis, Knee/enzymology , RNA, Long Noncoding/metabolism , Animals , Apoptosis , Arthritis, Experimental/enzymology , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Arthritis, Experimental/prevention & control , Case-Control Studies , Cell Line , Cell Proliferation , Chondrocytes/pathology , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Gene Expression Regulation, Enzymologic , Humans , MicroRNAs/genetics , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , Osteoarthritis, Knee/prevention & control , RNA, Long Noncoding/genetics , Rats, Wistar , Signal Transduction
6.
Biosci Rep ; 40(6)2020 06 26.
Article in English | MEDLINE | ID: mdl-32395770

ABSTRACT

BACKGROUND/AIMS: MicroRNAs (miRNAs) are involved in the pathogenesis of osteoarthritis (OA). The present study aimed to investigate the potential function of miR-122 in the development of OA and its potential molecular mechanisms. METHODS: The expression of miR-122, silent information regulator 1 (SIRT1), collagen II, aggrecan, matrix metalloproteinase (MMP) 13 (MMP13) and ADAMTS4 in OA cartilage was detected by RT-qPCR. Target gene prediction and screening, luciferase reporter assay were used to verify downstream target genes of miR-122. RESULTS: Compared with osteonecrosis, the expression of miR-122 was significantly increased in OA cartilage, while the expression of SIRT1 was significantly decreased. Overexpression of miR-122 increased the expression of extracellular matrix (ECM) catabolic factors, for example disintegrins, MMPs and metalloproteinases with platelet reaction protein motifs, and inhibited the expression of synthetic metabolic genes such as collagen II and aggregating proteoglycan. Inhibition of miR-122 expression had the opposite effect. Furthermore, SIRT1 was identified as a direct target of miR-122. SIRT1 was significantly inhibited by miR-122 overexpression. Knockdown of SIRT1 reversed the degradation of chondrocyte ECM by miR-122 inhibitors. CONCLUSION: The miR-122/SIRT1 axis can regulate the degradation of ECM in OA, thus providing new insights into the treatment of OA.


Subject(s)
Cartilage, Articular/enzymology , Chondrocytes/enzymology , Extracellular Matrix/enzymology , Knee Joint/enzymology , MicroRNAs/metabolism , Osteoarthritis, Knee/enzymology , Sirtuin 1/metabolism , Aged , Cartilage, Articular/pathology , Case-Control Studies , Cell Line, Tumor , Chondrocytes/pathology , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Female , Gene Expression Regulation, Enzymologic , Humans , Knee Joint/pathology , Male , MicroRNAs/genetics , Middle Aged , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , Proteolysis , Signal Transduction , Sirtuin 1/genetics
7.
Int J Rheum Dis ; 23(3): 435-442, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31957331

ABSTRACT

OBJECTIVE: The aim of this study was to investigate cross-sectional associations between serum levels of citrate and knee structural changes and cartilage enzymes in patients with knee osteoarthritis (OA). METHOD: A total of 137 subjects with symptomatic knee OA (mean age 55.0 years, range 34-74, 84% female) were included. Knee radiography was used to assess knee osteophytes, joint space narrowing (JSN) and radiographic OA assessed by Kellgren-Lawrence (K-L) grading system. T2-weighted fat-suppressed fast spin echo magnetic resonance imaging (MRI) was used to determine knee cartilage defects, bone marrow lesions (BMLs) and infrapatellar fat pad (IPFP) signal intensity alternations. Colorimetric fluorescence was used to measure the serum levels of citrate. Enzyme-linked immunosorbent assay was used to measure the serum cartilage enzymes including matrix metalloproteinase (MMP)-3 and MMP-13. RESULTS: After adjustment for potential confounders (age, sex, body mass index), serum citrate was negatively associated with knee osteophytes at the femoral site, cartilage defects at medial femoral site, total cartilage defects, and total BMLs (odds ratio [OR] 0.17-0.30, all P < .05). Meanwhile, serum citrate was negatively associated with IPFP signal intensity alteration (OR 0.30, P = .05) in multivariable analyses. Serum citrate was significantly and negatively associated with MMP-13 (ß -3106.37, P < .05) after adjustment for potential confounders. However, citrate was not significantly associated with MMP-3 in patients with knee OA. CONCLUSION: Serum citrate was negatively associated with knee structural changes including femoral osteophytes, cartilage defects, and BMLs and also serum MMP-13 in patients with knee OA, suggesting that low serum citrate may be a potential indicator for advanced knee OA.


