Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Front Immunol ; 12: 762016, 2021.
Article in English | MEDLINE | ID: mdl-34777378

ABSTRACT

Gout is caused by depositing monosodium urate (MSU) crystals within the articular area. The infiltration of neutrophils and monocytes drives the initial inflammatory response followed by lymphocytes. Interestingly, emerging evidence supports the view that in situ imbalance of T helper 17 cells (Th17)/regulatory T cells (Treg) impacts the subsequent damage to target tissues. Galectin-9 (Gal-9) is a modulator of innate and adaptive immunity with both pro- and anti-inflammatory functions, dependent upon its expression and cellular location. However, the specific cellular and molecular mechanisms by which Gal-9 modulates the inflammatory response in the onset and progression of gouty arthritis has yet to be elucidated. In this study, we sought to comprehensively characterise the functional role of exogenous Gal-9 in an in vivo model of MSU crystal-induced gouty inflammation by monitoring in situ neutrophils, monocytes and Th17/Treg recruited phenotypes and related cyto-chemokines profile. Treatment with Gal-9 revealed a dose-dependent reduction in joint inflammation scores, knee joint oedema and expression of different pro-inflammatory cyto-chemokines. Furthermore, flow cytometry analysis highlighted a significant modulation of infiltrating inflammatory monocytes (CD11b+/CD115+/LY6-Chi) and Th17 (CD4+/IL-17+)/Treg (CD4+/CD25+/FOXP-3+) cells following Gal-9 treatment. Collectively the results presented in this study indicate that the administration of Gal-9 could provide a new therapeutic strategy for preventing tissue damage in gouty arthritic inflammation and, possibly, in other inflammatory-based diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Gouty/drug therapy , Galectins/therapeutic use , Animals , Ankle Joint/immunology , Arthritis, Gouty/immunology , Cells, Cultured , Cytokines/immunology , Humans , Male , Mice , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Uric Acid
2.
Inflamm Res ; 70(3): 323-341, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33559709

ABSTRACT

OBJECTIVE: Tripterine (Trip) is frequently applied to alleviate inflammation in various diseases such as rheumatoid arthritis. Macrophages have both anti-inflammatory and pro-inflammatory functions. However, whether Trip can inhibit cell inflammation in gouty arthritis (GA) remains undiscovered and whether the mechanism involved in macrophage polarization is also undetermined. This paper aims to study the effects of Trip on inflammation and macrophage polarization in GA. METHODS: Monosodium urate (MSU) crystals were used to establish GA mouse models, and bone marrow-derived macrophages (BMDMs) were induced to construct GA cell models. Pretreatments of Trip and injection of Antagomir-449a/Agomir-449a were performed on mice for 6 days. The effects of Trip and miR-449 on toe swelling, joint damage of GA mouse were examined. The alternations on cell morphology, cell proliferation marker Ki67, inflammatory cytokines, NLRP3 inflammasome, and NF-κB signaling-related proteins were also determined both in vivo and in vitro. Dual-luciferase reporter gene assay and RIP assay were adopted to estimate the targeting relationship between miR-449a and NLRP3. RESULTS: GA mouse model had increased M1 macrophage, intensified inflammation response, along with suppressed miR-449a expression. Following administration of Trip attenuated cell inflammation, promoted macrophage polarize to M2 phenotype, elevated miR-449a expression, repressed the phosphorylation levels of NF-κB signaling-related proteins, and diminished IκBα expression in vivo and in vitro. However, inhibition of miR-449a hindered the favorable effect of Trip on GA and increased NLRP3 inflammasome expression. MiR-449a directly targeted NLRP3. Overexpression of NLRP3 partially eliminated the biological effects of miR-449a agonist. CONCLUSION: Trip regulates macrophage polarization through miR-449a/NLRP3 axis and the STAT3/NF-κB pathway to mitigate GA. The elucidation on the molecular mechanism of Trip in GA may provide theoretical guidance for clinical therapy of GA.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Gouty/drug therapy , MicroRNAs , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pentacyclic Triterpenes/therapeutic use , Animals , Ankle Joint/drug effects , Ankle Joint/immunology , Ankle Joint/pathology , Anti-Inflammatory Agents/pharmacology , Arthritis, Gouty/chemically induced , Cytokines/genetics , Cytokines/immunology , HEK293 Cells , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Inbred C57BL , NF-kappa B/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pentacyclic Triterpenes/pharmacology , Phenotype , STAT3 Transcription Factor/immunology , Toe Joint/drug effects , Toe Joint/immunology , Toe Joint/pathology , Uric Acid
3.
Int Immunopharmacol ; 87: 106830, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32738596

ABSTRACT

Berberine (BBR) is the effective constituent of Cortex phellodendri and was characterized as an excellent anti-microbial agent with significant anti-inflammatory effects. Previously, we had demonstrated that BBR alleviated the inflammatory response in adjuvant-induced arthritis (AA) rats by regulating polarization of macrophages. However, the exact mechanics by which BBR regulates macrophage polarization remained unclear. Here, we showed that BBR treatment had little influence on total number of macrophages in joints of AA rats, but increased the proportion of M2 macrophages and decreased the proportion of M1 macrophages. Meanwhile, we found BBR up-regulated the expression of AMP-activated protein kinase phosphorylation (p-AMPK) and down-regulated the expression of Hypoxia inducible factor 1α (HIF-1α) in synovial macrophages of AA rats. In vitro, using LPS-stimulated peritoneal macrophages from normal rats, we also verified that pretreatment with BBR promoted transition from M1 to M2 by up-regulating the expression of p-AMPK and suppressing the expression of HIF-1α. Compound C (an AMPK inhibitor) could abrogate the inhibition of BBR on migration of macrophages. Glycolysis of M1 suppressed by BBR through decreasing lactate export, glucose consumption, and increasing intracellular ATP content, which was remarkably reversed by Compound C. These findings indicated that anti-arthritis effect of BBR is associated with regulating energy metabolism of macrophages through AMPK/HIF-1α pathway.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Berberine/therapeutic use , AMP-Activated Protein Kinases/metabolism , Animals , Ankle Joint/drug effects , Ankle Joint/immunology , Ankle Joint/pathology , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Berberine/pharmacology , Cytokines/blood , Energy Metabolism/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Rats, Sprague-Dawley
4.
Int Immunopharmacol ; 77: 105919, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31655341

