Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 530
Filter
Add more filters

Publication year range
1.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 73-77, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836679

ABSTRACT

GABBR1 receptors have been implicated in the progression of rheumatoid arthritis (RA), and p38 MAP kinase (MAPK) was shown to be downregulated by GABA and result in unchecked production of pro-inflammatory cytokine. GABBR1 is a member of GABA receptors, and it is known to be upregulated and plays a vital role in RA. Glucocorticoids are efficient therapeutics in rheumatoid arthritis (RA) and are known to regulate GABA actions; therefore, we intended to investigate the potential of glucocorticoids in RA concerning the potential pathway GABBR1/MAPK. Joint specimens were obtained from collagen-induced arthritis mouse model. A double-blind semi-quantitative analysis of vascularity, cell infiltration, as well as lining thickness by help of a 4-point scale setting was used to assess joint inflammation. Expression of GABBR1 and p38 was evaluated immunohistochemically. In vitro peripheral blood (PB), synovial fluid (SF), and mononuclear cells (MCs) were acquired from RA mice. Western blotting was used for detecting expression of GABBR1 and p38 proteins. The presence of high levels of GABBR1 and p38 was prevalent in RA joints relative to healthy joints and related to the inflammation level. Glucocorticoid treatment alters GABBR1 along with p38 protein expression in joints while reducing joint inflammation. Ex vivo and in vitro assays revealed glucocorticoids have a direct impact on p38, such as the decreased GABBR1 expression level after dexamethasone incubation with SFMC. GABBR1 together with p38 expression in RA joints depends on local inflammation and can be targeted by glucocorticoids.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Glucocorticoids , p38 Mitogen-Activated Protein Kinases , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Animals , Glucocorticoids/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Mice , Male , Joints/pathology , Joints/drug effects , Joints/metabolism , Mice, Inbred DBA , Synovial Fluid/metabolism , Synovial Fluid/drug effects , Cellular Microenvironment/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects , Disease Models, Animal
2.
Int J Mol Sci ; 23(5)2022 Feb 22.
Article in English | MEDLINE | ID: mdl-35269552

ABSTRACT

(1) Background: Synovial fluid (SF) from knee joints with osteoarthritis (OA) has increased levels of phospholipids (PL). We have reported earlier that TGF-ß and IGF-1 stimulate fibroblast-like synoviocytes (FLS) to synthesize increased amounts of PLs. The current study examined whether IL-1ß induces the release of PLs in FLS and the underlying mechanism. (2) Methods: Cultured human OA FLS were treated with IL-1ß alone and with pathway inhibitors or with synthetic liver X receptor (LXR) agonists. Cholesterol hydroxylases, ABC transporters, apolipoproteins (APO), LXR, sterol regulatory binding proteins (SREBPs), and 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) were analyzed by RT-PCR, Western blot, and ELISA. The release of radiolabeled PLs from FLS was determined, and statistical analysis was performed using R (N = 5-9). (3) Results: Like synthetic LXR agonists, IL-1ß induced a 1.4-fold greater release of PLs from FLS. Simultaneously, IL-1ß upregulated the level of the PL transporter ABCA1 and of cholesterol hydroxylases CH25H and CYP7B1. IL-1ß and T0901317 stimulated the expression of SREBP1c, whereas only T0901317 enhanced SREBP2, HMGCR, APOE, LXRα, and ABCG1 additionally. (4) Conclusions: IL-1ß partially controls PL levels in OA-SF by affecting the release of PLs from FLS. Our data show that IL-1ß upregulates cholesterol hydroxylases and thus the formation of oxysterols, which, as natural agonists of LXR, increase the level of active ABCA1, in turn enhancing the release of PLs.


Subject(s)
Benzoates/pharmacology , Benzylamines/pharmacology , Interleukin-1beta/pharmacology , Osteoarthritis/metabolism , Phospholipids/metabolism , Synoviocytes/cytology , ATP Binding Cassette Transporter 1/genetics , Cells, Cultured , Cytochrome P450 Family 7/genetics , Gene Expression Regulation/drug effects , Humans , Liver X Receptors/genetics , Osteoarthritis/genetics , Steroid Hydroxylases/genetics , Synovial Fluid/cytology , Synovial Fluid/drug effects , Synovial Fluid/metabolism , Synoviocytes/drug effects , Synoviocytes/metabolism
3.
Rheumatology (Oxford) ; 60(10): 4899-4909, 2021 10 02.
Article in English | MEDLINE | ID: mdl-33493345

ABSTRACT

OBJECTIVES: Direct inhibition of M1 polarization of synovial macrophages may be a useful therapeutic treatment for OA and OA-associated synovitis. Frugoside (FGS) is a cardiac glycoside compound isolated and extracted from Calotropis gigantea. Cardiac glycosides possess interesting anti-inflammatory potential. However, the corresponding activity of FGS has not been reported. Therefore, our aim was to find direct evidence of the effects of FGS on synovial macrophage M1 polarization and OA control. METHODS: Collagenase was used to establish an experimental mouse OA model (CIOA) with considerable synovitis. Then, FGS was intra-articular administered. The mRNA and protein levels of iNOS were analysed by real-time PCR and Western blotting in vitro. Immunohistochemical and immunofluorescence staining were used to measure the expression of F4/80, iNOS, Col2α1 and MMP13 in vivo. The levels of pro-inflammatory cytokines in FGS-treated M1 macrophage culture supernatants were analysed by flow cytometry. RESULTS: FGS attenuates synovial inflammation and delays the development of OA in CIOA mice. Further results demonstrate that FGS inhibits macrophage M1 polarization in vitro and in vivo, which subsequently decreases the secretion of IL-6 and TNF-α, in turn delaying cartilage and extracellular matrix (ECM) degradation and chondrocyte hypertrophy. FGS inhibits macrophage M1 polarization by partially downregulating miR-155 levels. CONCLUSION: This study demonstrates that intra-articular injection of FGS is a potential strategy for OA prevention and treatment, even at an early stage of disease progression. This is a novel function of FGS and has promising future clinical applications.


