Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 704
Filter
1.
Acta Biomater ; 181: 297-307, 2024 06.
Article in English | MEDLINE | ID: mdl-38710401

ABSTRACT

The pericellular matrix (PCM) serves a critical role in signal transduction and mechanoprotection in chondrocytes. Osteoarthritis (OA) leads to a gradual deterioration of the cartilage, marked by a shift in the spatial arrangement of chondrocytes from initially isolated strands to large cell clusters in end-stage degeneration. These changes coincide with progressive enzymatic breakdown of the PCM. This study aims to assess the role and involvement of specific matrix metalloproteinases (MMPs) in PCM degradation during OA. We selected cartilage samples from 148 OA patients based on the predominant spatial chondrocyte patterns. The presence of various MMPs (-1,-2,-3,-7,-8,-9,-10,-12,-13) was identified by multiplexed immunoassays. For each pattern and identified MMP, the levels and activation states (pro-form vs. active form) were measured by zymograms and western blots. The localization of these MMPs was determined using immunohistochemical labeling. To verify these results, healthy cartilage was exposed to purified MMPs, and the consecutive structural integrity of the PCM was analyzed through immunolabeling and proximity ligation assay. Screening showed elevated levels of MMP-1,-2,-3,-7, and -13, with their expression profile showing a clear dependency of the degeneration stage. MMP-2 and -7 were localized in the PCM, whereas MMP-1,-7, and -13 were predominantly intracellular. We found that MMP-2 and -3 directly disrupt collagen type VI, and MMP-3 and -7 destroy perlecan. MMP-2, -3, and -7 emerge as central players in early PCM degradation in OA. With the disease's initial stages already displaying elevated peaks in MMP expression, this insight may guide early targeted therapies to halt abnormal PCM remodeling. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) causes a gradual deterioration of the articular cartilage, accompanied by a progressive breakdown of the pericellular matrix (PCM). The PCM's crucial function in protecting and transmitting signals within chondrocytes is impaired in OA. By studying 148 OA-patient cartilage samples, the involvement of matrix metalloproteinases (MMPs) in PCM breakdown was explored. Findings highlighted elevated levels of certain MMPs linked to different stages of degeneration. Notably, MMP-2, -3, and -7 were identified as potent contributors to early PCM degradation, disrupting key components like collagen type VI and perlecan. Understanding these MMPs' roles in initiating OA progression, especially in its early stages, provides insights into potential targets for interventions to preserve PCM integrity and potentially impeding OA advancement.


Subject(s)
Extracellular Matrix , Matrix Metalloproteinases , Osteoarthritis , Proteolysis , Humans , Matrix Metalloproteinases/metabolism , Osteoarthritis/pathology , Osteoarthritis/metabolism , Osteoarthritis/enzymology , Extracellular Matrix/metabolism , Male , Female , Middle Aged , Aged , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrocytes/enzymology , Cartilage, Articular/pathology , Cartilage, Articular/metabolism
2.
Haemophilia ; 28(1): 145-150, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34697874

ABSTRACT

INTRODUCTION: Studying the pathological changes of ligaments in patients with haemophilic arthritis (HA) has important significance for guiding the release of ligaments during total knee arthroplasty (TKA) and exploring interventions to prevent ligament lesions. AIM: This study was conducted to show the pathological changes and investigate the lysine oxidase (LOX) and matrix metalloproteinase (MMP)-1, -2, and -3 levels in the ligaments of patients with HA compared with those of patients with osteoarthritis (OA). METHODS: Ligaments obtained during the TKA were stained with Masson trichrome, Verhoeff-Van Gieson and haematoxylin and eosin to show the basic pathological changes. Collagen I, elastin, LOXs and MMP-1, -2, and -3 expression levels were detected via western blot. LOX and MMP-1, -2, and -3 mRNA expression levels were analysed via quantitative real-time PCR. RESULTS: Compared with OA ligaments, HA ligaments were constructed more loosely with wider gaps, more breaks, haemocytodeposition and local hypertrophy among the fibres. LOXs and MMP mRNA expression levels were upregulated in the HA tissues, which was consistent with the western blot results. Collagen I and elastin levels were also higher in patients with HA. CONCLUSIONS: The metabolism of the ligaments in patients with HA is more complex than in those with OA, and the ligaments of patients with HA have stronger healing and destruction processes. This pathology is related to iron overload and imbalanced inflammatory factors due to repeated intra-articular bleeding.


Subject(s)
Ligaments/enzymology , Osteoarthritis , Humans , Matrix Metalloproteinase 1 , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 3 , Osteoarthritis/enzymology , Protein-Lysine 6-Oxidase
3.
J Med Chem ; 65(2): 1567-1584, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34931844