ABSTRACT

BACKGROUND: Rheumatoid arthritis (RA) is a systematic, inflammatory, autoimmune disease, associated with a high number of disabilities. Increasing evidence has demonstrated that neutrophil extracellular trap (NET) formation plays a significant role in the pathogenesis and progression of RA. In this study, we have aimed to investigate the effects of polydatin (PD) on NET formation and its effects on disease activity in a collagen-induced arthritis (CIA) mouse model. METHODS: In the presence of PD or vehicle, neutrophils isolated from RA patients and mice were treated with phorbol 12-myristate 13-acetate (PMA) for 4 h, and NET formation investigated. For in vivo experiments, PD was administered intraperitoneally (45 mg/kg per day) to collagen-induced arthritis (CIA) mice. The incidence and severity of collagen-induced arthritis were assessed and NET deposition tested. RESULTS: In vitro, PD significantly suppressed NET formation of neutrophils from RA patients. Consistently, decreased NETs were observed in PD treated bone marrow-derived neutrophils. In CIA mouse model, PD treatment delayed the onset of arthritis and attenuated arthritis severity. Compared with vehicle-treated CIA mice, the deposition of NETs in ankle joints was also reduced in PD-treated CIA mice. CONCLUSION: In this study, we found that PD treatment markedly inhibited NET formation and protected CIA mice from the development of arthritis. These findings suggest that inhibition of NET formation by PD may serve as a novel mechanism for the treatment of RA.


Subject(s)
Arthritis, Experimental/drug therapy , Extracellular Traps/drug effects , Glucosides/therapeutic use , Protective Agents/therapeutic use , Stilbenes/therapeutic use , Animals , Ankle Joint/drug effects , Ankle Joint/immunology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Autoantibodies/blood , Glucosides/pharmacology , Humans , Male , Mice, Inbred DBA , Neutrophils/immunology , Protective Agents/pharmacology , Stilbenes/pharmacology
5.
Int J Mol Sci ; 20(20)2019 Oct 11.
Article in English | MEDLINE | ID: mdl-31614480

ABSTRACT

Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by symmetric polyarthritis with swelling and pain at synovial joints. In RA patients, delayed neutrophil apoptosis amplifies the inflammatory response and massively released neutrophil extracellular traps (NETs) induce tissue damage and provide self-antigens. Andrographolide (AD) is the major active labdane diterpenoid derived from Andrographis paniculata, which has multiple pharmacological effects, including hepatoprotection, anti-angiogenesis, anti-thrombosis, and anti-inflammation. In the present study, we investigated the effect of AD on an adjuvant-induced arthritis (AA) murine model of RA and found that AD alleviated murine arthritis by reducing neutrophil infiltration and NETosis in the ankle joints and relieved the systematic inflammation. In vitro experiments showed that AD accelerated the apoptosis of lipopolysaccharide-activated neutrophils and inhibited autophagy-dependent extracellular traps formation of neutrophils. These findings suggest that AD has considerable potential for RA therapy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Arthritis, Rheumatoid/drug therapy , Diterpenes/administration & dosage , Neutrophils/cytology , Animals , Ankle Joint/drug effects , Ankle Joint/immunology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Cells, Cultured , Cytokines/drug effects , Cytokines/metabolism , Disease Models, Animal , Diterpenes/pharmacology , Extracellular Traps/drug effects , Extracellular Traps/metabolism , Gene Expression Regulation/drug effects , Humans , Lipopolysaccharides/pharmacology , Male , Mice , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/immunology
6.
J Ethnopharmacol ; 236: 183-195, 2019 May 23.
Article in English | MEDLINE | ID: mdl-30849505

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Paullinia pinnata L. (Sapindaceae) is an African woody vine, traditionally used for the treatment of itch and pain-related conditions such as rheumatoid arthritis. AIM: This work evaluates, in vitro and in vivo, the anti-inflammatory and analgesic effects of aqueous (AEPP) and methanol (MEPP) extracts from Paullinia pinnata leaves. METHODS: AEPP and MEPP (100, 200 and 300 mg/kg/day) were administered orally in monoarthritic rats induced by a unilateral injection of 50 µl of Complete Freund's Adjuvant (CFA) in the ankle joint. During the 14 days of treatment, pain and inflammation were evaluated alternatively in both ankle and paw of the CFA-injected leg. Malondialdehyde (MDA) and glutathione (GSH) levels were assessed in serum and spinal cord. Histology of soft tissue of the ankle was also analyzed. For in vitro studies, AEPP and MEPP (10, 30 and 100 µg/ml) were evaluated against nitric oxide (NO) production by macrophages that were either non-stimulated or stimulated with LPS, 8-Br-AMPc and the mixture of both substances after 8 h exposure. These extracts were also evaluated on TNF-α and IL-1ß production in cells stimulated with LPS for 8 h. Finally, the ability of the extracts to bind to neuroactive receptors was evaluated in vitro using competitive binding assays with >45 molecular targets. RESULTS: AEPP and MEPP significantly reduced by 20-98% (p < 0.001) the inflammation and pain sensation in both the ankle and paw. AEPP significantly increased glutathione levels (p < 0.05) in serum. Both extracts reduced MDA production in serum and spinal cord (p < 0.001), and significantly improved tissue reorganization in treated arthritic rats. P. pinnata extracts did not affect NO production in non-stimulated macrophages but significantly reduced it by 47-88% in stimulated macrophages. AEPP and MEPP also significantly inhibited TNF-α (35-68%) and IL-1ß (31-36%) production in LPS stimulated macrophages. No cytotoxic effect of plant extracts was observed. MEPP showed concentration-dependent affinity for Sigma 2 receptors with an IC50 of 50 µg/ml. CONCLUSION: These results demonstrate the analgesic and anti-inflammatory effects of P. pinnata extracts on monoarthritis and further support its traditional use for pain and inflammation. These activities are at least partly due to the ability of these extracts to inhibit the production of NO, TNF-α, IL-1ß and to likely modulate Sigma 2 receptors.