Subject(s)
Digitoxigenin/analogs & derivatives , Macrophage Activation/drug effects , Macrophages/drug effects , MicroRNAs/antagonists & inhibitors , Osteoarthritis/drug therapy , Synovial Fluid/cytology , Animals , Blotting, Western , Digitoxigenin/therapeutic use , Disease Models, Animal , Disease Progression , Flow Cytometry , Fluorescent Antibody Technique , Macrophages/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Real-Time Polymerase Chain Reaction , Synovial Fluid/drug effects
4.
Eur Cell Mater ; 41: 616-632, 2021 06 06.
Article in English | MEDLINE | ID: mdl-34091884

ABSTRACT

In vitro models aim to recapitulate the in vivo situation. To more closely mimic the knee joint environment, current in vitro models need improvements to reflect the complexity of the native tissue. High molecular weight hyaluronan (hMwt HA) is one of the most abundant bioactive macromolecules in healthy synovial fluid, while shear and dynamic compression are two joint-relevant mechanical forces. The present study aimed at investigating the concomitant effect of joint-simulating mechanical loading (JSML) and hMwt HA-supplemented culture medium on the chondrogenic differentiation of primary human bone-marrow-derived mesenchymal stem cells (hBM-MSCs). hBM-MSC chondrogenesis was investigated over 28 d at the gene expression level and total DNA, sulphated glycosaminoglycan, TGF-ß1 production and safranin O staining were evaluated. The concomitant effect of hMwt HA culture medium and JSML significantly increased cartilage-like matrix deposition and sulphated glycosaminoglycan synthesis, especially during early chondrogenesis. A stabilisation of the hBM-MSC-derived chondrocyte phenotype was observed through the reduced upregulation of the hypertrophic marker collagen X and an increase in the chondrogenic collagen type II/X ratio. A combination of JSML and hMwt HA medium better reflects the complexity of the in vivo synovial joint environment. Thus, JSML and hMwt HA medium will be two important features for joint-related culture models to more accurately predict the in vivo outcome, therefore reducing the need for animal studies. Reducing in vitro artefacts would enable a more reliable prescreening of potential cartilage repair therapies.


Subject(s)
Cell Differentiation/drug effects , Chondrogenesis/drug effects , Hyaluronic Acid/pharmacology , Mesenchymal Stem Cells/drug effects , Aged , Cartilage/drug effects , Cartilage/metabolism , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Collagen Type II/metabolism , Collagen Type X/metabolism , DNA/metabolism , Female , Humans , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Synovial Fluid/drug effects , Synovial Fluid/metabolism , Tissue Engineering/methods , Tissue Scaffolds/chemistry
5.
Int J Mol Sci ; 22(19)2021 Sep 23.
Article in English | MEDLINE | ID: mdl-34638561

ABSTRACT

Tumor necrosis factor (TNF) is a regulator of several chronic inflammatory diseases, such as rheumatoid arthritis. Although anti-TNF biologics have been used in clinic, they render several drawbacks, such as patients' progressive immunodeficiency and loss of response, high cost, and intravenous administration. In order to find new potential anti-TNF small molecule inhibitors, we employed an in silico approach, aiming to find natural products, analogs of Ampelopsin H, a compound that blocks the formation of TNF active trimer. Two out of nine commercially available compounds tested, Nepalensinol B and Miyabenol A, efficiently reduced TNF-induced cytotoxicity in L929 cells and production of chemokines in mice joints' synovial fibroblasts, while Nepalensinol B also abolished TNF-TNFR1 binding in non-toxic concentrations. The binding mode of the compounds was further investigated by molecular dynamics and free energy calculation studies, using and advancing the Enalos Asclepios pipeline. Conclusively, we propose that Nepalensinol B, characterized by the lowest free energy of binding and by a higher number of hydrogen bonds with TNF, qualifies as a potential lead compound for TNF inhibitors' drug development. Finally, the upgraded Enalos Asclepios pipeline can be used for improved identification of new therapeutics against TNF-mediated chronic inflammatory diseases, providing state-of-the-art insight on their binding mode.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Biological Products/chemistry , Biological Products/pharmacology , Drug Discovery/methods , Tumor Necrosis Factor Inhibitors/chemistry , Tumor Necrosis Factor Inhibitors/pharmacology , Animals , Binding Sites/drug effects , Cell Line , Cell Survival/drug effects , Computer Simulation , Drug Design , Fibroblasts/drug effects , Mice , Primary Cell Culture , Synovial Fluid/drug effects , Tumor Necrosis Factor-alpha/toxicity
6.
Int J Mol Sci ; 22(22)2021 Nov 22.
Article in English | MEDLINE | ID: mdl-34830460