Subject(s)
Cartilage, Articular/enzymology , Citric Acid/blood , Knee Joint/enzymology , Matrix Metalloproteinase 13/blood , Osteoarthritis, Knee/blood , Adult , Aged , Biomarkers/blood , Cartilage, Articular/diagnostic imaging , Cross-Sectional Studies , Female , Humans , Knee Joint/diagnostic imaging , Male , Middle Aged , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/enzymology , Prognosis
8.
Biosci Rep ; 40(1)2020 01 31.
Article in English | MEDLINE | ID: mdl-31868209

ABSTRACT

Osteoarthritis is mainly caused by a degenerative joint disorder, which is characterized by the gradual degradation of articular cartilage and synovial inflammation. The chondrocyte, the unique resident cell type of articular cartilage, is crucial for the development of osteoarthritis. Previous studies revealed that P21-activated kinase-1 (PAK1) was responsible for the initiation of inflammation. The purpose of the present study was to determine the potential role of PAK1 in osteoarthritis. The level of PAK1 expression was measured by Western blot and quantitative real-time PCR in articular cartilage from osteoarthritis model rats and patients with osteoarthritis. In addition, the functional role of aberrant PAK1 expression was detected in the chondrocytes. We found that the expression of PAK1 was significantly increased in chondrocytes treated with osteoarthritis-related factors. Increased expression of PAK1 was also observed in knee articular cartilage samples from patients with osteoarthritis and osteoarthritis model rats. PAK1 was found to inhibit chondrocytes proliferation and to promote the production of inflammatory cytokines in cartilages chondrocytes. Furthermore, we found that PAK1 modulated the production of extracellular matrix and cartilage degrading enzymes in chondrocytes. Results of the present studies demonstrated that PAK1 might play an important role in the pathogenesis of osteoarthritis.


Subject(s)
Arthritis, Experimental/enzymology , Cartilage, Articular/enzymology , Chondrocytes/enzymology , Knee Joint/enzymology , Osteoarthritis, Knee/enzymology , p21-Activated Kinases/metabolism , Aged , Aged, 80 and over , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Cell Proliferation , Cells, Cultured , Chondrocytes/pathology , Cytokines/metabolism , Female , Humans , Inflammation Mediators/metabolism , Knee Joint/pathology , Male , Middle Aged , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation , p21-Activated Kinases/genetics
9.
J Orthop Res ; 37(11): 2429-2436, 2019 11.
Article in English | MEDLINE | ID: mdl-31304988

ABSTRACT

Knee injuries cause structural damage and acute inflammation that initiates the development of post-traumatic osteoarthritis (PTOA). NADPH oxidase 4 (Nox4), a member of a family of enzymes that generates reactive oxygen species (ROS), plays a pivotal role in normal development of the musculoskeletal system, but may increase ROS production to harmful levels after joint injury. The role of ROS in both normal joint homeostasis and injury is poorly understood, but inhibition of excessive ROS production by Nox4 after joint injury could be protective to the joint, decreasing oxidative stress, and initiation of PTOA. Knee injuries were simulated using inflammatory cytokines in cultured primary human chondrocytes and a non-invasive mouse model of PTOA in C57BL/6N and Nox4 knockout mice. There is an acute decrease in Nox4 activity within 24 h after injury in both systems, followed by a subsequent sustained low-level increase, a novel finding not seen in any other system. Inhibition of Nox4 activity by GKT137831 was protective against early structural changes after non-invasive knee injury in a mouse model. Nox4 knockout mice had significant differences in structural and mechanical properties of bone, providing further evidence for the role of Nox4 in development of joint tissues and biochemical response after joint injury. Nox4 plays a significant role in the acute phase after joint injury, and targeted inhibition of inflammation caused by Nox4 may be protective against early joint changes in the pathogenesis of PTOA. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2429-2436, 2019.