Subject(s)
Analgesics/pharmacology , Anti-Inflammatory Agents/pharmacology , Arthralgia/drug therapy , Arthritis, Rheumatoid/drug therapy , Medicine, African Traditional/methods , Paullinia/chemistry , Plant Extracts/pharmacology , Analgesics/isolation & purification , Analgesics/therapeutic use , Animals , Ankle Joint/drug effects , Ankle Joint/immunology , Ankle Joint/pathology , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/therapeutic use , Arthralgia/etiology , Arthralgia/pathology , Arthritis, Experimental/complications , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Drug Evaluation, Preclinical , Female , Freund's Adjuvant/administration & dosage , Freund's Adjuvant/immunology , Humans , Male , Methanol/chemistry , Nitric Oxide/metabolism , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Plant Leaves/chemistry , Rats , Rats, Wistar , Treatment Outcome , Water/chemistry
7.
Injury ; 48(7): 1287-1295, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28551052

ABSTRACT

Osteochondral allografts are used to treat many different conditions as acute traumatic large-sized lesions, degenerative osteoarthritis, osteochondritis dissecans, avascular necrosis or in case of failure of previous procedures particularly in young patients for whom primary prosthesis is not desirable. Fresh allografts present the advantage of having mature viable hyaline cartilage, not causing donor morbidity, allowing the restoration of even large defects in a single surgical session. Conversely, they could account for risks of disease transmission, immunologic reactions, and for limited availability. The present review aimed to analyze published studies of the last decade in which patients received fresh osteochondral allografts by dividing them for knee or ankle regenerative purposes. We wish to report the observed failure rates and particularly to collect any other reported side effect or outcome for identifying major problems and limits linked to the procedure and for delineating possible future researches and approaches. The overall success rates resulted ranging from 5.3% to 48.3% in the ankle at a mean follow up of 3.3 years and from 0% to 85.7% in the knee at a mean follow up of 7.1 years. Among other outcomes, occurrence or progression of arthritis, osteolysis, graft instability, fractures, nonunions, edema and infections were recorded. Overall, the lack of well designed randomized and controlled clinical trials, of immunological determination of the anti-donor antibodies development and of local and systemic biomarkers to detect reaction to the graft seems to be the major drawback. Improvements in these limiting factors might be desirable in order to enhance the clinical scenario of a well-established and successful procedure to give, especially for young patients, a real regeneration of the joint.


Subject(s)
Ankle Joint/surgery , Bone Transplantation , Knee Joint/surgery , Osteoarthritis/surgery , Osteochondritis Dissecans/surgery , Osteonecrosis/surgery , Transplantation, Homologous , Ankle Joint/immunology , Ankle Joint/physiopathology , Bone Transplantation/adverse effects , Cartilage, Articular/cytology , Cartilage, Articular/surgery , Follow-Up Studies , Graft Rejection , Humans , Knee Joint/immunology , Knee Joint/physiopathology , Observational Studies as Topic , Osteoarthritis/physiopathology , Osteochondritis Dissecans/physiopathology , Osteonecrosis/physiopathology , Prospective Studies , Regeneration/immunology , Retrospective Studies , Transplantation, Homologous/adverse effects , Treatment Failure
8.
Arthritis Rheumatol ; 68(10): 2476-86, 2016 10.
Article in English | MEDLINE | ID: mdl-27111864

ABSTRACT

OBJECTIVE: The spondyloarthritides (SpA) are a group of rheumatic diseases characterized by ossification and inflammation of entheseal tissue, the region where tendon attaches to bone. Interleukin-23 (IL-23) is involved in the pathogenesis of SpA by acting on IL-23 receptor (IL-23R) expressed on enthesis-resident lymphocytes. Upon IL-23 binding, CD3+CD4-CD8- tissue-resident lymphocytes secrete IL-17A and IL-22, leading to inflammation, bone loss, and ossification. Knowledge about enthesis-resident lymphocytes remains fragmentary, and the contribution of entheseal γ/δ T cells in particular is not clear. This study was undertaken to investigate the presence of γ/δ T cells in the enthesis. METHODS: We used 2-photon microscopy and flow cytometry to analyze entheseal lymphocytes from C57BL/6, Tcrd-H2BeGFP, Rorc-GFP, and IL-23R-eGFP mice. To analyze entheseal γ/δ T cells in IL-23-induced inflammation, Tcrd-H2BeGFP mice were crossed with mice of the susceptible B10.RIII background. Hydrodynamic injection of IL-23 minicircle DNA was performed for overexpression of IL-23 and induction of inflammation. Light-sheet fluorescence microscopy was used to visualize arthritic inflammation. RESULTS: Activated Vγ6+CD27- γ/δ T cells were abundant in uninflamed entheseal tissue and constituted the large majority of retinoic acid receptor-related orphan nuclear receptor γt (RORγt)+IL-23R+ enthesis-resident lymphocytes. Fetal thymus-dependent γ/δ T cells were the main source of IL-17A at the enthesis. Under inflammatory conditions, γ/δ T cells increased in number at the Achilles tendon enthesis, aortic root, and adjacent to the ciliary body. CONCLUSION: Entheseal γ/δ T cells are derived from fetal thymus and are maintained as self-renewing tissue-resident cells. As main IL-17A producers within tissues exposed to mechanical stress including enthesis, γ/δ T cells are key players in the pathogenesis of IL-23-induced local inflammation.