ABSTRACT

TGF ß-activated kinase 1 (TAK1) is an important participant in inflammatory pathogenesis for diseases such as rheumatoid arthritis (RA) and gouty arthritis. The central position it occupies between the mitogen activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) pathways makes it an attractive therapeutic target. As this field has developed in recent years, several novel inhibitors have been presented as having specific activity that reduces the TAK1 function either covalently as in the case of 5Z-7-oxozeanol (5Z7O) or reversibly (NG-25). However, the mechanism through which takinib elicits its anti-inflammatory activity remains elusive. While this inhibitor shows great promise, a thorough analysis of its inhibitor function and its potential off-target effects is necessary before addressing its clinical potential or its use in inflammatory conditions. An analysis through Western blot showed an unexpected increase in IL-1ß-induced TAK1 phosphorylation-a prerequisite for and indicator of its functional potential-by takinib while simultaneously demonstrating the inhibition of the JAK/STAT pathway in human rheumatoid arthritis synovial fibroblasts (RASFs) in vitro. In THP-1 monocyte-derived macrophages, takinib again led to the lipopolysaccharide-induced phosphorylation of TAK1 without a marked inhibition of the TAK1 downstream effectors, namely, of c-Jun N-terminal kinase (JNK), phospho-c-Jun, NF-κB phospho-p65 or phospho-IκBα. Taken together, these findings indicate that takinib inhibits inflammation in these cells by targeting multiple signaling pathways, most notably the JAK/STAT pathway in human RASFs.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Benzamides/pharmacology , Benzimidazoles/pharmacology , MAP Kinase Kinase Kinases/genetics , STAT3 Transcription Factor/genetics , Synovial Fluid/drug effects , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Janus Kinases/genetics , Lactones/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Monocytes/drug effects , NF-kappa B/genetics , Resorcinols/pharmacology , Signal Transduction/drug effects , Synovial Fluid/metabolism , Synovial Membrane/drug effects
7.
Inflammopharmacology ; 29(5): 1427-1442, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34350509

ABSTRACT

BACKGROUND: Imbalance between apoptosis and autophagy in fibroblast-like synoviocytes (FLS) is one of the pathogenic mechanisms responsible for their abnormal proliferation in rheumatoid arthritis (RA). Methotrexate (MTX) demonstrated limited efficacy in amending this imbalance in fluid-derived (fd)-FLS. The active compound of black tea Theaflavin 3,3'-digallate (TF3) may be effective in restoring apoptosis-autophagy imbalance in (fd)-FLS. The combined effect of MTX + TF3 upon the same is yet to be elucidated. OBJECTIVE: To evaluate the effect of MTX + TF3 on fd-FLS to induce apoptosis and inhibit autophagy through Endoplasmic Reticulum (ER) stress-mediated pathways. METHODS: FLS from synovial fluid of 11 RA and 10 osteoarthritis patients were cultured after treatment with MTX/TF3 or a combination of MTX (125 nM) and TF3(10 µM) and the following parameters were evaluated. C-reactive protein, cytokines (TNF-α, IL-6), angiogenic markers were quantified by ELISA. fd-FLS viability was determined by MTT assay and apoptosis by flow cytometry. ER stress markers were estimated by RT-PCR (IRE1A, spliced-XBP-1) and immunoblotting (Grp78, Hsp70, CHOP, HIF-1α). Immunoblot studies were done to evaluate apoptotic (Bcl-2, Bax, Caspases) and autophagic (Beclin1, LC3b, p62) proteins. RESULTS: MTX (IC25) and TF3 (IC50) both in single doses could down-regulate the levels of pro-inflammatory and angiogenic markers. Combinatorial treatment modulated autophagosomal proteins in fd-FLS and induced apoptosis by regulating ER stress response. CONCLUSION: Disruption in homeostasis between apoptosis and autophagy in fd-FLS might be an underlying phenomenon in the progression of pathophysiology in RA. Co-administration of MTX + TF3 successfully restored the homeostasis by inducing apoptosis.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Biflavonoids/pharmacology , Catechin/analogs & derivatives , Methotrexate/pharmacology , Adult , Antirheumatic Agents/administration & dosage , Apoptosis/drug effects , Arthritis, Rheumatoid/physiopathology , Autophagy/drug effects , Biflavonoids/administration & dosage , Catechin/administration & dosage , Catechin/pharmacology , Cells, Cultured , Disease Progression , Drug Synergism , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Inhibitory Concentration 50 , Male , Methotrexate/administration & dosage , Middle Aged , Osteoarthritis/drug therapy , Osteoarthritis/physiopathology , Synovial Fluid/cytology , Synovial Fluid/drug effects , Synoviocytes/cytology , Synoviocytes/drug effects
8.
Toxicol Appl Pharmacol ; 391: 114917, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32044269

ABSTRACT

The hyperplastic phenotype of fibroblast-like synoviocytes (FLSs) plays an important role for synovitis, chronic inflammation and joint destruction in rheumatoid arthritis (RA). Interleukin 17A (IL-17A), a signature pro-inflammatory cytokine effectively influences the hyperplastic transformation of FLS cells and synovial pannus growth. IL-17A cytokine signalling participates in RA pathology by regulating an array of pro-inflammatory mediators and osteoclastogenesis. Cyanidin, a key flavonoid inhibits IL-17A/IL-17 receptor A (IL-17RA) interaction and alleviates progression and disease severity of psoriasis and asthma. However, the therapeutic efficacy of cyanidin on IL-17A cytokine signalling in RA remains unknown. In the present study, cyanidin inhibited IL-17A induced migratory and proliferative capacity of FLS cells derived from adjuvant-induced arthritis (AA) rats. Cyanidin treatment reduced IL-17A mediated reprogramming of AA-FLS cells to overexpress IL-17RA. In addition, significantly decreased expression of IL-17A dependent cyr61, IL-23, GM-CSF, and TLR3 were observed in AA-FLS cells in response to cyanidin. At the molecular level, cyanidin modulated IL-17/IL-17RA dependent JAK/STAT-3 signalling in AA-FLS cells. Importantly, cyanidin activated PIAS3 protein to suppress STAT-3 specific transcriptional activation in AA-FLS cells. Cyanidin treatment to AA rats attenuated clinical symptoms, synovial pannus growth, immune cell infiltration, and bone erosion. Cyanidin reduced serum level of IL-23 and GM-CSF and expression of Cyr 61 and TLR3 in the synovial tissue of AA rats. Notably, the level of p-STAT-3 protein was significantly decreased in the synovial tissue of AA rats treated with cyanidin. This study provides the first evidence that cyanidin can be used as IL-17/17RA signalling targeting therapeutic drug for the treatment of RA and this need to be investigated in RA patients.