ABSTRACT

The serine/threonine kinase SGK1 is an activator of the ß-catenin pathway and a powerful stimulator of cartilage degradation that is found to be upregulated under genomic control in diseased osteoarthritic cartilage. Today, no oral disease-modifying treatments are available and chronic treatment in this indication sets high requirements for the drug selectivity, pharmacokinetic, and safety profile. We describe the identification of a highly selective druglike 1H-pyrazolo[3,4-d]pyrimidine SGK1 inhibitor 17a that matches both safety and pharmacokinetic requirements for oral dosing. Rational compound design was facilitated by a novel hSGK1 co-crystal structure, and multiple ligand-based computer models were applied to guide the chemical optimization of the compound ADMET and selectivity profiles. Compounds were selected for subchronic proof of mechanism studies in the mouse femoral head cartilage explant model, and compound 17a emerged as a druglike SGK1 inhibitor, with a highly optimized profile suitable for oral dosing as a novel, potentially disease-modifying agent for osteoarthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Disease Models, Animal , Immediate-Early Proteins/antagonists & inhibitors , Microsomes, Liver/drug effects , Osteoarthritis/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/chemistry , Animals , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Ligands , Male , Mice , Mice, Inbred C57BL , Osteoarthritis/enzymology , Osteoarthritis/pathology , Protein Kinase Inhibitors/chemistry , Rats , Rats, Sprague-Dawley
4.
Sci Rep ; 11(1): 23120, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34848799

ABSTRACT

The pathophysiology of osteoarthritis (OA) includes the destruction of subchondral bone tissue and inflammation of the synovium. Thus, an effective disease-modifying treatment should act on both of these pathogenetic components. It is known that cSrc kinase is involved in bone and cartilage remodeling, and SYK kinase is associated with the inflammatory component. Thus the aim of this study was to characterize the mechanism of action and efficacy of a small molecule multikinase inhibitor MT-SYK-03 targeting SYK and cSrc kinases among others in different in vitro and in vivo arthritis models. The selectivity of MT-SYK-03 kinase inhibition was assayed on a panel of 341 kinases. The compound was evaluated in a set of in vitro models of OA and in vivo OA and RA models: surgically-induced arthritis (SIA), monosodium iodoacetate-induced arthritis (MIA), collagen-induced arthritis (CIA), adjuvant-induced arthritis (AIA). MT-SYK-03 inhibited cSrc and SYK with IC50 of 14.2 and 23 nM respectively. Only five kinases were inhibited > 90% at 500 nM of MT-SYK-03. In in vitro OA models MT-SYK-03 reduced hypertrophic changes of chondrocytes, bone resorption, and inhibited SYK-mediated inflammatory signaling. MT-SYK-03 showed preferential distribution to joint and bone tissue (in rats) and revealed disease-modifying activity in vivo by halving the depth of cartilage erosion in rat SIA model, and increasing the pain threshold in rat MIA model. Chondroprotective and antiresorptive effects were shown in a monotherapy regime and in combination with methotrexate (MTX) in murine and rat CIA models; an immune-mediated inflammation in rat AIA model was decreased. The obtained preclinical data support inhibition of cSrc and SYK as a viable strategy for disease-modifying treatment of OA. A Phase 2 clinical study of MT-SYK-03 is to be started.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/enzymology , Bone and Bones/drug effects , CSK Tyrosine-Protein Kinase/antagonists & inhibitors , Cartilage/drug effects , Osteoarthritis/drug therapy , Osteoarthritis/enzymology , Syk Kinase/antagonists & inhibitors , Animals , Arthritis, Experimental/pathology , Bone Resorption/pathology , Chondrocytes/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Humans , Inflammation , Inhibitory Concentration 50 , Iodoacetic Acid/pharmacology , Lipopolysaccharide Receptors/biosynthesis , Male , Mice , Monocytes/cytology , Protective Agents/pharmacology , Rabbits , Rats , Rats, Sprague-Dawley , Rats, Wistar , Synovial Membrane/pathology
5.
Sci Rep ; 11(1): 17020, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34426599

ABSTRACT

Temporomandibular joint OA (TMJOA) is a common degenerative joint disease, leads to structural damage and ultimately loss of function. Matrix degradation is one of the first pathogenesis during the progression of OA, it was effective to inhibit matrix degradation to block the development of OA. In this study, an in vivo model (compressive mechanical force) and an in vitro model (IL-1ß) were used to induce OA-like changes in TMJ cartilage and chondrocytes. We revealed lysyl oxidase like-2 (LOXL2) play a critical role in TMJOA. LOXL2 expression decreased in mechanical stress/IL-ß induced TMJOA-like lesions in both in vivo models and in vitro models. Furthermore, recombinant LOXL2 (rhLOXL2) treatment ameliorated the degenerative changes induced by mechanical stress in vivo, including the thinning cartilage, down-expression of collagen II and proteoglycan, and over-expression of TNF-a, while LOXL2 antibody (anti-LOXL2) treatment exacerbated these changes. Mechanistically, the protection of LOXL2 in chondrocytes was induced partly through activation of the Integrin/FAK pathway. The inhibition of the Integrin/FAK pathway could neutralized the effects caused by rhLOXL2. Collectively, our study suggests that the LOXL2 plays a protective role in mechanical stress induced TMJOA-like changes, and the Integrin/FAK pathway may be a key downstream pathway in this process.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrins/metabolism , Osteoarthritis/enzymology , Signal Transduction , Animals , Biomechanical Phenomena , Cartilage/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Collagen Type II/metabolism , Down-Regulation , Extracellular Matrix/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , Male , Mandible/pathology , Rats, Sprague-Dawley , Stress, Mechanical , Temporomandibular Joint/pathology , Tumor Necrosis Factor-alpha/metabolism
6.
Clin Appl Thromb Hemost ; 27: 1076029621995569, 2021.
Article in English | MEDLINE | ID: mdl-33754883