Subject(s)
Achilles Tendon/immunology , Aortic Valve/immunology , Ciliary Body/immunology , Interleukin-23/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Spondylarthropathies/immunology , T-Lymphocyte Subsets/immunology , Achilles Tendon/pathology , Animals , Ankle Joint/immunology , Ankle Joint/pathology , Aortic Valve/pathology , Ciliary Body/pathology , Enthesopathy/immunology , Enthesopathy/pathology , Flow Cytometry , Green Fluorescent Proteins/genetics , Interleukin-17/immunology , Interleukins/immunology , Mice , Mice, Transgenic , Microscopy, Fluorescence , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Spondylarthropathies/pathology , T-Lymphocyte Subsets/pathology , X-Ray Microtomography , Interleukin-22
9.
Rheumatol Int ; 33(10): 2597-605, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23722874

ABSTRACT

We have recently reported that CD4(+) T cells synthesize and secrete catecholamines that facilitate a shift of T helper 1 (Th1)/Th2 balance toward Th2 polarization. In this study, we used an animal model of human rheumatoid arthritis, collagen type II-induced arthritis (CIA), to explore relationship between catecholamine production in CD4(+) T cells and Th1-/Th2-mediated joint inflammation. Histopathological observation of ankle joints of CIA mice displayed an evident inflammatory change on day 35 and a major damage to bones on day 55 post-immunization. Expression of Th1-specific transcription factor, T-bet, and cytokines, IL-2 and IFN-γ, and Th2-specific transcription factor, GATA-3, and cytokines, IL-4 and IL-10, was all upregulated on days 35 and 55 post-immunization, but the elevated Th1 response tended to decrease and the enhanced Th2 response tended to increase with the CIA progression. Expression of tyrosine hydroxylase (TH), a rate-limiting enzyme for synthesis of catecholamines, dramatically increased in ankle joints of CIA mice, although this increase was reduced on day 55 relative to that on day 35 post-immunization. In synovial tissue of CIA ankle joints but not normal joints, CD4-, T-bet-, GATA-3-, and TH-immunoreactive cells were found. Importantly, co-expressed cells with CD4 and TH, T-bet and TH, and GATA-3 and TH were observed in synovial tissue of CIA ankle joints. These results suggest that an increase in catecholamine production occurs in inflamed joints of CIA. The catecholamines are, at least in part, from Th1 and Th2 cells, and they may be related to joint inflammatory alleviation in CIA progression.


Subject(s)
Ankle Joint/metabolism , Arthritis, Experimental/metabolism , CD4-Positive T-Lymphocytes/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Ankle Joint/immunology , Ankle Joint/pathology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Disease Progression , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred DBA , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology
10.
PLoS One ; 8(2): e56349, 2013.
Article in English | MEDLINE | ID: mdl-23431370

ABSTRACT

Rheumatoid arthritis (RA) is the most common arthritis and is mainly characterized by symmetric polyarticular joint disorders. Our previous study demonstrated a novel small molecule compound (Z)-N-(3-Chlorophenyl)-2-(4-((2,4-dioxothiazolidin-5-ylidene) methyl) phenoxy) acet-amide (SKLB023) showed potently anti-arthritic effects in a rat arthritis model, however, the underlying mechanisms for this are largely unknown. Both NF-κB and macrophages were reported to play important roles in the pathologic processes of RA. The purposes of this study were to indicate whether NF-κB and macrophages contributed to anti-arthritic effects of SKLB023 in two experimental arthritis models. Our results showed that SKLB023 could significantly improve joint inflammation and cartilage destruction both in adjuvant induced arthritis (AIA) and collagen-induced arthritis (CIA) models. We further found that the binding activation of NF-κB to DNA in joint tissues and RAW264.7 macrophages were suppressed by SKLB023. SKLB023 also inhibited the NF-κB activity in peritoneal macrophages by luciferase assay. Furthermore, the number of macrophages in synovial tissues was decreased after the treatment of different doses of SKLB023. The levels of TNF-α, IL-1ß, and IL-6 in plasma, and the levels of TNF-α, NO, and IL-1ß in peritoneal macrophages were down-regulated by SKLB023. Finally, SKLB023 attenuated the expression of iNOS and COX-2 in vivo and suppressed the phosphorylations of components of the mitogen-activated protein kinases (MAPKs). These observations identify a novel function for SKLB023 as an inhibitor of NF-κB in macrophages of RA, highlighting that SKLB023 was a potential therapeutic strategy for RA.


Subject(s)
Acetanilides/pharmacology , Ankle Joint/pathology , Anti-Inflammatory Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Cartilage, Articular/pathology , Macrophages/drug effects , NF-kappa B/metabolism , Thiazolidinediones/pharmacology , Acetanilides/therapeutic use , Animals , Ankle Joint/drug effects , Ankle Joint/immunology , Anti-Inflammatory Agents/therapeutic use , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Cartilage, Articular/drug effects , Cartilage, Articular/immunology , Cell Line , Cyclooxygenase 2/metabolism , Cytokines/blood , Disease Models, Animal , Female , Inflammation Mediators/blood , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred DBA , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Protein Processing, Post-Translational , Rats , Rats, Inbred Lew , Signal Transduction/drug effects , Thiazolidinediones/therapeutic use
11.
J Immunol Methods ; 386(1-2): 78-84, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-22982203