Subject(s)
Anthocyanins/therapeutic use , Arthritis, Rheumatoid/drug therapy , Fibroblasts/drug effects , Interleukin-17/metabolism , Signal Transduction/drug effects , Synovial Fluid/drug effects , Synoviocytes/drug effects , Animals , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/pathology , Cell Movement/drug effects , Disease Progression , Fibroblasts/pathology , Gene Expression Regulation/drug effects , Janus Kinases/biosynthesis , Janus Kinases/genetics , Male , Rats , Rats, Wistar , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/genetics , Synovial Membrane/pathology , Synoviocytes/pathology
9.
Reumatismo ; 72(3): 125-130, 2020 Nov 19.
Article in English | MEDLINE | ID: mdl-33213125

ABSTRACT

The aim of this study was to evaluate the effect of an oral preparation containing a naturally occurring matrix of hydrolyzed collagen type II, chondroitin sulfate (CS), and hyaluronic acid (HA), and bioactive oligopeptides of natural hydrolyzed keratin (K) in patients affected by knee OA through the evaluation of synovial fluid (SF) and clinical changes before and after treatment. Thirty patients with knee OA and swollen joint were included in the study and submitted to arthrocentesis. Patients were randomized in two groups: 1) the treatment group (N.15) took a dietary supplement containing 120 mg HA, 240 mg CS and 300 mg K once a day for 4 weeks; 2) the control group (N.15) was only submitted to arthrocentesis. Patient symptoms were evaluated at the beginning and at the end of the study by the WOMAC self-assessment questionnaire, the Lequesne algofunctional index, and the VAS forms. SF changes were evaluated by measuring local inflammatory indices, cytokines IL-1ß, IL-8, IL-6, IL-10 and GM-CSF. The group of patients treated with the oral supplement showed an improvement in the clinical indices WOMAC (p<0.01), Lequesne (p=0.014) and VAS pain (p<0.01). On the contrary, no significant changes were found in the control group. The SF collected from the treated group showed a reduction of IL-8 (p=0.015), IL-6 and IL-10 levels, while no changes in cytokines were observed in the control group. This pilot study suggests that an oral administration of a preparation containing a combination of HA, CS and K can improve some clinical parameters and affect cytokine concentrations in SF in patients with knee OA.


Subject(s)
Chondroitin Sulfates/administration & dosage , Collagen Type II/administration & dosage , Hyaluronic Acid/administration & dosage , Keratins/administration & dosage , Osteoarthritis, Knee/drug therapy , Synovial Fluid/chemistry , Administration, Oral , Arthrocentesis , Drug Combinations , Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Humans , Interleukin-10/analysis , Interleukin-1beta/analysis , Interleukin-6/analysis , Interleukin-8/analysis , Middle Aged , Pilot Projects , Symptom Assessment/methods , Synovial Fluid/drug effects
10.
Med Sci Monit ; 25: 7488-7498, 2019 Oct 06.
Article in English | MEDLINE | ID: mdl-31587011

ABSTRACT

BACKGROUND Osteoarthritis (OA) is a joint disease characterized by articular cartilage degeneration and inflammation. We have previously clarified that a xanthan gum (XG) preparation exerts ameliorating effect on a rabbit OA model by regulating matrix metalloproteinase (MMP)-1 and MMP-3, which are critical proteins in the Wnt3a/ß-catenin pathway. Thus, it is reasonable to predict that the Wnt3a/ß-catenin pathway is involved in the treatment of OA with XG. MATERIAL AND METHODS The effect of XG in OA model animals were observed by hematoxylin and eosin staining (HE), Safranin O staining, and Fast Green staining. Articular cartilage degradation on the medial plateau sides was quantified using the modified Pritzker OARSI score. The levels of IL-6, TNF-alpha, and IL-1ß in synovial fluid were determined with ELISA. The protective effect of XG in rat chondrocytes was assessed by CCK8 assay. Moreover, activation of the Wnt3a/ß-catenin pathway and the expression of MMP13, ADAMTS5, aggrecan, and collagen II under the influence of XG was measured by Western blot and qRT-PCR. RESULTS Our results showed that XG reduced the OARSI score and the concentration of inflammatory cytokines in OA after intra-articular injection. XG acted on Wnt3a/ß-catenin in ATDC5 cells in a dose-dependent manner and exhibited a protective effect. XG also decreased the expression of MMP13 and ADAMTS5 and rescued the inhibition of aggrecan and collagen II expression in SNP-stimulated chondrocytes. CONCLUSIONS These results indicate that the effects of XG are related to the Wnt3a/ß-catenin pathway and XG suppresses matrix degradation by inhibiting the expression of MMPs and ADAMTS and promotes aggrecan and collagen II content in the ECM, indicating its favorable potential for use in OA therapy.


Subject(s)
Osteoarthritis/drug therapy , Polysaccharides, Bacterial/pharmacology , Wnt Signaling Pathway/drug effects , ADAMTS5 Protein/metabolism , Animals , Cartilage/drug effects , Cartilage/metabolism , Cartilage/pathology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Line , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Disease Models, Animal , Inflammation/metabolism , Male , Matrix Metalloproteinase 13/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Rabbits , Rats , Rats, Sprague-Dawley , Synovial Fluid/drug effects , Synovial Fluid/metabolism , Wnt3A Protein/metabolism , beta Catenin/metabolism
11.
BMC Musculoskelet Disord ; 20(1): 424, 2019 Sep 12.
Article in English | MEDLINE | ID: mdl-31511072