ABSTRACT

Osteoarthritis (OA) is a chronic condition marked by joint pain, inflammation and loss of articular cartilage, that can be treated with total joint arthroplasty (TJA) at end stages. TJA is marked by post-operative inflammation, which directly effects levels of cartilage degradation biomarkers, proteoglycan-4 (PRG4) and matrix metalloproteinase-9 (MMP-9). PRG4 is a protective glycoprotein that is decreased in individuals with OA. MMP-9 is a matrix metalloproteinase that contributes to articular cartilage loss and is elevated in OA patients. It is upregulated by pro-inflammatory markers, such as IL-1, IL-6 and CRP. This study aims to elucidate the immediate post-operative changes in levels of PRG4, MMP-9, IL-6, CRP, and WBC in patients undergoing TJA to clarify the role of inflammation in recovery after surgery and in the overall pathogenesis of OA. Blood was collected at 3 time points (day 0, day 1 post-operatively, and days 5-7 post-operatively), from 63 patients undergoing TJA due to OA, and levels of these biomarkers were quantified. IL-6, CRP, WBC and MMP-9 were lowest at day 0, highest at day 1, and stabilized at an intermediate level at days 5-7. Meanwhile, PRG4 followed the opposite trend. These studies suggest that IL-6, CRP and WBC showed predictable fluctuations, with pro-inflammatory biomarkers upregulating MMP-9 and downregulating PRG4. Measuring these biomarkers may help expose the role of inflammation in the post-surgical recovery of TJA patients and in long-term pathogenesis of OA. These levels may help risk stratify patients pre-operatively and help develop individualized post-surgical plans.


Subject(s)
Arthroplasty, Replacement, Hip , Inflammation/metabolism , Matrix Metalloproteinase 9/metabolism , Osteoarthritis/metabolism , Osteoarthritis/surgery , Proteoglycans/metabolism , Female , Humans , Inflammation/enzymology , Male , Osteoarthritis/enzymology
7.
Sci Rep ; 11(1): 5682, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33707504

ABSTRACT

WNT ligands can activate several signalling cascades of pivotal importance during development and regenerative processes. Their de-regulation has been associated with the onset of different diseases. Here we investigated the role of the WNT/Calcium Calmodulin Kinase II (CaMKII) pathway in osteoarthritis. We identified Heme Oxygenase I (HMOX1) and Sox-9 as specific markers of the WNT/CaMKII signalling in articular chondrocytes through a microarray analysis. We showed that the expression of the activated form of CaMKII, phospho-CaMKII, was increased in human and murine osteoarthritis and the expression of HMOX1 was accordingly reduced, demonstrating the activation of the pathway during disease progression. To elucidate its function, we administered the CaMKII inhibitor KN93 to mice in which osteoarthritis was induced by resection of the anterior horn of the medial meniscus and of the medial collateral ligament in the knee joint. Pharmacological blockade of CaMKII exacerbated cartilage damage and bone remodelling. Finally, we showed that CaMKII inhibition in articular chondrocytes upregulated the expression of matrix remodelling enzymes alone and in combination with Interleukin 1. These results suggest an important homeostatic role of the WNT/CaMKII signalling in osteoarthritis which could be exploited in the future for therapeutic purposes.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cartilage, Articular/enzymology , Cartilage, Articular/pathology , Homeostasis , Osteoarthritis/enzymology , Osteoarthritis/pathology , Aged , Animals , Bone Remodeling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cattle , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , Female , Gene Expression Regulation, Enzymologic , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Interleukin-1beta/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Models, Biological , Osteoarthritis/genetics , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcriptome/genetics , Up-Regulation , Wnt3 Protein/metabolism
8.
Biomed Pharmacother ; 135: 111199, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33401221

ABSTRACT

Osteoarthritis (OA) is a heterogeneous disease that is consistently difficult to treat due to the complexity of the regulatory network involved in OA pathogenesis, especially in terms of cartilage degeneration. As a C-2 epimer of glucose, d-mannose can alleviate bone loss and repress immunopathology by upregulating regulatory T cells; however, the role of d-mannose in OA-related cartilage degeneration remains unknown. In this study, we investigated the chondroprotective effect of d-mannose in vitro and in vivo on OA. We found that incubating interleukin (IL)-1ß-treated rat chondrocytes with d-mannose restrained OA degeneration by elevating cell proliferation, strongly activating autophagy, reducing apoptosis, and downregulating catabolism. Additionally, oral gavage administration of d-mannose to monosodium iodoacetate (MIA)-treated rats revealed that a median (1.25 g/kg/day) rather than high or low dose of d-mannose suppressed OA progression and attenuated OA development based on lower macroscopic scores for cartilage, decreased histological scores for cartilage and synovium, strongly activated autophagy, and downregulated catabolism. In terms of a downstream mechanism, we showed that d-mannose might attenuate OA degeneration by activating autophagy in IL-1ß-treated rat chondrocytes by promoting the phosphorylation of 5' AMP-activated protein kinase (AMPK). Our in vitro findings revealed that d-mannose delayed IL-1ß-induced OA degeneration in rat chondrocytes by enhancing autophagy activation through the AMPK pathway. Furthermore, the in vivo results indicated that a median dose of d-mannose suppressed MIA-induced OA development. These results suggested that d-mannose exhibits chondroprotective effects and represents a potential disease-modifying drug and novel therapeutic agent for OA.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/drug effects , Chondrocytes/drug effects , Interleukin-1beta/toxicity , Joints/drug effects , Mannose/pharmacology , Osteoarthritis/prevention & control , Animals , Antirheumatic Agents/pharmacology , Apoptosis/drug effects , Cells, Cultured , Chondrocytes/enzymology , Chondrocytes/pathology , Disease Models, Animal , Iodoacetic Acid , Joints/enzymology , Joints/pathology , Male , Osteoarthritis/chemically induced , Osteoarthritis/enzymology , Osteoarthritis/pathology , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction
9.
Biomed Pharmacother ; 133: 111089, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33378983