Subject(s)
Anterior Cruciate Ligament Injuries/complications , Chondrocytes/enzymology , NADPH Oxidase 4/metabolism , Osteoarthritis, Knee/enzymology , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Adolescent , Adult , Animals , Drug Evaluation, Preclinical , Female , Humans , Hydrogen Peroxide/metabolism , Male , Mice, Knockout , Middle Aged , NADPH Oxidase 4/antagonists & inhibitors , NADPH Oxidase 4/genetics , Osteoarthritis, Knee/etiology , Osteoarthritis, Knee/prevention & control , Primary Cell Culture , Pyrazoles/pharmacology , Pyrazolones , Pyridines/pharmacology , Pyridones , Young Adult
10.
Osteoarthritis Cartilage ; 27(5): 833-843, 2019 05.
Article in English | MEDLINE | ID: mdl-30685487

ABSTRACT

OBJECTIVE: We investigated the effects of 29-kDa amino-terminal fibronectin fragment (29-kDa FN-f) on xylosyltransferase-1 (XT-1), an essential anabolic enzyme that catalyzes the initial and rate-determining step in glycosaminoglycan chain synthesis, in human primary chondrocytes. METHODS: Proteoglycan and XT-1 expression in cartilage tissue was analyzed using safranin O staining and immunohistochemistry. The effects of 29-kDa FN-f on XT-1 expression and its relevant signaling pathway were analyzed by quantitative real-time-PCR, immunoblotting, chromatin immunoprecipitation, and immunoprecipitation assays. The receptors for 29-kDa FN-f were investigated using small interference RNA and blocking antibodies. RESULTS: The expression of XT-1 was significantly lower in human osteoarthritis cartilage than in normal cartilage. Intra-articular injection of 29-kDa FN-f reduced proteoglycan levels and XT-1 expression in murine cartilage. In addition, in 29-kDa FN-f-treated cells, XT-1 expression was significantly suppressed at both the mRNA and protein levels, modulated by the transcription factors specificity protein 1 (Sp1), Sp3, and activator protein 1 (AP-1). The 29-kDa FN-f suppressed the binding of Sp1 to the promoter region of XT-1 and enhanced the binding of Sp3 and AP-1. Inhibition of mitogen-activated protein kinase and nuclear factor kappa B signaling pathways restored the 29-kDa FN-f-inhibited XT-1 expression, along with the altered expression of Sp1 and Sp3. Blockading toll-like receptor 2 (TLR-2) and integrin α5ß1 via siRNA and blocking antibodies revealed that the effects of 29-kDa FN-f on XT-1 expression were mediated through the TLR-2 and integrin α5ß1 signaling pathways. CONCLUSION: These results demonstrate that 29-kDa FN-f negatively affects cartilage anabolism by regulating glycosaminoglycan formation through XT-1.


Subject(s)
Cartilage, Articular/enzymology , Chondrocytes/enzymology , Enzyme Inhibitors/pharmacology , Fibronectins/pharmacology , Osteoarthritis, Knee/enzymology , Pentosyltransferases/antagonists & inhibitors , Aged , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/drug effects , Down-Regulation/drug effects , Female , Humans , Male , Mice, Inbred C57BL , Middle Aged , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , Pentosyltransferases/biosynthesis , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Peptide Fragments/pharmacology , RNA, Messenger/genetics , Signal Transduction/drug effects , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription, Genetic , UDP Xylose-Protein Xylosyltransferase
11.
Orthop Surg ; 11(1): 117-125, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30560591