ABSTRACT

Complete Freund's adjuvant (CFA) containing the inactivated mycobacterium has been conventionally used to induce typical arthritis in rats. In comparison, incomplete Freund's adjuvant (IFA), lacking the mycobacterium, can only induce less severe arthritis. However, the key components responsible for the arthritogenic effect of the whole mycobacterium are rarely known. Although mycobacterial heat-shock protein 65 (MHSP65) specific humoral and cellular immune responses were detected in rats with arthritis induced by CFA, MHSP65 alone cannot induce arthritis. In this study, we replaced the whole mycobacterium in CFA with recombinant MHSP65 (rMHSP65), prepared rMHSP65-IFA emulsion by mixing rMHSP65 and IFA, and investigated whether rMHSP65-IFA could induce arthritis in rats as CFA did. We found that intradermal injection of the rMHSP65-IFA emulsion induced arthritic lesions in testing animals to the same degree as those induced by CFA, manifested by severe swelling in hind paws, and synovial thickening, cartilage erosion and lymphocytes infiltration in ankle joints. Notably, the rMHSP65-IFA recipe also induced the production of anti-dsDNA and -rMHSP65 antibodies in rats. These results thus demonstrate that rMHSP65 can be used to substitute the inactivated mycobacteria in CFA to induce typical arthritis in rats.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Bacterial Proteins/immunology , Chaperonin 60/immunology , Freund's Adjuvant/immunology , Lipids/immunology , Animals , Ankle Joint/immunology , Antibodies, Antinuclear/metabolism , Humans , Male , Models, Animal , Rats , Rats, Wistar , Synovial Membrane/pathology
12.
J Immunol ; 189(5): 2488-501, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22851707

ABSTRACT

Localized elevation in type I IFN has been uniquely linked to the severe Lyme arthritis that develops in C3H mice infected with the spirochete Borrelia burgdorferi. In this study, the dynamic interactions that result in generation of these responses were further examined in C3H mice carrying the type I IFN receptor gene ablation, which effectively blocks all autocrine/paracrine signaling crucial to induction of downstream effectors. Reciprocal radiation chimeras between C3H and IFNAR1⁻/⁻ mice implicated both radiation-sensitive and radiation-resistant cells of the joint tissue in the proarthritic induction of type I IFN. Ex vivo analysis of cells from the naive joint revealed CD45⁺ cells residing in the tissue to be uniquely capable of initiating the type I IFN response to B. burgdorferi. Type I IFN responses were analyzed in real time by lineage sorting of cells from infected joint tissue. This demonstrated that myeloid cells, endothelial cells, and fibroblasts were responsible for propagating the robust IFN response, which peaked at day 7 postinfection and rapidly resolved. Endothelial cells and fibroblasts were the dominant sources of IFN signature transcripts in the joint tissue. Fibroblasts were also the major early source of chemokines associated with polymorphonuclear leukocyte and monocyte/macrophage infiltration, thus providing a focal point for arthritis development. These findings suggest joint-localized interactions among related and unrelated stromal, endothelial, and myeloid cell lineages that may be broadly applicable to understanding the pathogeneses of diseases associated with type I IFN signature, including systemic lupus erythematosus and some rheumatoid arthritides.


Subject(s)
Arthritis, Experimental/immunology , Fibroblasts/immunology , Interferon Type I/biosynthesis , Lyme Disease/immunology , Myeloid Cells/immunology , Up-Regulation/immunology , Animals , Ankle Joint/immunology , Ankle Joint/microbiology , Ankle Joint/pathology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Borrelia burgdorferi/immunology , Borrelia burgdorferi/pathogenicity , Cells, Cultured , Chemokines/biosynthesis , Chemokines/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Interferon Type I/deficiency , Interferon Type I/genetics , Lyme Disease/metabolism , Lyme Disease/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Cells/pathology , Severity of Illness Index , Signal Transduction/immunology , Transcription, Genetic/immunology , Up-Regulation/genetics
13.
Arthritis Res Ther ; 14(3): R137, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-22676399

ABSTRACT

INTRODUCTION: We sought to investigate the capacity of interleukin (IL)-7 to enhance collagen-induced arthritis and to study by what mechanisms this is achieved. METHODS: Mice received multiple injections with IL-7 or phosphate-buffered saline (PBS) as a control. Arthritis severity and incidence were determined by visual examination of the paws. Joint destruction was determined by assessing radiographs and immunohistochemistry of the ankle joints. Total cellularity and numbers of T-cell and B-cell subsets were assessed, as well as ex vivo production of interferon-γ (IFN-γ), IL-17, and IL-4. Proinflammatory mediators were measured in serum with multianalyte profiling. RESULTS: IL-7 increased arthritis severity and radiology-assessed joint destruction. This was consistent with IL-7-increased intensity of cell infiltrates, bone erosions, and cartilage damage. Splenic CD19+ B cells and CD19+/GL7+ germinal center B cells, as well as CD4 and CD8 numbers, were increased by IL-7. IL-7 expanded memory T cells, associated with increased percentages of IFN-γ-, IL-4-, and IL-17-producing CD4+ T cells. On antigen restimulation of draining lymph node cells in vitro IL-7 treatment was found to increase IFN-γ and IL-17 production, whereas IL-4 was reduced. IL-7 also increased concentrations of proinflammatory mediators, indicative of T-cell activation (sCD40L), vascular activation (VCAM-1, VEGF), tissue destruction (fibroblast growth factor-basic (FGF-b), LIF), and chemotaxis (MIP-1γ, MIP-3ß, lymphotactin, MDC, and MCP-5). CONCLUSIONS: In arthritic mice, IL-7 causes expansion of T and B cells, associated with increased levels of proinflammatory mediators. IL-7 intensifies arthritis severity and joint destruction, accompanied by increased Th1 and Th17 activity. These data indicate that IL-7 could be an important mediator in arthritic conditions and that targeting IL-7 or its receptor represent novel therapeutic strategies.