ABSTRACT

Osteoarthritis is a chronic, systemic and inflammatory disease. However, the pathogenesis and understanding of RA are still limited. Ubiquitin-specific protease 13 (USP13) belongs to the deubiquitinating enzyme (DUB) superfamily, and has been implicated in various cellular events. Nevertheless, its potential on RA progression has little to be investigated. In the present study, we found that USP13 expression was markedly up-regulated in synovial tissue samples from patients with RA, and was down-regulated in human fibroblast-like synoviocytes (H-FLSs) stimulated by interleukin-1ß (IL-1ß), tumor necrosis factor alpha (TNF-α) or lipopolysaccharide (LPS). We then showed that over-expressing USP13 markedly suppressed inflammatory response, oxidative stress and apoptosis in H-FLSs upon IL-1ß or TNF-α challenge, whereas USP13 knockdown exhibited detrimental effects. In addition, USP13-induced protective effects were associated with the improvement of nuclear factor erythroid 2-related factor 2 (Nrf-2) and the repression of Casapse-3. Furthermore, phosphatase and tensin homolog (PTEN) expression was greatly improved by USP13 in H-FLSs upon IL-1ß or TNF-α treatment, whereas phosphorylated AKT expression was diminished. In response to IL-1ß or TNF-α exposure, nuclear transcription factor κB (NF-κB) signaling pathway was activated, whereas being significantly restrained in H-FLSs over-expressing USP13. Mechanistically, USP13 directly interacted with PTEN. Of note, we found that USP13-regulated cellular processes including inflammation, oxidative stress and apoptotic cell death were partly dependent on AKT activation. Furthermore, USP13 over-expression effectively inhibited osteoclastogenesis and osteoclast-associated gene expression. The in vivo experiments finally confirmed that USP13 dramatically repressed synovial hyperplasia, inflammatory cell infiltration, cartilage damage and bone loss in collagen-induced arthritis (CIA) mice via the same molecular mechanisms detected in vitro. Taken together, these findings suggested that targeting USP13 may provide feasible therapies for RA.


Subject(s)
Apoptosis , Arthritis, Experimental/prevention & control , Bone Remodeling , Endopeptidases/metabolism , Joints/enzymology , Osteoarthritis/prevention & control , Oxidative Stress , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ubiquitin-Specific Proteases/metabolism , Aged , Animals , Arthritis, Experimental/enzymology , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Cells, Cultured , Collagen Type II , Endopeptidases/genetics , Humans , Hyperplasia , Joints/pathology , Male , Mice, Inbred C57BL , Mice, Inbred DBA , Middle Aged , Osteoarthritis/enzymology , Osteoarthritis/genetics , Osteoarthritis/pathology , Osteoclasts/enzymology , Osteoclasts/pathology , Osteogenesis , PTEN Phosphohydrolase/genetics , Signal Transduction , Synoviocytes/enzymology , Synoviocytes/pathology , Ubiquitin-Specific Proteases/genetics
10.
Int Immunopharmacol ; 91: 107191, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33359852

ABSTRACT

This study aims to investigate the effects and mechanisms of parathyroid hormone [1-34] (PTH1-34) on TNF-α-stimulated mice chondrocytes, as well as cartilage from a meniscus injury induced osteoarthritis (MIO) mice model. The C57BL/6J mice received medial meniscectomy, and then administrated with PTH1-34. The results showed that PTH1-34 administration decreased secondary allodynia and the pain-related transcripts. The IHC, ELISA, Micro-CT imaging and histopathology analysis revealed the significantly improved subchondral plate thickness and bone porosity, the reduced pro-inflammatory cytokines in serum and joint fluid. In vitro, mice chondrocyte was treated with TNF-α or co-cultured with synovial cells. The results showed that TNF-α markedly upregulated the MMP13 expression, and the ERK1/2, NF-κB or PI3K signaling pathway inhibitors could reverse the induction effect of TNF-α on expression of MMP13 in chondrocytes. PTH1-34 alone has no effect on the expression of MMP13 and NF-κB signaling pathways, but the PTH1-34 could reverse the induction effect of TNF-α on MMP13 expression and NF-κB signaling pathway activation in chondrocytes. In addition, PTH1-34 administration inhibited the expression of TNF-α and MMP13, and chondrocyte viability, while the PKA repressor reversed the effect of PTH1-34 in chondrocytes co-cultured with synovial cells. In conclusion, PTH1-34 has an obvious analgesic and anti-inflammatory effect, inhibits the matrix synthesis and alleviates the progression of osteoarthritis. In vitro, PTH1-34 inhibited TNF-α expression and antagonized TNF-α-induced MMP13 expression via the PKA pathway and the NF-κB signaling pathways, respectively.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Arthralgia/prevention & control , Chondrocytes/drug effects , Joints/drug effects , Matrix Metalloproteinase 13/metabolism , Meniscus/drug effects , Osteoarthritis/prevention & control , Teriparatide/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Arthralgia/enzymology , Arthralgia/etiology , Cells, Cultured , Chondrocytes/enzymology , Chondrocytes/pathology , Coculture Techniques , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Joints/enzymology , Joints/pathology , Meniscectomy , Meniscus/enzymology , Meniscus/pathology , Meniscus/surgery , Mice, Inbred C57BL , NF-kappa B/metabolism , Osteoarthritis/enzymology , Osteoarthritis/etiology , Osteoarthritis/pathology , Signal Transduction , Synovial Membrane/drug effects , Synovial Membrane/enzymology , Synovial Membrane/pathology
11.
J Cell Physiol ; 236(3): 2205-2213, 2021 03.
Article in English | MEDLINE | ID: mdl-32808296