ABSTRACT

OBJECTIVE: To investigate the association between curcumin and the differentially expressed genes (DEG) in synovial tissues of osteoarthritis. METHODS: Microarray analysis was used to screen for the DEG in osteoarthritis synovial cells. Curcumin-related genes were identified through the drug-gene interaction network STITCH (http://stitch.embl.de/cgi/input.pl). Expression levels of fibronectin 1 (FN1) and collagen III protein were measured by western blot. MTT assay was used to examine the effects of different concentrations of curcumin on cell viability. Western blot and quantitative real-time polymerase chain reaction were used to validate the different expression levels of matrix metalloproteinase-3 (MMP3). Clone formation assay, flow cytometry, and the TUNEL method were conducted for detecting the cell proliferation and apoptosis rate. RESULTS: In the two chips of GSE1919 and GSE55235, the average expression of MMP3 in the osteoarthritis group was 63.7% and 12.9% higher than that of the healthy control, respectively. The results of western blot also showed that the average expression of MMP3 in 30 osteoarthritis patients was 132% higher than that of the healthy group, which confirmed that MMP3 was highly expressed in osteoarthritis group. The results of MTT showed that at 72 h, the cell viability of 40 µmol/L curcumin was the lowest and 79.6% lower than for the 0 µmol/L group, so the final curcumin concentration of 40 µmol/L was selected for subsequent experiments. Western blot results further showed that the expression of MMP3 was 44% lower in the untreated groups compared with the curcumin group, and the expressions of FN1 and collagen III were increased by 112% and 84%, respectively, which indicated that curcumin inhibited MMP3 expression and decreased osteoarthritis synovial cell activity. Cloning formation experiments showed that cell numbers increased by 75% and 20.5% in untreated and curcumin groups, and compared with the untreated group, the cells in the curcumin group decreased by 30.8%. Flow cytometry showed that the apoptotic rate in the curcumin group increased by 85.1% compared with the untreated group, but for a single group, MMP3 decreased the apoptotic rate by 53.9% and 46.7%, respectively. CONCLUSIONS: MMP3 was highly expressed in osteoarthritis synovial cells. Curcumin could reduce cell viability, inhibit cell proliferation, increase cell apoptosis, and eventually alleviate inflammation of osteoarthritis by inhibiting the expression of MMP3.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Curcumin/pharmacology , Matrix Metalloproteinase 3/biosynthesis , Osteoarthritis, Knee/pathology , Synovial Membrane/drug effects , Apoptosis/drug effects , Case-Control Studies , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Humans , Matrix Metalloproteinase 3/genetics , Osteoarthritis, Knee/enzymology , Synovial Membrane/enzymology , Synovial Membrane/pathology
12.
PLoS One ; 13(9): e0203944, 2018.
Article in English | MEDLINE | ID: mdl-30222787

ABSTRACT

Recent studies have shown that superoxide dismutase 1 (SOD1), SOD2, and SOD3 are significantly decreased in human osteoarthritic cartilage. SOD activity is a marker that can be used to comprehensively evaluate the enzymatic capacities of SOD1, SOD2, and SOD3; however, the trend of SOD activity in end-stage osteoarthritic tissues remains unknown. In the present study, we found that SOD activity in end-stage osteoarthritic synovium of the knee was significantly lower than that in control synovium without the influence of age. The SOD activity was significantly lower in the end-stage knee osteoarthritic cartilage than in the control, but a weak negative correlation was observed between aging and SOD activity. However, SOD activity in end-stage hip osteoarthritic cartilage was significantly lower than that in control cartilage without the influence of aging. The relationship between osteoarthritis and SOD activity was stronger than the relationship between aging and SOD activity. These results indicate that direct regulation of SOD activity in joint tissues may lead to suppression of osteoarthritis progression.


Subject(s)
Cartilage, Articular/enzymology , Osteoarthritis, Hip/enzymology , Osteoarthritis, Knee/enzymology , Superoxide Dismutase/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Aging/metabolism , Case-Control Studies , Female , Humans , Male , Malondialdehyde/metabolism , Middle Aged , Reactive Oxygen Species/metabolism , Synovial Membrane/enzymology , Young Adult
13.
Biomed Pharmacother ; 106: 1169-1174, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30119184