ABSTRACT

BACKGROUND: Intra-articular (IA) injection of hyaluronic acid (HA) (IA-HA) is a well-recognized treatment option for pain associated with symptomatic knee osteoarthritis (OA). IA-HA products differ in their HA content, molecular weight, cross-linking, and source of HA. These differences are assumed to affect the biocompatibility of the IA-HA products once injected inside the knee joint. METHODS: In the present study, we investigated the biocompatibility of three multiple-injection IA-HA products available in the global market. These included SUPARTZ FX™, a medium range molecular weight HA derived from rooster comb (Avian-HA); ORTHOVISC®, a high range molecular weight HA obtained through biological fermentation (Bio-HA); and SYNVISC®, a high molecular weight cross-linked hyaluronan derived from rooster comb (Avian-CL-HA). Rabbit knee joint tissues were histologically and biochemically examined after IA injection of the products. Furthermore, we compared the amounts of impurities in the IA-HA products. RESULTS: IA injection of Avian-CL-HA into rabbit knee joints induced the aggregation of inflammatory cells, infiltration of eosinophils, and an increase in the number of cells in the synovial fluid. However, these effects were not seen in the Avian-HA and Bio-HA groups. The residual protein content and the contaminant levels of bacterial endotoxins were below the limit of quantitation in all HA products. Avian-CL-HA contained relatively a large amount of (1 → 3)-ß-D-glucan, but this was below the lower limit of quantification in the other HA products. CONCLUSIONS: The present results clearly demonstrate that the biocompatibility of Avian-HA is comparable to that of Bio-HA, and they were both considered to have a favorable safety profile for the treatment of symptomatic OA of the knee. However, immunostimulatory activity was observed after injection of Avian-CL-HA: this might be a result of its unique cross-linking structure and/or the considerable amount of (1 → 3)-ß-D-glucan impurity present in the formulation.


Subject(s)
Hyaluronic Acid/analogs & derivatives , Viscosupplements/administration & dosage , Animals , Drug Contamination , Humans , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/adverse effects , Injections, Intra-Articular , Knee Joint/drug effects , Male , Materials Testing , Models, Animal , Osteoarthritis, Knee/drug therapy , Rabbits , Synovial Fluid/cytology , Synovial Fluid/drug effects , Viscosupplements/adverse effects
12.
Drug Dev Res ; 80(3): 360-367, 2019 05.
Article in English | MEDLINE | ID: mdl-30609097

ABSTRACT

Hit, Lead & Candidate Discovery This study investigated the effects of a natural phenolic compound quercetin on surgical-induced osteoarthritis (OA) in rabbits. Forty-eight New Zealand White rabbits were used to establish OA model by Hulth modified method, and subsequently randomized into untreated OA group (treatment was drinking water), celecoxib treated group (celecoxib 100 mg kg-1 by gavage), and quercetin treated group (25 mg kg-1 by gavage). Sixteen nonoperated rabbits served as the normal controls (drinking water was given). The treatment (length: 4 weeks) started on the 5th week postoperation when the OA pathological changes were manifested. Expressions of superoxide dismutase (SOD), matrix metalloproteinase-13 (MMP-13) and tissue inhibitor of metalloproteinases-1 (TIMP-1) in serum, synovial fluid, and synovial tissue were measured at 8 and 12 weeks postoperation. Pathological analysis was performed with synovial tissue section and Osteoarthritis Research Society International histopathology grading and staging scores were determined. The quercetin treated group showed higher SOD and TIMP-1 expressions but lower MMP-13 expression than untreated OA group in the serum, synovial fluid and synovial tissues (p < .05). There was no significant difference in the SOD, MMP-13 and TIMP-1 expressions between the quercetin-treated and celecoxib-treated groups. The MMP-13/TIMP-1 ratio of the quercetin treated group was significantly lower than that of the untreated OA group (p < .05). Quercetin can up-regulate SOD and TIMP-1, down-regulate MMP-13, and improve the degeneration of OA through weakening the oxidative stress responses and inhibiting the degradation of cartilage extracellular matrix.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Osteoarthritis, Knee/drug therapy , Quercetin/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Disease Models, Animal , Knee Joint/drug effects , Knee Joint/pathology , Matrix Metalloproteinase 13/blood , Matrix Metalloproteinase 13/metabolism , Osteoarthritis, Knee/blood , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Oxidative Stress/drug effects , Quercetin/pharmacology , Rabbits , Superoxide Dismutase/blood , Superoxide Dismutase/metabolism , Synovial Fluid/drug effects , Synovial Fluid/metabolism , Tissue Inhibitor of Metalloproteinase-1/blood , Tissue Inhibitor of Metalloproteinase-1/metabolism
13.
Drug Dev Res ; 80(3): 353-359, 2019 05.
Article in English | MEDLINE | ID: mdl-30663793

ABSTRACT

Preclinical Research & Development Curcumin has been shown to possess a series of beneficial effects, such as antiinflammatory, antioxidant, analgesic, and promoting healing. However, the effect and relative mechanism of curcumin on knee osteoarthritis (OA) have not been elucidated. The aim of this study is to explore the protective effect of curcumin on monosodium iodoacetate (MIA)-induced OA. Forty-eight rats were randomized into four experimental groups: control group, OA group, OA + PBS group, and OA + curcumin group, respectively. A single intraarticular injection of MIA was applied to establish the rat model of knee OA. Hematoxylin-eosin staining was used to evaluate histological changes of knee joint. The paw withdrawal threshold was collected and the expression of synovial fluid cytokine levels was measured by ELISA. The protein expression of TRL-4, MyD88, p-IκBα, NF-κB, TNF-α, IL-1ß, and IL6 was measured by western blot. Treating with curcumin can significantly reduce joint diameter and Mankin's score, and increase the paw withdrawal threshold. The expression of synovial fluid inflammatory biomarkers, IL-6, IL-1ß, and TNF-α in the OA + curcumin group were lower than that in OA and OA + PBS group. The protein expression of the TLR4 receptor was increased in the OA, OA + PBS, and OA + curcumin group compared to the control group. However, curcumin treatment can significantly decrease the expression of MyD88, p-IκBα, NF-κB, TNF-α, IL-1ß, and IL6 in OA + curcumin group. These findings may indicate that curcumin could block TLR4/NF-κB signal pathway, and reduce inflammation level to prevent knee wound in OA rats. Curcumin may be a feasible kind of medicament in the treatment of knee OA.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Curcumin/therapeutic use , Osteoarthritis, Knee/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Curcumin/pharmacology , Cytokines/immunology , Female , Iodoacetic Acid , Knee Joint/drug effects , Knee Joint/immunology , Knee Joint/pathology , Myeloid Differentiation Factor 88/immunology , NF-kappa B/immunology , Osteoarthritis, Knee/chemically induced , Osteoarthritis, Knee/immunology , Osteoarthritis, Knee/pathology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Synovial Fluid/drug effects , Synovial Fluid/immunology , Toll-Like Receptor 4/immunology
14.
J Cell Mol Med ; 22(4): 2449-2457, 2018 04.
Article in English | MEDLINE | ID: mdl-29383874