Subject(s)
Arthritis, Experimental/immunology , B-Lymphocytes/immunology , Interleukin-7/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Ankle Joint/immunology , Ankle Joint/pathology , Arthritis, Experimental/pathology , Flow Cytometry , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred DBA
14.
J Ethnopharmacol ; 141(1): 290-300, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22391142

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Paeoniflorin (Pae) is extracted from the root of paeonia lactiflora which have attracted attention for anti-rheumatic and immune modulating properties. AIM OF THE STUDY: To investigate the role of PI3K/Akt/mTOR signaling mediated by BAFF/BAFF-R in antibodies production and the regulation of Pae on the signaling pathway in rats with collagen-induced arthritis (CIA). MATERIALS AND METHODS: CIA rats were randomly separated into different groups and treated with Pae (25, 100mg/kg) from day 18 to day 38 after immunization. The effects of Pae on B lymphocytes of CIA rats were evaluated by the levels of BAFF, anti-CII antibody, IgA, IgG and IgM, and the expressions of BAFF-R, PI3K, p-Akt and mTOR. RESULTS: In CIA rats, the levels of anti-CII antibody, IgA, IgG and IgM in serum enhanced, BAFF, BAFF-R, PI3K, p-Akt and mTOR were highly expressed. Pae (100mg/kg) obviously decreased arthritis score, relieved ankle and paw swelling, improved spleen histopathology in CIA rats, decreased the levels of IgA, IgM, IgG and anti-CII antibody, and significantly decreased the expressions of BAFF, BAFF-R, PI3K, p-Akt and mTOR. CONCLUSION: PI3K/Akt/mTOR signaling mediated by BAFF/BAFF-R participates in antibodies production by B lymphocytes of CIA rats. Pae had therapeutic effects on rats with CIA. These effects might be relative to regulating PI3K/Akt/mTOR signal mediated by BAFF/BAFF-R, and down regulate the antibodies production further.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/drug therapy , Autoantibodies/blood , B-Cell Activating Factor/immunology , B-Cell Activation Factor Receptor/immunology , Benzoates/pharmacology , Bridged-Ring Compounds/pharmacology , Glucosides/pharmacology , Paeonia , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Ankle Joint/drug effects , Ankle Joint/enzymology , Ankle Joint/immunology , Ankle Joint/pathology , Anti-Inflammatory Agents/isolation & purification , Arthritis, Experimental/enzymology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , Benzoates/isolation & purification , Bridged-Ring Compounds/isolation & purification , Glucosides/isolation & purification , Male , Monoterpenes , Paeonia/chemistry , Phosphorylation , Phytotherapy , Plant Roots , Plants, Medicinal , Rats , Rats, Sprague-Dawley , Spleen/drug effects , Spleen/enzymology , Spleen/immunology , Spleen/pathology , Time Factors
15.
Int Immunol ; 24(6): 379-88, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22354915

ABSTRACT

T lymphocytes are key inflammatory cells contributing significantly to the pathogenesis of Rheumatoid arthritis (RA). Biological treatments targeting T lymphocytes may provide an efficient approach for treatment of RA. CTLA4-FasL, a fusion product of extracellular domains of CTLA4 and FasL, integrating two inhibitory elements against T cells into one molecule, might be a desirable derivative of engineered soluble FasL or CTLA4 and have therapeutic potential in RA. The aim of this study was to investigate whether simultaneous induction of Fas-mediated apoptosis and blockade of co-stimulation signal by CTLA4-FasL gene delivery has a suppressive effect on adjuvant-induced arthritis (AIA) in Lewis rats. Recombinant adeno-associated virus (rAAV) vectors encoding rat CTLA4-FasL fusion gene (rAAV.CTLA4-FasL) or enhanced green fluorescent protein (rAAV.EGFP) were injected intraarticularly into both ankle joints after immunization. The ankles were monitored by measures of clinical, histological and inflammatory cytokines' changes. Treatment using rAAV.CTLA4-FasL resulted in a significant suppression of AIA compared with rAAV.EGFP control, as reflected in the mainly clinical signs including articular index, ankle joint thickness and paw swelling and typically histological characters of arthritic joints including synovial hyperplasia, inflammatory cells infiltration and cartilage degradation. Treatment with rAAV.CTLA4-FasL also significantly decreased the levels of key proinflammatory cytokines in AIA joints. Moreover, local productions of transgene mRNA and protein of CTLA4-FasL were found in injected joints without systemic distribution. Our results indicate that rAAV.CTLA4-FasL profoundly suppressed experimental model of RA, implicating the potential therapeutic applications for suppression of RA by local joint delivery of CTLA4-FasL.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/therapy , CTLA-4 Antigen/immunology , Fas Ligand Protein/immunology , Animals , Ankle Joint/immunology , Ankle Joint/metabolism , Ankle Joint/pathology , Arthritis, Experimental/genetics , Blotting, Western , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Cytokines/immunology , Cytokines/metabolism , Dependovirus/genetics , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , Gene Expression , Gene Transfer Techniques , Genetic Therapy/methods , HEK293 Cells , Humans , Injections, Intra-Articular , Protein Binding/immunology , Rats , Rats, Inbred Lew , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , fas Receptor/immunology , fas Receptor/metabolism
16.
J Ethnopharmacol ; 137(3): 1381-7, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21855621

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The fruit of Thladiantha dubia has been used in Chinese manchu nationality folk medicine for treatment of various pains, such as rheumatic pain, lumbocrural pain, and dysmenorrhea. The aim of the present study was to evaluate the anti-rheumatic effect of Thladiantha dubia fruit crude polysaccharide (TF-P) fraction in rats. MATERIALS AND METHODS: TF-P was evaluated in mice for analgesic activity using acetic acid-induced abdominal contractions test and for anti-inflammatory activity using xylene-induced ear oedema model. Moreover, rheumatoid arthritis (RA) was induced by injection of CFA into the subplantar surface of the hind paw of the male Wistar rats. Joint swelling was measured. In order to evaluate the effect of TF-P on disease progression, proinflammatory mediators (TNF-α and IL-2), and antioxidant indicators (SOD, MDA, and NO) were determined in rats induced by CFA. Pathologic changes of RA in rats were also observed under light microscope. RESULTS AND CONCLUSIONS: In the present study, TF-P at the dose of 200mg/kg (according to the clinical traditional dosage) significantly reduced writhings and stretchings induced by the acetic acid in mice. TF-P significantly inhibited xylene-induced ear oedema in mice, compared with control group. TF-P significantly inhibited inoculated and non-inoculated joint swellings in rats induced by CFA. TF-P had no effect of body weight in rats. The histopathological analysis suggested that TF-P obviously alleviated the degree of RA rats. TF-P treated rats preserved a nearly normal histological architecture of the joint. Results of the present study confirm the use of Thladiantha dubia traditionally for the treatment of painful and inflammatory conditions. Those results suggest TF-P has protective and therapeutic effects on RA rats induced by CFA.


Subject(s)
Analgesics/pharmacology , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Cucurbitaceae , Drugs, Chinese Herbal/pharmacology , Edema/prevention & control , Pain/prevention & control , Polysaccharides/pharmacology , Acetic Acid , Analgesics/isolation & purification , Animals , Ankle Joint/drug effects , Ankle Joint/immunology , Ankle Joint/metabolism , Ankle Joint/pathology , Antirheumatic Agents/isolation & purification , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Cucurbitaceae/chemistry , Disease Models, Animal , Disease Progression , Drugs, Chinese Herbal/isolation & purification , Edema/chemically induced , Female , Freund's Adjuvant , Fruit , Inflammation Mediators/blood , Interleukin-2/blood , Male , Malondialdehyde/blood , Mice , Nitric Oxide/blood , Pain/chemically induced , Pain Measurement , Plants, Medicinal , Polysaccharides/isolation & purification , Rats , Rats, Wistar , Superoxide Dismutase/blood , Tumor Necrosis Factor-alpha/blood , Xylenes
17.
Arthritis Rheum ; 63(7): 1878-87, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21391201

ABSTRACT

OBJECTIVE: The interleukin-7 (IL-7)-related cytokine thymic stromal lymphopoietin (TSLP) is a potent activator of myeloid dendritic cells, enhancing Th2-mediated hypersensitivity, and it has been implicated in the pathogenesis of atopic diseases. Although intraarticular concentrations of TSLP have been shown to be increased in patients with rheumatoid arthritis (RA), the functional capacities of TSLP in arthritis are poorly studied. The purpose of this study was to investigate the effects of TSLP administration and TSLP receptor deficiency on immune activation, arthritis severity, and tissue destruction in T cell-driven arthritis models of RA. METHODS: Immunopathology was studied in arthritic mice that were given multiple injections of murine recombinant TSLP and in mice that were deficient in the TSLP receptor (TSLPR(-/-)). Arthritis severity and incidence were determined by visual examination of the paws. Joint destruction was determined by assessing radiographs and the immunohistochemistry of ankle joints. Total cellularity and numbers of T cell subsets were assessed. Proinflammatory mediators were measured by multianalyte profiling of serum or paw protein extracts. RESULTS: Administration of TSLP significantly exacerbated the severity of collagen-induced arthritis and the joint damage that was associated with increased T cell activation. Furthermore, TSLPR(-/-) mice had less severe arthritis than did wild-type mice. TSLPR(-/-) mice had diminished concentrations of local proinflammatory and catabolic mediators, including IL-17, IL-1ß, IL-6, basic fibroblast growth factor, and matrix metalloproteinase 9, while levels of the regulatory cytokines IL-10 and IL-13 were increased. CONCLUSION: TSLP and its receptor enhance Th17-driven arthritis and tissue destruction in experimental arthritis. The increased expression of TSLP as well as the increased number of TSLPR-expressing cells in the joints of patients with RA suggest that TSLP and its receptor constitute novel therapeutic targets in RA.


Subject(s)
Ankle Joint/diagnostic imaging , Arthritis, Experimental/metabolism , Cytokines/metabolism , Immunoglobulins/metabolism , Receptors, Cytokine/metabolism , Animals , Ankle Joint/immunology , Ankle Joint/metabolism , Arthritis, Experimental/diagnostic imaging , Arthritis, Experimental/immunology , Cytokines/immunology , Flow Cytometry , Immunoglobulins/immunology , Inflammation/diagnostic imaging , Inflammation/immunology , Inflammation/metabolism , Interleukin-7/immunology , Interleukin-7/metabolism , Mice , Mice, Knockout , Radiography , Receptors, Cytokine/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Thymic Stromal Lymphopoietin
18.
Clin Exp Immunol ; 163(2): 225-34, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21155990

ABSTRACT

Transmembrane activator and calcium modulator and cyclophilin ligand interactor-immunoglobulin (TACI-Ig) is a human fusion protein that binds and neutralizes both B lymphocyte stimulator (BLyS), a cytokine shown to be a key regulator of B cell maturation, proliferation and survival, and a proliferation-inducing ligand (APRIL). Rat adjuvant arthritis (AA) is an experimental animal model of rheumatoid arthritis (RA), which is mainly dependent on T cells and neutrophil-mediated cytokine production. The purpose of the present study was to investigate the effects of TACI-Ig on rat AA. Rat AA was induced by intradermal injection of 0·1 ml complete Freund's adjuvant (CFA). TACI-Ig (0·7, 2·1 and 6·3 mg/kg), recombinant human tumour necrosis factor-α receptor (rhTNFR) : Fc (2·8 mg/kg) and IgG-Fc (6·3 mg/kg) were administered subcutaneously every other day from days 16 to 34 after immunization. Arthritis was evaluated by arthritis global assessment and swollen joint count (SJC). The ankle joint and spleen were harvested for histopathological examination. Spleen index and thymus index were calculated. The levels of BLyS, interleukin (IL)-17, interferon (IFN)-γ, IgG1, IgG2a and IgM in AA rat spleen were measured by enzyme-linked immunosorbent assay. Administration of TACI-Ig significantly reduced the arthritis global assessment and SJC, decreased spleen index and ameliorated histopathological manifestations of rat AA. Suppressing the levels of BLyS, IL-17, IFN-γ and Ig in AA rat spleen were observed after administration of TACI-Ig. These results showed that TACI-Ig significantly inhibited the degree of rat AA, and the inhibitory effects might be associated with its ability to reduce BLyS, proinflammatory cytokines and Ig levels in spleen.