ABSTRACT

Osteoarthritis (OA) is a degenerative joint disease that is commonly accompanied by inflammation. Scoparone is a biologically active constituent isolated from Artemisia capillaris and possesses anti-inflammatory activity. However, the effect of scoparone on inflammatory response in OA has not been authenticated. The aim of this study was to evaluate the role of scoparone in OA in vitro. Our results showed that IL-1ß treatment significantly inhibited the cell viability of chondrocytes, whereas the inhibition effect was attenuated by scoparone in a dose-dependent manner. IL-1ß also efficiently induced the production of nitric oxide (NO), prostaglandin E2 (PGE2), MMP-3, MMP-13, ADAMTS-4 and ADAMTS-5 in chondrocytes. However, scoparone dose-dependently suppressed the induction. In addition, scoparone repressed IL-1ß-induced the expression of iNOS and COX-2 in chondrocytes. Furthermore, the activation of the PI3K/Akt/NF-κB pathway induced by IL-1ß was diminished by scoparone treatment. Taken together, these findings indicated that scoparone inhibited the expression of inflammatory mediators in IL-1ß-induced chondrocytes via regulating the PI3K/Akt/NF-κB pathway. Thus, scoparone may be used as a new therapeutic agent for the treatment of OA.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chondrocytes/drug effects , Coumarins/pharmacology , Inflammation Mediators/metabolism , Interleukin-1beta/pharmacology , NF-kappa B/metabolism , Osteoarthritis, Knee/drug therapy , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , ADAMTS Proteins/metabolism , Cell Survival/drug effects , Cells, Cultured , Chondrocytes/enzymology , Chondrocytes/pathology , Dose-Response Relationship, Drug , Humans , Matrix Metalloproteinases, Secreted/metabolism , Middle Aged , Osteoarthritis, Knee/enzymology , Osteoarthritis, Knee/pathology , Phosphorylation , Signal Transduction/drug effects
14.
J Lipid Res ; 59(9): 1763-1770, 2018 09.
Article in English | MEDLINE | ID: mdl-29986999

ABSTRACT

Omega-6 FAs are inflammatory mediators that are increased in joints with osteoarthritis (OA), but their association with OA progression is not yet well defined. To investigate the relationship between omega-6 FAs and knee OA, we measured with LC-MS the levels of 22 omega-6 lipids (arachidonic acid, linoleic acid, and 20 oxylipins) in synovial fluid (SF) from 112 knees of 102 individuals (58 with knee OA; 44 controls). We hypothesized that oxylipin metabolites would increase in OA knee SF and with radiographically progressive disease. We validated results by comparing samples from affected and unaffected knees in 10 individuals with unilateral OA. In adjusted analysis, SF levels of three omega-6 oxylipins [prostaglandin D2, 11,12-dihydroxyeicosatrienoic acid (DHET), and 14,15-DHET] were associated with OA. Of these, 11,12-DHET and 14,15-DHET were higher in affected versus unaffected knees of people with unilateral disease (P < 0.014 and P < 0.003, respectively). Levels of these and 8,9-DHET were also associated with radiographic progression over 3.3 years in 87 individuals. Circulating levels of all three were associated with gene variants at the soluble epoxide hydrolase enzyme. Lipidomic profiling in SF identified an additional inflammatory pathway associated with knee OA and radiographic progression.


Subject(s)
Epoxide Hydrolases/chemistry , Epoxide Hydrolases/metabolism , Osteoarthritis, Knee/metabolism , Oxylipins/chemistry , Oxylipins/metabolism , Aged , Case-Control Studies , Disease Progression , Epoxide Hydrolases/genetics , Female , Humans , Male , Osteoarthritis, Knee/diagnostic imaging , Osteoarthritis, Knee/enzymology , Osteoarthritis, Knee/genetics , Polymorphism, Single Nucleotide , Solubility , Tomography, X-Ray Computed
15.
J Biol Chem ; 293(31): 12259-12270, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29929979

ABSTRACT

Certain dysregulated chondrocyte metabolic adaptive responses such as decreased activity of the master regulator of energy metabolism AMP-activated protein kinase (AMPK) promote osteoarthritis (OA). Metabolism intersects with epigenetic and transcriptional responses. Hence, we studied chondrocyte ATP-citrate lyase (ACLY), which generates acetyl-CoA from mitochondrial-derived citrate, and modulates acetylation of histones and transcription factors. We assessed ACLY in normal and OA human knee chondrocytes and cartilages by Western blotting and immunohistochemistry, and quantified acetyl-CoA fluorometrically. We examined histone and transcription factor lysine acetylation by Western blotting, and assessed histone H3K9 and H3K27 occupancy of iNOS, MMP3, and MMP13 promoters by chromatin immunoprecipitation (ChIP) and quantitative PCR (qPCR). We analyzed iNOS, MMP3, MMP13, aggrecan (ACAN), and Col2a1 gene expression by RT-qPCR. Glucose availability regulated ACLY expression and function, nucleocytosolic acetyl-CoA, and histone acetylation. Human knee OA chondrocytes exhibited increased ACLY activation (assessed by Ser-455 phosphorylation), associated with increased H3K9 and H3K27 acetylation. Inhibition of ACLY attenuated IL-1ß-induced transcription of iNOS, MMP3, and MMP13 by suppressing acetylation of p65 NF-κB, H3K9, and H3K27, blunted release of NO, MMP3, and MMP13, and also reduced SOX9 acetylation that promoted SOX9 nuclear translocation, leading to increased aggrecan and Col2a1 mRNA expression. ACLY is a novel player involved in regulation of cartilage matrix metabolism. Increased ACLY activity in OA chondrocytes increased nucleocytosolic acetyl-CoA, leading to increased matrix catabolism via dysregulated histone and transcription factor acetylation. Pharmacologic ACLY inhibition in OA chondrocytes globally reverses these changes and stimulates matrix gene expression and AMPK activation, supporting translational investigation in OA.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Cartilage, Articular/enzymology , Chondrocytes/enzymology , Extracellular Matrix/enzymology , Osteoarthritis, Knee/enzymology , ATP Citrate (pro-S)-Lyase/genetics , Acetyl Coenzyme A/metabolism , Acetylation , Aggrecans/genetics , Aggrecans/metabolism , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Histones/metabolism , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/metabolism
16.
J Orthop Res ; 36(10): 2728-2735, 2018 10.
Article in English | MEDLINE | ID: mdl-29761560

ABSTRACT

Proper control of protein synthesis is vital for tissue homeostasis and its deregulation is characteristic of many disorders including osteoarthritis (OA). The objectives of this work were to analyze and correlate changes in activity of the translation apparatus associated with cartilage degeneration in an animal model of OA. Osteoarthritis was induced surgically in rats by anterior cruciate ligament transection (ACLT). Using a modified Mankin scoring system and analysis of protein expression we demonstrated, that mechanistic target of rapamycin complex 1 (mTORC1)-mediated 4E-BP1 phosphorylation was detected significantly earlier than other mTORC1-mediated modifications, such as p70S6K and ULK1 phosphorylation. 4E-BP1 is an inhibitor of cap-dependent translation those functions are inhibited by mTORC1 mediated phosphorylation. This signaling event not only preceded prominent signs of cartilage degeneration but also the increase in global protein synthesis rate. These results suggest that abnormal mTORC1 activity is one of the primary dysregulations observed in OA cartilage. Importantly, it is distributed disproportionately between targets, with 4E-BP1 being phosphorylated earlier than other downstream targets. Thus, our work provides new insights into the sequence of molecular events leading to cartilage destruction in OA and identifies translational control as an important regulatory hub involved in initiating OA. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:2728-2735, 2018.


Subject(s)
Carrier Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Osteoarthritis, Knee/enzymology , Phosphoproteins/metabolism , Animals , Anterior Cruciate Ligament Injuries/complications , Disease Progression , Extracellular Matrix/metabolism , Intracellular Signaling Peptides and Proteins , Male , Osteoarthritis, Knee/etiology , Protein Biosynthesis , Rats, Sprague-Dawley
17.
Mol Med Rep ; 18(1): 541-549, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29749508

ABSTRACT

The aim of the present study was to investigate the role of microRNA (miR)­27a­3p in osteoarthritis (OA). Reverse transcription­quantitative polymerase chain reaction and western blotting were performed to determine the expression of miR­27a­3p and aggrecanase­2 (ADAMTS5) in cartilage tissues from patients with OA and healthy controls, and also in interleukin (IL)­1ß­treated primary human chondrocytes. Primary human chondrocytes were transfected with miR­27a­3p. A luciferase reporter assay was used to validate the direct contact between miR­27a­3p and its putative binding site in the 3'­untranslated region ADAMTS5 mRNA. Furthermore, the effects of IL­1ß­induced activation of mitogen­activated protein kinase (MAPK) and nuclear factor (NF)­κB on miR­27a­3p were evaluated using specific inhibitors. The results revealed that the level of miR­27a­3p was reduced in OA cartilage tissues compared with those of normal controls. In addition, decreased miR­27a­3p and increased ADAMTS5 expression was observed in a time­ and dose­dependent manner in chondrocytes treated with IL­1ß. Furthermore, overexpression of miR­27a­3p suppressed the expression of ADAMTS5 in human chondrocytes induced by IL­1ß. miR­27a­3p overexpression also decreased the luciferase activity of the wild­type ADAMTS5 reporter plasmid. Mutation of the miR­27a­3p binding site in the 3'­untranslated region of ADAMTS5 mRNA abolished the miR­27a­3p­mediated repression of reporter activity. Furthermore, the use of specific inhibitors demonstrated that IL­1ß may regulate miR­27a­3p expression via NF­κB and MAPK signaling pathways in chondrocytes. The present study concluded that miR­27a­3p was downregulated in human OA and was suppressed by IL­1ß, and functions as a crucial regulator of ADAMTS5 in OA chondrocytes. In addition, IL­1ß­mediated suppression of miR­27a­3p activity may occur via the MAPK and NF­κB pathways. The present study may provide a novel strategy for clinical treatment of OA caused by upregulation of miR­27a­3p.