ABSTRACT

Synovial fibroblasts (SFs) play a crucial role in the inflammatory process of rheumatoid arthritis (RA). The highly activated NF-κB signal in SFs is responsible for most of the synovial inflammation associated with this disease. In this study, we have developed an SF-targeting liposomal system that encapsulates the NF-κB-blocking peptide (NBD peptide) HAP-lipo/NBD. HAP-lipo/NBDs demonstrated efficient SF-specific targeting in vitro and in vivo. Our study also showed a significant inhibitory effect of HAP-lipo/NBD on NF-κB activation, inflammatory cytokine release and SF migration capability after zymosan stimulation. Furthermore, the systemic administration of HAP-lipo/NBDs significantly inhibited synovial inflammation and improved the pathological scores of arthritis induced by zymosan. Thus, these results suggest that an SF-targeting NF-κB-blocking strategy is a potential approach for the development of alternative, targeted anti-RA therapies.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Inflammation/drug therapy , Liposomes/administration & dosage , Peptides/administration & dosage , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Fibroblasts/drug effects , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/pathology , Liposomes/chemistry , NF-kappa B/antagonists & inhibitors , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Peptides/chemistry , Peptides/genetics , Signal Transduction/drug effects , Synovial Fluid/drug effects , Zymosan/toxicity
15.
Article in English | MEDLINE | ID: mdl-30107253

ABSTRACT

Rheumatoid arthritis is a chronic inflammatory joint disease characterized by synovial proliferation and tissue destruction. Pro-inflammatory cytokines like interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) play a key role in the disease process and elevate energy expenditure, which further increases the joint pain and stiffness. The present study was undertaken to explore the anti-arthritic potential of fenugreek mucilage in adjuvant induced arthritic rats. In the present study, paw volume was measured on the 7th, 14th and 21st day. Finally, animals were anaesthetized; blood samples and tissues were collected for the assay of inflammatory enzymes like cyclooxygenase, lipoxygenase; evaluated the level of cytokines like IL-6, TNF-α, arthritic index and rheumatoid factor. Fenugreek mucilage exhibited maximum percentage of edema inhibition at a dose of 75 mg/kg on 21st day of adjuvant arthritis. The effect was higher than that of standard drug, indomethacin. The activities of inflammatory enzymes and concentration of mediators were decreased on treatment with fenugreek mucilage. Cytology of synovial fluid showed mild inflammation with normal synoviocytes (mesothelial cells) tried to bring back to normal characteristics on supplementation with fenugreek mucilage. Based on the observations, it can be suggested that fenugreek mucilage possesses promising anti-arthritic property and it can be used as a therapeutic agent for arthritis.


Subject(s)
Arthritis, Experimental/prevention & control , Arthritis, Rheumatoid/prevention & control , Plant Extracts/pharmacology , Plant Mucilage/pharmacology , Trigonella/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cytokines/metabolism , Drug Evaluation, Preclinical , Edema/metabolism , Edema/pathology , Edema/prevention & control , Female , Indomethacin/pharmacology , Inflammation Mediators/metabolism , Phytotherapy , Rats, Sprague-Dawley , Synovial Fluid/drug effects , Synovial Fluid/metabolism
16.
Clin Immunol ; 176: 1-11, 2017 03.
Article in English | MEDLINE | ID: mdl-28025136

ABSTRACT

Juvenile idiopathic arthritis (JIA) has three major onset types with widely varying clinical features. We assessed the natural killer T (NKT) cell function in patients with different JIA subtypes, and found systemic patients exhibited lower NKT cell counts, perforin and granzyme B expression, while the pauciarticular and polyarticular patients displayed higher perforin and granzyme B expression as compared with the controls. The synovial fluid had more NKT cells with higher levels of perforin, granzyme B, and tumour necrosis factor (TNF)-α than peripheral cells. The polyarticular patients that responded to etanercept had lower NKT cell counts, intracellular perforin, granzyme B and the mean fluorescence intensity of TNF-α than the patients that did not respond. Treatment with etanercept reduced the granzyme B and perforin, interferon (IFN)-γ and TNF-α expression in NKT cells in the responsive group. Therefore, a higher NKT cell function may indicate a decreased response to etanercept in polyarticular patients.