ABSTRACT

Osteoarthritis (OA) is a progressive degenerative joint disorder characterized by synovial inflammation. Interleukin-6 (IL-6) is a key proinflammatory cytokine in OA progression. Particulate matter 2.5 (PM2.5) exposure increases the risk of different diseases, including OA. Up until now, no studies have described any association between PM2.5 and IL-6 expression in human OA synovial fibroblasts (OASFs). Here, our data show that PM2.5 concentration- and time-dependently promoted IL-6 synthesis in human OASFs. We also found that reactive oxygen species (ROS) generation potentiated the effects of PM2.5 on IL-6 production. ASK1, ERK, p38, and JNK inhibitors reduced PM2.5-induced increases of IL-6 expression. Treatment of OASFs with PM2.5 promoted phosphorylation of these signaling cascades. We also found that PM2.5 enhanced c-Jun phosphorylation and its translocation into the nucleus. Thus, PM2.5 increases IL-6 production in human OASFs via the ROS, ASK1, ERK, p38, JNK, and AP-1 signaling pathways. Our evidence links PM2.5 with OA progression.


Subject(s)
Fibroblasts/pathology , Interleukin-6/biosynthesis , MAP Kinase Kinase Kinase 5/metabolism , Osteoarthritis/enzymology , Osteoarthritis/pathology , Particulate Matter/toxicity , Synovial Membrane/pathology , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System/drug effects , Models, Biological , Reactive Oxygen Species/metabolism , Transcription Factor AP-1/metabolism , Up-Regulation/drug effects
12.
Int J Mol Sci ; 21(24)2020 Dec 12.
Article in English | MEDLINE | ID: mdl-33322825

ABSTRACT

This article provides a brief review of the pathophysiology of osteoarthritis and the ontogeny of chondrocytes and details how physical exercise improves the health of osteoarthritic joints and enhances the potential of autologous chondrocyte implants, matrix-induced autologous chondrocyte implants, and mesenchymal stem cell implants for the successful treatment of damaged articular cartilage and subchondral bone. In response to exercise, articular chondrocytes increase their production of glycosaminoglycans, bone morphogenic proteins, and anti-inflammatory cytokines and decrease their production of proinflammatory cytokines and matrix-degrading metalloproteinases. These changes are associated with improvements in cartilage organization and reductions in cartilage degeneration. Studies in humans indicate that exercise enhances joint recruitment of bone marrow-derived mesenchymal stem cells and upregulates their expression of osteogenic and chondrogenic genes, osteogenic microRNAs, and osteogenic growth factors. Rodent experiments demonstrate that exercise enhances the osteogenic potential of bone marrow-derived mesenchymal stem cells while diminishing their adipogenic potential, and that exercise done after stem cell implantation may benefit stem cell transplant viability. Physical exercise also exerts a beneficial effect on the skeletal system by decreasing immune cell production of osteoclastogenic cytokines interleukin-1ß, tumor necrosis factor-α, and interferon-γ, while increasing their production of antiosteoclastogenic cytokines interleukin-10 and transforming growth factor-ß. In conclusion, physical exercise done both by bone marrow-derived mesenchymal stem cell donors and recipients and by autologous chondrocyte donor recipients may improve the outcome of osteochondral regeneration therapy and improve skeletal health by downregulating osteoclastogenic cytokine production and upregulating antiosteoclastogenic cytokine production by circulating immune cells.


Subject(s)
Chondrocytes/metabolism , Exercise/physiology , Mesenchymal Stem Cells/metabolism , Osteoarthritis/physiopathology , Osteogenesis , Physical Conditioning, Animal/physiology , Regeneration/genetics , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/enzymology , Cartilage, Articular/pathology , Cytokines/metabolism , Glycosaminoglycans/metabolism , Humans , Metalloproteases/metabolism , Osteoarthritis/enzymology , Osteoarthritis/immunology , Osteoarthritis/therapy , Osteogenesis/genetics , Osteogenesis/immunology , Osteogenesis/physiology , Regeneration/immunology , Regeneration/physiology , Stem Cell Transplantation
13.
Aging (Albany NY) ; 12(20): 20163-20183, 2020 10 25.
Article in English | MEDLINE | ID: mdl-33099538

ABSTRACT

Osteoarthritis (OA) is one of the most painful and widespread chronic degenerative joint diseases and is characterized by destructed articular cartilage and inflamed joints. Previously, our findings indicated that circular RNA ciRS-7 (ciRS-7)/microRNA 7 (miR-7) axis is abnormally expressed in OA, and regulates proliferation, inflammatory responses, and apoptosis of interleukin-1ß (IL-1ß)-stimulated chondrocytes. However, its underlying role in OA remains unknown. In this study, we first validated cartilage degradation and defection of autophagy in samples of OA patients. IL-1ß initially stimulated autophagy of chondrocytes, and ultimately significantly suppressed autophagy. Upregulated ciRS-7/down-regulated miR-7 aggravated IL-1ß-induced cartilage degradation, and restrained autophagy in vitro. Gene sequencing and bioinformatics analysis performed on a control group, IL-1ß group, and IL-1ß+miR-7-mimics group demonstrated that seven of the most significant mRNA candidates were enriched in the interleukin-17 (IL-17) signaling pathway. Increased IL-17A levels were also observed by qRT-PCR and ELISA. In addition, it was revealed that the ciRS-7/miR-7 axis ameliorated cartilage degradation and defection of autophagy by PI3K/AKT/mTOR activation in IL-1ß-induced chondrocytes. Furthermore, an OA model was established in rats with medial meniscus destabilization. miR-7-siRNA-expressing lentiviruses alleviated surgical resection-induced cartilage destruction of OA mice, whereas miR-7 mimics worsened the effects. Thus, these findings revealed that the mechanism of the ciRS-7/miR-7 axis involved regulating OA progression and provided valuable directions for OA treatment.


Subject(s)
Autophagy , Cartilage, Articular/enzymology , Chondrocytes/enzymology , Interleukin-17/metabolism , MicroRNAs/metabolism , Osteoarthritis/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy/drug effects , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Case-Control Studies , Cell Line , Chondrocytes/drug effects , Chondrocytes/pathology , Disease Models, Animal , Gene Expression Regulation , Humans , Interleukin-17/genetics , Interleukin-1beta/pharmacology , Male , MicroRNAs/genetics , Osteoarthritis/genetics , Osteoarthritis/pathology , RNA, Long Noncoding/genetics , Rats, Sprague-Dawley , Signal Transduction , Transcriptome
14.
Int Immunopharmacol ; 89(Pt B): 107079, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33096361

ABSTRACT

Osteoarthritis (OA) is a progressive and degenerative joint disease. Aloin is a bitter and yellow-brown-coloured compound from the Aloe plant and is allowed for use in foods as a "natural flavour". In our study, we examined the protective effects of Aloin on the inhibition of OA development as well as its underlying mechanism in both in vitro and vivo experiments. In in-vitro experiments, the protective effect of aloin on the anabolism and catabolism of the extracellular matrix (ECM) induced by IL-1 ß in chondrocytes by inhibiting the expression of pro-inflammatory factors, including TNF-α (p = 0.016), IL-6 (p = 0.006), iNOS (p = 0.001) and COX-2 (p = 0.006). Mechanistically, Aloin suppressed the IL-1ß-induced activation of the PI3K/Akt/NF-κB signalling pathway cascades. Moreover, molecular docking studies demonstrated that Aloin bound strongly to PI3K. In vivo, Aloin ameliorated the OA process in the destabilization of the medial meniscus (DMM) model. In summary, our findings demonstrate that Aloin ameliorates the progression of OA via the PI3K/Akt/NF-κB signalling pathways, which supports Aloin as a promising therapeutic agent for the treatment of OA.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chondrocytes/drug effects , Emodin/analogs & derivatives , Joints/drug effects , NF-kappa B/metabolism , Osteoarthritis/prevention & control , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cells, Cultured , Chondrocytes/enzymology , Chondrocytes/pathology , Disease Models, Animal , Emodin/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Interleukin-1beta/pharmacology , Joints/enzymology , Joints/pathology , Male , Mice, Inbred C57BL , Molecular Docking Simulation , Osteoarthritis/enzymology , Osteoarthritis/pathology , Phosphorylation , Signal Transduction
15.
Int J Mol Sci ; 21(14)2020 Jul 13.
Article in English | MEDLINE | ID: mdl-32668590

ABSTRACT

Osteoarthritis (OA) is the most common type of arthritis that occurs in an aged population. It affects any joints in the body and degenerates the articular cartilage and the subchondral bone. Despite the pathophysiology of OA being different, cartilage resorption is still a symbol of osteoarthritis. Matrix metalloproteinases (MMPs) are important proteolytic enzymes that degrade extra-cellular matrix proteins (ECM) in the body. MMPs contribute to the turnover of cartilage and its break down; their levels have increased in the joint tissues of OA patients. Application of chondroprotective drugs neutralize the activities of MMPs. Natural products derived from herbs and plants developed as traditional medicine have been paid attention to, due to their potential biological effects. The therapeutic value of natural products in OA has increased in reputation due to their clinical impact and insignificant side effects. Several MMPs inhibitor have been used as therapeutic drugs, for a long time. Recently, different types of compounds were reviewed for their biological activities. In this review, we summarize numerous natural products for the development of MMPs inhibitors in arthritic diseases and describe the major signaling targets that were involved for the treatments of these destructive joint diseases.


Subject(s)
Biological Products/therapeutic use , Matrix Metalloproteinase Inhibitors/therapeutic use , Osteoarthritis/drug therapy , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/enzymology , Cartilage, Articular/pathology , Chondrocytes/drug effects , Chondrocytes/enzymology , Cytokines/physiology , Drug Evaluation, Preclinical , Extracellular Matrix Proteins/metabolism , Forecasting , Humans , Iodoacetic Acid/toxicity , Models, Animal , NF-kappa B/metabolism , Osteoarthritis/chemically induced , Osteoarthritis/enzymology , Osteoarthritis/pathology , Rats , Self Medication , Tetradecanoylphorbol Acetate/toxicity
16.
J Ethnopharmacol ; 261: 113074, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-32534115

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Kyung-Bang Gumiganghwal-tang tablet (GMGHT) is a standardized Korean Medicine that could treat a cold, headache, arthralgia and fever. Although GMGHT has been used for arthritis-related diseases including a sprain, arthralgia, unspecified arthritis and knee arthritis, there is no pre-clinical evidence to treat osteoarthritis (OA). This study determined the drug dosage and the mechanisms of GMGHT for OA. METHODS: OA was induced by intra-articular monoiodoacetic acid (MIA) injection in Sprague-Dawley rats. As calculated from the human equivalent dose formula, GMGHT was orally administered at the doses of 9.86, 98.6 and 986 mg/kg for 4 weeks. The arthritis score was performed by a blind test, and histological changes in articular cartilage were indicated by hematoxylin and eosin, Safranin O and toluidine blue staining. SW1353 chondrocytes were stimulated by interleukin (IL)-1ß recombinant to analyze the expressions of Type II collagen, matrix metalloproteinases (MMPs) and nuclear factor (NF)-κB. RESULTS: Rough and punctate surfaces of the femoral condyle induced by MIA, were recovered by the GMGHT treatment. The arthritis score was significantly improved in the 968 mg/kg of GMGHT-treated cartilage. Loss of chondrocytes and proteoglycan were ameliorated at the deep zone of the subchondral bone plate by the GMGHT administration in OA rats. The expression of Type II collagen was increased, while MMP-1, -3 and -13 levels were decreased in the GMGHT-treated SW1353 chondrocytes. In addition, the GMGHT treatment regulated NF-κB activation along with IL-6, transforming growth factor-ß and IL-12 production. CONCLUSIONS: GMGHT promoted the recovery of articular cartilage damage by inhibiting MMPs, accompanied with its anti-inflammatory effects in OA. GMGHT might be an alternative therapeutic treatment for OA.


Subject(s)
Arthritis, Experimental/prevention & control , Cartilage, Articular/drug effects , Joints/drug effects , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinases, Secreted/antagonists & inhibitors , Osteoarthritis/prevention & control , Plant Extracts/pharmacology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/enzymology , Arthritis, Experimental/pathology , Cartilage, Articular/enzymology , Cartilage, Articular/pathology , Cell Line, Tumor , Chondrocytes/drug effects , Chondrocytes/enzymology , Chondrocytes/pathology , Collagen Type II/metabolism , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Iodoacetic Acid , Joints/enzymology , Joints/pathology , Male , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinases, Secreted/genetics , Matrix Metalloproteinases, Secreted/metabolism , Osteoarthritis/chemically induced , Osteoarthritis/enzymology , Osteoarthritis/pathology , Rats, Sprague-Dawley
17.
Biosci Rep ; 40(5)2020 05 29.
Article in English | MEDLINE | ID: mdl-32426819

ABSTRACT

This research aimed to explore the role of period circadian clock 2 (Per2) in the evolution of osteoarthritis (OA) and the relevant mechanisms. Per2 messenger RNA (mRNA) and protein levels were markedly reduced in NHAC-kn cells treated with 5 µg/ml lipopolysaccharide (LPS) for 12 h. Then, pcDNA3.1-Per2 and si-Per2 were recruited to boost and reduce the expression of Per2, respectively. MTT assay, apoptosis analysis and enzyme-linked immunosorbent assay (ELISA) results showed that Per2 increased cell proliferation, while inhibited apoptosis and inflammation. Furthermore, the PTEN/PI3K/Akt signalling pathway was activated by Per2 overexpression; the CO-IP data confirmed that Per2 specifically bound to PTEN. Through employing IGF-1, a PI3K activator, we determined that Per2-mediated inflammation response in LPS-stimulated NHAC-kn cells through the PTEN/PI3K/Akt signalling pathway. In summary, the present study indicates that Per2 may serve as a novel therapeutic target through activating the PTEN/PI3K/Akt signalling pathway.