Subject(s)
Cartilage, Articular/metabolism , Interleukin-1beta/metabolism , MicroRNAs/genetics , Osteoarthritis, Knee/metabolism , Signal Transduction , ADAMTS5 Protein/genetics , Adolescent , Adult , Aged , Cartilage, Articular/enzymology , Cells, Cultured , Child , Female , Gene Expression Regulation , Humans , Male , MicroRNAs/metabolism , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteoarthritis, Knee/enzymology , Osteoarthritis, Knee/etiology
18.
Arthritis Rheumatol ; 70(7): 1030-1041, 2018 07.
Article in English | MEDLINE | ID: mdl-29457374

ABSTRACT

OBJECTIVE: Osteoarthritis (OA) chondrocytes exhibit impairment of autophagy, one arm of the proteostasis network that coordinates proteome and organelle quality control and degradation. Deficient proteostasis impacts differentiation and viability, and inflammatory processes in aging and disease. The present study was undertaken to assess ubiquitin proteasome system proteasomal function in OA chondrocytes. METHODS: We evaluated human knee cartilage by immunohistochemistry, and assessed proteasomal function, levels of proteasomal core subunits and chaperones, and autophagy in cultured chondrocytes. Assays included Western blotting, quantitative reverse transcription-polymerase chain reaction, proteasomal protease activity assessment, and cell immunofluorescence analysis. RESULTS: Human knee OA cartilage exhibited polyubiquitin accumulation, with increased ubiquitin K48-linked polyubiquitinated proteins in situ, suggesting proteasomal impairment. Cultured OA chondrocytes demonstrated accumulation of K48 polyubiquitinated proteins, significantly reduced 20S proteasome core protease activity, and decreased levels of phosphorylated FOXO4 and proteasome 26S subunit, non-ATPase 11 (PSMD11), a FOXO4-inducible promoter of proteasomal activation. Levels of proteasome subunit ß type 3 (PSMB3), PSMB5, PSMB6, and proteasome assembly chaperone 1 were not decreased in OA chondrocytes. In normal chondrocytes, PSMD11 small interfering RNA knockdown stimulated certain autophagy machinery elements, increased extracellular nitric oxide (NO) levels, and reduced chondrocytic master transcription factor SOX9 protein and messenger RNA (mRNA) and aggrecan (AGC1) mRNA. PSMD11 gain-of- function by transfection increased proteasomal function, increased levels of SOX9-induced AGC1 mRNA, stimulated elements of the autophagic machinery, and inhibited extracellular levels of interleukin-1-induced NO and matrix metalloproteinase 13 in OA chondrocytes. CONCLUSION: Deficient PSMD11, associated with reduced phosphorylated FOXO4, promotes impaired proteasomal function in OA chondrocytes, dysregulation of chondrocytic homeostasis, and decreased levels of SOX9 mRNA, SOX9 protein, and AGC1 mRNA. Chondrocyte proteasomal impairment may be a therapeutic target for OA.


Subject(s)
Aggrecans/metabolism , Chondrocytes/metabolism , Osteoarthritis, Knee/enzymology , Proteasome Endopeptidase Complex/physiology , SOX9 Transcription Factor/metabolism , Cartilage, Articular/cytology , Cell Culture Techniques , Humans , Knee Joint/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA, Messenger/metabolism
19.
J Bone Miner Res ; 33(5): 945-958, 2018 05.
Article in English | MEDLINE | ID: mdl-29314205

ABSTRACT

Cdc42, a member of Rho family small guanosine triphosphatases (GTPases), is critical for cartilage development. We investigated the roles of Cdc42 in osteoarthritis and explored the potential mechanism underlying Cdc42-mediated articular cartilage degeneration and subchondral bone deterioration. Cdc42 is highly expressed in both articular cartilage and subchondral bone in a mouse osteoarthritis model with surgical destabilization of the medial meniscus (DMM) in the knee joints. Specifically, genetic disruption of Cdc42, knockdown of Cdc42 expression, or inhibition of Cdc42 activity robustly attenuates the DMM-induced destruction, hypertrophy, high expression of matrix metallopeptidase-13 and collagen X, and activation of Stat3 in articular cartilages. Notably, genetic disruption of Cdc42, knockdown of Cdc42 expression or inhibition of Cdc42 activity significantly restored the increased numbers of mesenchymal stem cells, osteoprogenitors, osteoblasts, osteoclasts, and neovascularized vessels, the increased bone mass, and the activated Erk1/2, Smad1/5 and Smad2 in subchondral bone of DMM-operated mice. Mechanistically, Cdc42 mediates interleukin-1ß-induced interleukin-6 production and subsequent Jak/Stat3 activation to regulate chondrocytic inflammation, and also lies upstream of Erk/Smads to regulate subchondral bone remodeling during transform growth factor-ß1 signaling. Cdc42 is apparently required for both articular cartilage degeneration and subchondral bone deterioration of osteoarthritis, thus, interventions targeting Cdc42 have potential in osteoarthritic therapy. © 2018 American Society for Bone and Mineral Research.


Subject(s)
Bone and Bones/enzymology , Cartilage, Articular/enzymology , Knee Joint/enzymology , Osteoarthritis, Knee/enzymology , cdc42 GTP-Binding Protein/biosynthesis , Animals , Bone and Bones/pathology , Cartilage, Articular/pathology , Collagen Type X/genetics , Collagen Type X/metabolism , Gene Expression Regulation, Enzymologic , Interleukin-6/genetics , Interleukin-6/metabolism , Knee Joint/pathology , Male , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , Osteoblasts/enzymology , Osteoblasts/pathology , Osteoclasts/enzymology , Osteoclasts/pathology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Smad Proteins/genetics , Smad Proteins/metabolism , Transforming Growth Factor beta1 , cdc42 GTP-Binding Protein/genetics
20.
DNA Cell Biol ; 37(3): 271-277, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29356574

ABSTRACT

Pyruvate kinase M2 (PKM2) has been wildly verified to modulate glycolysis in tumor cells. However, the role of PKM2 on the glycolysis of osteoarthritis (OA) chondrocytes is still unclear. In present study, we investigate the function of PKM2 on OA chondrocyte glycolysis and the collagen matrix generation in vitro. Results showed that PKM2 was upregulated in OA chondrocytes compared with healthy control chondrocytes. In OA chondrocytes, ATP expression was lower compared with healthy control chondrocytes. Loss-of-function experiment showed that PKM2 knockdown mediated by lentivirus transfection could significantly suppress the glucose consumption and lactate secretion levels and decrease glucose transporter-1 (Glut-1), lactate dehydrogenase A (LDHA), and hypoxia inducible factor 1-alpha (HIF-1α), indicating the inhibition of PKM2 knockdown on glycolysis. Moreover, Cell Counting Kit-8 (CCK-8), flow cytometry, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay showed that PKM2 knockdown inhibited OA chondrocyte proliferation and promoted the apoptosis. Western blot and immunocytochemical staining showed that PKM2 knockdown downregulated the expression levels of COL2A1 and SOX-9. In summary, our results conclude that PKM2 modulates the glycolysis and extracellular matrix generation, providing the vital role of PKM2 on OA pathogenesis and a novel therapeutic target for OA.


Subject(s)
Chondrocytes/enzymology , Extracellular Matrix/enzymology , Glycolysis , Osteoarthritis, Knee/enzymology , Pyruvate Kinase/physiology , Adult , Cell Proliferation , Cells, Cultured , Enzyme Induction , Humans , Middle Aged , Osteoarthritis, Knee/pathology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...