Subject(s)
Arthritis, Juvenile/drug therapy , Arthritis, Juvenile/immunology , CD3 Complex/immunology , Etanercept/therapeutic use , Natural Killer T-Cells/immunology , Cell Count/methods , Child , Child, Preschool , Female , Granzymes/immunology , Humans , Interferon-gamma/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Natural Killer T-Cells/drug effects , Perforin/immunology , Synovial Fluid/drug effects , Synovial Fluid/immunology , Tumor Necrosis Factor-alpha/immunology
17.
Osteoarthritis Cartilage ; 25(11): 1890-1899, 2017 11.
Article in English | MEDLINE | ID: mdl-28736247

ABSTRACT

OBJECTIVE: Phospholipids (PLs), together with hyaluronan and lubricin, are involved in boundary lubrication within human articular joints. Levels of lubricants in synovial fluid (SF) have been found to be associated with the health status of the joint. However, the biosynthesis and release of PLs within human joints remains poorly understood. This study contributes to our understanding of the effects of cytokines on the biosynthesis of PLs using cultured fibroblast-like synoviocytes (FLS) from human osteoarthritic knee joints. METHODS: Cultured FLS were stimulated with IL-1ß, TNFα, IL-6, or inhibitors of cell signaling pathways such as QNZ, SB203580 and SP600125 in the presence of stable isotope-labeled precursors of PLs. Lipids were extracted and quantified using electrospray ionization tandem mass spectrometry (ESI-MS/MS). RESULTS: Our analyses provide for the first time a detailed overview of PL species being synthesized by FLS. IL-1ß increased the biosynthesis of both phosphatidylethanolamine (PE) and PE-based plasmalogens. We show here that the NF-κB, p38 MAPK and JNK signaling pathways are all involved in IL-1ß-induced PL biosynthesis. IL-6 had no impact on PLs, whereas TNFα increased the biosynthesis of all PL classes. CONCLUSION: The biosynthesis of various PLs is controlled by IL-1ß and TNFα. Our detailed PL species analysis revealed that FLS can partly contribute to the elevated PL levels found in human osteoarthritis (OA) SF. IL-1ß in particular stimulates PE and PE-based plasmalogens which can act as cell-protective antioxidants. These results suggest that during OA progression, FLS undergo alterations in their PL composition to adapt to the new diseased environment.


Subject(s)
Cytokines/pharmacology , Enzyme Inhibitors/pharmacology , Interleukin-1beta/pharmacology , Osteoarthritis, Knee/metabolism , Phospholipids/biosynthesis , Synoviocytes/drug effects , Aged , Aged, 80 and over , Anthracenes/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Glycoproteins/metabolism , Humans , Hyaluronic Acid/metabolism , Imidazoles/pharmacology , Interleukin-6/pharmacology , Knee Joint/cytology , MAP Kinase Signaling System/drug effects , Male , Middle Aged , NF-kappa B/drug effects , NF-kappa B/metabolism , Pyridines/pharmacology , Signal Transduction/drug effects , Spectrometry, Mass, Electrospray Ionization , Synovial Fluid/drug effects , Synovial Fluid/metabolism , Synoviocytes/metabolism , Tandem Mass Spectrometry , Tumor Necrosis Factor-alpha/pharmacology , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
18.
BMC Musculoskelet Disord ; 18(1): 281, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28662656

ABSTRACT

BACKGROUND: This study was performed to evaluate the antioxidative and anti-inflammatory effects of vitamin E on oxidative stress in the plasma, synovial fluid, and synovial tissue of patients with knee osteoarthritis. METHODS: Seventy-two patients with late-stage knee osteoarthritis scheduled for total knee arthroplasty were randomized to take oral placebo (Group A) or 400 IU of vitamin E (Group B) once a day for 2 months before undergoing surgery. The blood levels of endpoints indicating oxidative stress or antioxidant capacity, Knee Society Score (KSS), Western Ontario and McMaster Universities Osteoarthritis Index score (WOMAC), and adverse effects were compared before and after the intervention between the two groups. At surgery, these redox endpoints and histological findings were compared between the synovial fluid and synovial tissue. RESULTS: In blood samples, the pre-intervention of oxidative stress and antioxidative capacity were not different between Group A and Group B. In post-intervention blood samples, the Malondialdehyde (Group A 1.34 ± 0.10, Group B 1.00 ± 0.09, p < 0.02), Alpha tocopherol (Group A 15.92 ± 1.08, Group B 24.65 ± 1.47, p < 0.01) and Trolox equivalent antioxidant capacity (Group A 4.22 ± 0.10, Group B 5.04 ± 0.10, 0 < 0.01) were significantly different between Group A and Group B. In synovial fluid samples, the Malondialdehyde (Group A 1.42 ± 0.12, Group B 1.06 ± 1.08, p 0.01), Alphatocopherol (Group A 4.51, Group B 7.03, p < 0.01), Trolox equivalent antioxidant capacity (Group A, 1.89 ± 0.06, Group B 2.19 ± 0.10) were significantly different between Group A and Group B. The pre-intervention WOMAC score and KSS score were not different between Group A and Group B. The post-intervention WOMAC score was significantly improved in all categories in Group B (Pain: Group A 27.26 ± 0.89, Group B 19.19 ± 1.43, p < 0.01; Stiffness: Group A 8.23 ± 0.79, Group B 5.45 ± 0.73, p 0.01; Function: Group A 94.77 ± 4.22, Group B 72.74 ± 6.55, p < 0.01). The post-intervention KSS score was significantly improved in all categories in Group B (Clinical: Group A 25.31 ± 14.33, Group B 33.52 ± 16.96, p < 0.01; Functional: Group A 41.43 ± 16.11, Group B 51.61 ± 19.60, p 0.02). Significantly fewer synovial tissue cells were stained with nitrotyrosine and hematoxylin-eosin in Group B than in Group A. There were no differences in adverse effects or surgical complications between the groups. CONCLUSION: Vitamin E is an effective antioxidant that can improve clinical symptoms and reduce oxidative stress conditions in patients with late-stage knee osteoarthritis. TRIAL REGISTRATION: This research project had been approved for registration at Thai Clinical Trials Registry (TCTR) since 2016-08-28 11:26:32 (Retrospective registered). The TCTR identification number is TCTR20160828001 .