Subject(s)
Chondrocytes/enzymology , Osteoarthritis/enzymology , PTEN Phosphohydrolase/metabolism , Period Circadian Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Apoptosis , Cell Line , Cell Proliferation , Chondrocytes/drug effects , Chondrocytes/pathology , Cytokines/genetics , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Osteoarthritis/genetics , Osteoarthritis/pathology , Period Circadian Proteins/genetics , Signal Transduction
18.
Biosci Rep ; 40(6)2020 06 26.
Article in English | MEDLINE | ID: mdl-32432314

ABSTRACT

PURPOSE: To explore the regulatory mechanism of miR-137 and transcription factor 4 (TCF4) in the progression of osteoarthritis (OA). PATIENTS AND METHODS: The expressions of miR-137 and TCF4 were detected in OA cartilage tissue, chondrocytes and OA rat cartilage tissue. miR-137 and TCF4 were up-regulated or down-regulated and transfected into chondrocytes and OA rat cartilage tissue. The gene expression, protein level, cell proliferation, apoptosis and inflammatory factors were detected, respectively. LPS and anterior cruciate ligament transection (ACLT) on the right knee were used to induce chondrocyte inflammation and establish rat OA model, respectively. RESULTS: miR-137 was low expressed in cartilage tissue of OA group, while TCF4 expression and protein level were significantly higher, showing significant negative correlation. In LPS group, chondrocyte activity was significantly inhibited, cell apoptosis ability was significantly enhanced, and the levels of inflammatory factors TNF-α, IL-1ß, IL-6 were significantly increased. However, the above results were significantly improved after the up-regulation of miR-137 or down-regulation of TCF4. Double luciferase report revealed that miR-137 and TCF4 had targeted relationship. LPS induced activation of AMPK/NF-κB pathway and higher level of apoptosis. AMPK/NF-κB pathway inhibitor C could inhibit activation of this pathway, and up-regulation of miR-137 or down-regulation of TCF4 could significantly weaken the regulation of LPS on the pathway and apoptosis. Analysis of OA rat model showed that over-expression of miR-137 could inhibit up-regulation of inflammatory factors and activation of AMPK/NF-κB pathway. CONCLUSION: miR-137 targets the inhibition of TCF4 to reverse the progression of OA through the AMPK/NF-κB signaling pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Chondrocytes/enzymology , MicroRNAs/metabolism , NF-kappa B/metabolism , Osteoarthritis/enzymology , Transcription Factor 4/metabolism , Animals , Apoptosis , Case-Control Studies , Cell Proliferation , Cells, Cultured , Chondrocytes/pathology , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Humans , Male , MicroRNAs/genetics , Osteoarthritis/genetics , Osteoarthritis/pathology , Rats, Sprague-Dawley , Signal Transduction , Transcription Factor 4/genetics
19.
J Orthop Res ; 38(10): 2104-2112, 2020 10.
Article in English | MEDLINE | ID: mdl-32285964

ABSTRACT

Reactive oxygen species (ROS) generated by the NADPH oxidase (Nox) enzymes are important short-range signaling molecules. They have been extensively studied in the physiology and pathophysiology of the cardiovascular system, where they have important roles in vascular inflammation, angiogenesis, hypertension, cardiac injury, stroke, and aging. Increasing evidence demonstrates that ROS and Nox enzymes also affect bone homeostasis and osteoporosis, and more recent studies implicate ROS and Nox enzymes in both inflammatory arthritis and osteoarthritis. Mechanistically, this connection may be through the effects of ROS on signal transduction. ROS affect both transforming growth factor-ß/Smad signaling, interleukin-1ß/nuclear factor-kappa B signaling, and the resulting changes in matrix metalloproteinase expression. The purpose of this review is to describe the role of Nox enzymes in the physiology and pathobiology of bone and joints and to highlight the potential of therapeutically targeting the Nox enzymes.


Subject(s)
Bone and Bones/enzymology , Cartilage, Articular/enzymology , NADPH Oxidases/metabolism , Osteoarthritis/enzymology , Animals , Homeostasis , Humans , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/classification
20.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 34(3): 362-366, 2020 Mar 15.
Article in Chinese | MEDLINE | ID: mdl-32174084

ABSTRACT

OBJECTIVE: To study the expressions of Renin, angiotensin converting enzyme (ACE), angiotensin receptor 1 (AT1R), and AT2R in synovial tissue of osteoarthritis (OA) at different stages. METHODS: The patients who were treated with upper knee amputation because of trauma or total knee arthroplasty for OA between January 2018 and December 2018 were enrolled. Among them, 32 patients who met the selection criteria were included in the study. According to the Kellgren-Lawrence (K-L) X-ray classification, they were allocated to normal synovial group (group A, n=9), moderate OA synovial group (group B, n=11, K-L level 3), and advanced OA synovial group (group C, n=12, K-L level 4). The relative expressions of Renin, ACE, AT1R, and AT2R mRNAs and proteins were detected by real-time fluorescence quantitative PCR (qRT-PCR) and Western blot. RESULTS: The relative expressions of Renin, ACE, and AT1R mRNAs and proteins were significantly higher in group B and group C than in group A ( P<0.05). The relative expressions of ACE and AT1R mRNAs and proteins and Renin protein were significantly higher in group C than in group B ( P<0.05). However, the relative expressions of AT2R mRNA and protein were lower in group B and group C than in group A ( P<0.05), and in group C than in group B ( P<0.05). CONCLUSION: The expressions of Renin, ACE, and AT1R in synovial tissue of osteoarthritis significantly increase as the K-L level increased, and the expression of AT2R decreases. Renin, ACE, AT1R, and AT2R have a certain degree of correlation with the development of OA.


Subject(s)
Osteoarthritis/enzymology , Peptidyl-Dipeptidase A/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Renin/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...