Subject(s)
Arthritis, Experimental/drug therapy , Recombinant Fusion Proteins/therapeutic use , Adjuvants, Immunologic/adverse effects , Animals , Ankle Joint/immunology , Ankle Joint/pathology , B-Cell Activating Factor/analysis , B-Cell Activating Factor/immunology , Immunoglobulin G/immunology , Immunoglobulin M/analysis , Immunoglobulin M/immunology , Interferon-gamma/analysis , Interferon-gamma/immunology , Interleukin-17/analysis , Interleukin-17/immunology , Male , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor/immunology , Spleen/immunology , Spleen/pathology
19.
J Immunol ; 184(6): 3233-41, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20173024

ABSTRACT

Angiogenesis is an early and a critical event in the pathogenesis of rheumatoid arthritis (RA). Neovascularization is dependent on endothelial cell activation, migration and proliferation, and inhibition of angiogenesis may provide a novel therapeutic approach in RA. In this study, we document a novel role of IL-17 in mediating angiogenesis. Local expression of IL-17 in mouse ankles increases vascularity. We further demonstrate that IL-17 is angiogenic by showing its ability to promote blood vessel growth in Matrigel plugs in vivo. Additionally, IL-17, in concentrations present in the RA joint, induces human lung microvascular endothelial cell (HMVEC) migration mediated through the PI3K/AKT1 pathway. Furthermore, suppression of the PI3K pathway markedly reduces IL-17-induced tube formation. We also show that both IL-17-induced HMVEC chemotaxis and tube formation are mediated primarily through IL-17 receptor C. Neutralization of either IL-17 in RA synovial fluids or IL-17 receptor C on HMVECs significantly reduces the induction of HMVEC migration by RA synovial fluid. Finally, RA synovial fluid immunoneutralized with anti-IL-17 and antivascular endothelial growth factor does not reduce HMVEC migration beyond the effect detected by immunodepleting each factor alone. These observations identify a novel function for IL-17 as an angiogenic mediator in RA, supporting IL-17 as a therapeutic target in RA.


Subject(s)
Angiogenic Proteins/physiology , Arthritis, Experimental/immunology , Arthritis, Experimental/physiopathology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/physiopathology , Interleukin-17/physiology , Angiogenic Proteins/antagonists & inhibitors , Angiogenic Proteins/biosynthesis , Animals , Ankle Joint/blood supply , Ankle Joint/immunology , Ankle Joint/pathology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , Cell Line , Cell Migration Inhibition/immunology , Cell Movement/immunology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interleukin-17/antagonists & inhibitors , Interleukin-17/biosynthesis , Lung/blood supply , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred C57BL , Receptors, Interleukin-17/physiology , Synovial Fluid/cytology , Synovial Fluid/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/physiology
20.
J Immunol ; 183(10): 6689-97, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19846884

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation in joints and subsequent destruction of cartilage and bone. Inflammatory mediators such as PGs and proinflammatory cytokines contribute to RA progress. Pin1, a peptidyl prolyl isomerase, plays important pathophysiological roles in several diseases, including cancer and neurodegeneration. We found that both Pin1 and cyclooxygenase-2 (COX-2) were highly expressed in ankle tissues of type II collagen-induced RA mice. HTB-94 cells overexpressing Pin1 and primary cultured human chondrocytes showed increased basal expression of proinflammatory proteins (COX-2, inducible NO synthase, TNF-alpha, and IL-1beta). Site-directed mutagenesis revealed that Pin1-mediated transcriptional activation of COX-2 was coordinately regulated by NF-kappaB, CREB, and C/EBP. Gel shift, reporter gene, and Western blot analyses confirmed that NF-kappaB, CREB, and C/EBP were consistently activated in chondrocytes overexpressing Pin1. Treatment of RA mice with juglone, a chemical inhibitor of Pin1, significantly reduced RA progress and COX-2 expression in the ankle tissues. Moreover, juglone dose dependently decreased the basal COX-2 expression in primary cultured chondrocytes from RA patients. These results demonstrate that Pin1 induction during RA progress stimulates proinflammatory protein expression by activating NF-kappaB, CREB, and C/EBP, and suggest that Pin1 is a potential therapeutic target of RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Chondrocytes/immunology , Naphthoquinones/therapeutic use , Peptidylprolyl Isomerase/immunology , Animals , Ankle Joint/drug effects , Ankle Joint/immunology , Ankle Joint/pathology , Arthritis, Rheumatoid/drug therapy , Cell Line , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Collagen Type II/pharmacology , Cyclic AMP Response Element-Binding Protein/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/immunology , Cyclooxygenase 2/metabolism , Genes, Reporter/genetics , Genes, Reporter/immunology , Humans , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice , NF-kappa B/immunology , NF-kappa B/metabolism , NIMA-Interacting Peptidylprolyl Isomerase , Naphthoquinones/administration & dosage , Nitric Oxide Synthase Type II/immunology , Nitric Oxide Synthase Type II/metabolism , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/metabolism , Steroid Isomerases/immunology , Steroid Isomerases/metabolism , Transfection , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...