Subject(s)
Antioxidants/therapeutic use , Osteoarthritis, Knee/drug therapy , Oxidative Stress/drug effects , Vitamin E/therapeutic use , Aged , Antioxidants/pharmacology , Blood/drug effects , Blood/metabolism , Female , Humans , Male , Synovial Fluid/drug effects , Synovial Fluid/metabolism , Vitamin E/pharmacology
19.
BMC Musculoskelet Disord ; 18(1): 283, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28668088

ABSTRACT

BACKGROUND: Recent studies have suggested that the tumor necrosis factor-α (TNF-α) pathway is a potential target for the management of osteoarthritis (OA). Transforming growth factor (TGF)-ß-activated kinase 1 (TAK1) is essential in several cytokine-mediated cascades, including the TNF-α, interleukin-1 (IL-1), and TGF-ß pathways. The role of TAK1 in synovial tissue in OA is not fully understood. Using synovial cells harvested from OA patients during surgery, we investigated whether TAK1 inhibition suppresses production of TNF-α-induced extracellular matrix degrading enzymes and expression of pain-related molecules. METHODS: Synovial tissues were harvested from ten subjects with radiographic evidence of osteoarthritis (OA) during total knee arthroplasty. Synovial cells were cultured and stimulated with control (culture media), 10 ng/mL human recombinant TNF-α, or 10 ng/mL TNF-α and 10 µM of the TAK1 inhibitor (5Z)-7-oxozeaenol for 24 h. Real-time polymerase chain reaction (PCR) analysis was used to monitor expression of mRNA of the extracellular matrix degrading enzymes matrix metalloproteinase-3 (MMP-3) and a disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type 1 motif, 4 (ADAMTS-4); and of the pain-related molecules cyclooxygenase-2 (COX-2), microsomal prostaglandin E synthase-1 (mPGES-1), and nerve growth factor (NGF). MMP-3 and NGF protein concentrations in cell supernatant were measured by enzyme-linked immunosorbent assay (ELISA). COX-2, mPGES-1 and ADAMTS-4 protein expression was also evaluated by western blotting. RESULTS: TNF-α stimulated increases in ADAMTS-4 and MMP3 mRNA (2.0-fold and 1.6-fold, respectively, p < 0.05) and protein expression (21.5-fold and 2.0-fold, respectively). Treatment with the TAK1 inihibitor (5Z)-7-oxozeaenol reduced ADAMTS-4 and MMP3 mRNA (0.5-fold and 0.6-fold, respectively) and protein expression (1.4-fold and 0.5-fold, respectively) in OA synovial cells. COX-2, mPGES-1 and NGF mRNA (11.2-fold, 3.1-fold and 2.7-fold, respectively) and protein expression (3.0-fold, 2.7-fold and 2.2-fold, respectively) were increased by TNF-α. (5Z)-7-oxozeaenol treatment reduced mPGES1 and NGF mRNA (1.5-fold and 0.8-fold, respectively) and protein (1.5-fold and 0.5-fold, respectively). CONCLUSION: TAK1 plays an important role in the regulation of TNF-α induced extracellular matrix degrading enzymes and pain-related molecule expression. TAK1 may be a potential target for therapeutic strategies aimed at preventing osteoarthritis progression and pain.


Subject(s)
Arthralgia/metabolism , Extracellular Matrix/enzymology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/physiology , Synovial Fluid/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Aged , Aged, 80 and over , Arthralgia/surgery , Arthroplasty, Replacement, Knee/trends , Cells, Cultured , Extracellular Matrix/drug effects , Female , Gene Expression , Humans , Lactones/pharmacology , Male , Middle Aged , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/surgery , Resorcinols/pharmacology , Synovial Fluid/drug effects
20.
BMC Complement Altern Med ; 17(1): 537, 2017 Dec 16.
Article in English | MEDLINE | ID: mdl-29246219

ABSTRACT

BACKGROUND: The standardized maritime pine bark extract (Pycnogenol®) has previously shown symptom alleviating effects in patients suffering from moderate forms of knee osteoarthritis (OA). The cellular mechanisms for this positive impact are so far unknown. The purpose of the present randomized pilot controlled study was to span the knowledge gap between the reported clinical effects of Pycnogenol® and its in vivo mechanism of action in OA patients. METHODS: Thirty three patients with severe OA scheduled for a knee arthroplasty either received 100 mg of Pycnogenol® twice daily or no treatment (control group) three weeks before surgery. Cartilage, synovial fluid and serum samples were collected during surgical intervention. Relative gene expression of cartilage homeostasis markers were analyzed in the patients' chondrocytes. Inflammatory and cartilage metabolism mediators were investigated in serum and synovial fluid samples. RESULTS: The oral intake of Pycnogenol® downregulated the gene expression of various cartilage degradation markers in the patients' chondrocytes, the decrease of MMP3, MMP13 and the pro-inflammatory cytokine IL1B were statistically significant (p ≤ 0.05). Additionally, protein concentrations of ADAMTS-5 in serum were reduced significantly (p ≤ 0.05) after three weeks intake of the pine bark extract. CONCLUSIONS: This is the first report about positive cellular effects of a dietary supplement on key catabolic and inflammatory markers in patients with severe OA. The results provide a rational basis for understanding previously reported clinical effects of Pycnogenol® on symptom scores of patients suffering from OA. TRIAL REGISTRATION: ISRCTN10754119 . Retrospectively registered 08/10/2015.


Subject(s)
Cartilage/drug effects , Flavonoids/pharmacology , Osteoarthritis, Knee/drug therapy , Osteoarthritis, Knee/metabolism , Synovial Fluid/drug effects , Aged , Biomarkers/analysis , Cartilage/chemistry , Collagenases/blood , Female , Flavonoids/administration & dosage , Flavonoids/therapeutic use , Humans , Interleukin-1beta/blood , Male , Middle Aged , Pilot Projects , Plant Extracts , Synovial Fluid/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL