Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Endocr J ; 70(7): 731-743, 2023 Jul 28.
Article in English | MEDLINE | ID: mdl-37164685

ABSTRACT

Glucocorticoids (GCs) are the important stress hormones and widely prescribed as drugs. Although stress has been suggested as a promoter of tumor progression, the direct influence of GCs on metastasis of tumor is not fully understood. Metastasis is a major cause of death in pancreatic cancer patients. In the present study, we investigated the effect of GCs on progression of pancreatic cancer and elucidated the underlying mechanism. It was found that GCs significantly promote cell adhesion, migration, and invasion of pancreatic cancer cells in vitro and their lung metastasis in vivo. Further mechanistic studies showed that GCs notably up-regulate the expression of a trans-membrane glycoprotein, mucin 1 (MUC1) and increase the activation of AKT. Inhibiting MUC1 expression not only attenuates the activation of AKT, but also significantly reduces the promoting effects of GCs on cell adhesion, migration, invasion, and lung metastasis of pancreatic cancer cells. Moreover, GCs not only significantly up-regulate expression of Rho-associated kinase 1/2 (ROCK1/2) and matrix metalloproteinase 3 and 7 (MMP3/7), but also activate ROCK2, which are also involved in the pro-migratory and pro-invasive effects of GCs in pancreatic cancer cells. Taken together, our findings reveal that GCs promote metastasis of pancreatic cancer cells through complex mechanism. MUC1-PI3K/AKT pathway, ROCK1/2 and MMP3/7 are involved in the promoting effect of GCs on cell migration, invasion and metastasis in pancreatic cancer cells. These results suggest the importance of reducing stress and GCs administration in patients with pancreatic cancer to avoid an increased risk of cancer metastasis.


Subject(s)
Cell Adhesion , Cell Movement , Glucocorticoids , Lung Neoplasms , Neoplasm Invasiveness , Neoplasm Metastasis , Pancreatic Neoplasms , Glucocorticoids/pharmacology , Humans , Lung Neoplasms/pathology , Pancreatic Neoplasms/pathology , Cell Adhesion/drug effects , Cell Movement/drug effects , Neoplasm Invasiveness/pathology , rho-Associated Kinases/drug effects , rho-Associated Kinases/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
2.
Arthritis Rheumatol ; 73(7): 1145-1154, 2021 07.
Article in English | MEDLINE | ID: mdl-33512089

ABSTRACT

OBJECTIVE: We have previously reported that stimulation of mouse bone marrow-derived macrophages with tumor necrosis factor (TNF) and interleukin-6 (IL-6) induces differentiation of osteoclast-like cells. We undertook this study to clarify the characterization and function of human TNF and IL-6-induced osteoclasts using peripheral blood collected from patients with rheumatoid arthritis (RA) and healthy donors. METHODS: Peripheral blood monocytes were cultured with a combination of TNF and IL-6, TNF alone, IL-6 alone, or with RANKL, and their bone resorption ability was evaluated. Expression levels of NFATc1, proinflammatory cytokines, and matrix metalloproteinase 3 were analyzed. The effects of NFAT inhibitor and JAK inhibitor were examined. Furthermore, the relationship between the number of TNF and IL-6-induced osteoclasts or RANKL-induced osteoclasts differentiated from peripheral blood mononuclear cells (PBMCs) in patients with RA and the modified total Sharp score (mTSS) or whole-body bone mineral density (BMD) was examined. RESULTS: Peripheral blood monocytes stimulated with a TNF and IL-6-induced osteoclasts were shown to demonstrate the ability to absorb bone matrix. Cell differentiation was not inhibited by the addition of osteoprotegerin. Stimulation with a combination of TNF and IL-6 promoted NFATc1 expression, whereas the NFAT and JAK inhibitors prevented TNF and IL-6-induced osteoclast formation. Expression levels of IL1ß, TNF, IL12p40, and MMP3 were significantly increased in TNF and IL-6-induced osteoclasts, but not in RANKL-induced osteoclasts. The number of TNF and IL-6-induced osteoclasts differentiated from PBMCs in patients with RA positively correlated with the mTSS, whereas RANKL-induced osteoclast numbers negatively correlated with the whole-body BMD of the same patients. CONCLUSION: Our results demonstrate that TNF and IL-6-induced osteoclasts may contribute to the pathology of inflammatory arthritis associated with joint destruction, such as RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Bone Resorption/immunology , Interleukin-6/immunology , Osteoclasts/immunology , Tumor Necrosis Factor-alpha/immunology , Aged , Arthritis, Rheumatoid/diagnostic imaging , Arthritis, Rheumatoid/metabolism , Bone Density , Bone Resorption/diagnostic imaging , Bone Resorption/metabolism , Case-Control Studies , Cytokines/drug effects , Cytokines/immunology , Cytokines/metabolism , Female , Humans , Interleukin-12 Subunit p40/drug effects , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Interleukin-1beta/drug effects , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-6/pharmacology , Male , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/immunology , Matrix Metalloproteinase 3/metabolism , Middle Aged , NFATC Transcription Factors/drug effects , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Osteogenesis/immunology , RANK Ligand/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
3.
J Cell Physiol ; 236(7): 5147-5161, 2021 07.
Article in English | MEDLINE | ID: mdl-33319933

ABSTRACT

Endothelial-to-mesenchymal transition (EndMT) and fibroblast-to-myofibroblast (FibroMF) differentiation are frequently reported in organ fibrosis. Stromelysin1, a matrix metalloprotease-3 (MMP3) has been indicated in vascular pathologies and organ injuries that often lead to fibrosis. In the current study, we investigated the role of stromelysin1 in EndMT and FibroMF differentiation, which is currently unknown. In our results, whereas TGFß2 treatment of endothelial cells (ECs) induced EndMT associated with increased expression of stromelysin1 and mesenchymal markers such as α-smooth muscle actin (αSMA), N-cadherin, and activin linked kinase-5 (ALK5), inhibition of stromelysin1 blunted TGFß2-induced EndMT. In contrast, treatment of NIH-3T3 fibroblasts with TGFß1 promoted FibroMF differentiation accompanied by increased expression of αSMA, N-cadherin, and ALK5. Intriguingly, stromelysin1 inhibition in TGFß1-stimulated myofibroblasts further exacerbated fibroproliferation with increased FibroMF marker expression. Gene Expression Omnibus (GEO) data analysis indicated increased stromelysin1 expression associated with EndMT and decreased stromelysin1 expression in human pulmonary fibrosis fibroblasts. In conclusion, our study has identified that EndMT and FibroMF differentiation are reciprocally regulated by stromelysin1.


Subject(s)
Cell Differentiation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Fibroblasts/cytology , Matrix Metalloproteinase 3/metabolism , Myofibroblasts/cytology , 3T3 Cells , Actins/biosynthesis , Animals , Antigens, CD/biosynthesis , Cadherins/biosynthesis , Cell Line , Endothelial Cells/metabolism , Fibrosis/pathology , Humans , Matrix Metalloproteinase 3/drug effects , Mice , Receptor, Transforming Growth Factor-beta Type I/biosynthesis , Transforming Growth Factor beta2/pharmacology
4.
Medicine (Baltimore) ; 99(43): e22841, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33120813

ABSTRACT

BACKGROUND: A large number of clinical studies have confirmed that after treatment with traditional Chinese medicine components such as sinomenine (SIN), the matrix -metalloproteinase3 (MMP-3) level of patients with rheumatoid arthritis (RA) shows a significant decrease, whereas MMP-3 can be involved in degrading bone matrix in humans, so in the progression of bone and joint injury in patients with RA, serum MMP-3 can be used as an important biochemical marker. The traditional Chinese medicine components commonly used in clinical practice include total glucosides of paeony (TGP), SIN, and tripterygium glycosides, which have the characteristics of disease-modifyinganti-rheumatic drugs and non-steroidal anti-inflammatory drugs, while they can reduce the toxic side effects of methotrexate (MTX), and their combination with other drugs such as MTX and leflunomide (HWA486) has become an important regimen for the treatment of RA in clinical practice. Therefore, we designed this study protocol to evaluate the adjuvant effect of commonly used traditional Chinese medicine components combined with MTX in the treatment of osteoarticular injury in RA. METHODS: The search time was set from January 2000 to September 2020 in this study. EMBASE database, Cochrane Library, PubMed, Web of Science, Science Direct, Chinese National Knowledge Infrastructure, China Biology Medicine disc (CBM), Chinese Scientifific Journals Database (VIP), and Wanfang Database were used as search sources to select the traditional Chinese medicine components that reduce MMP-3 and use MTX in the treatment of RA. Clinical randomized controlled trials were used, and inclusion criteria and exclusion criteria were set for screening. In this study, MMP-3, erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), cyclic peptide containing citrulline (CCP) and rheumatoid factor (RF) were used as the main outcomes, and the improvement of Disease Activity Score 28 (DAS28), joint bone mineral density, Clinical Disease Activity Index (CDAI), and other clinically relevant symptoms was selected as the secondary outcomes. Revman software version 5.3 was used for statistical analysis of data and risk assessment of deviation in this meta-analysis. In this study, one researcher performed study direction selection, literature inquiry, and literature download, and 2 independent reviewers performed literature data extraction and literature quality assessment. Dichotomized data are expressed as relative risk, continuous data are expressed as mean difference or standard mean difference, and finally fixed-effect model or random-effect model is used for synthesis according to the heterogeneity of data. RESULTS: To evaluate the effect of downregulation of MMP-3 level by traditional Chinese medicine components combined with MTX on the progression of bone injury in patients with RA by serum MMP-3, ESR, CRP, CCP, and RF. CONCLUSION: This study protocol can be used to evaluate the efficacy and safety of traditional Chinese medicine components combined with MTX in the treatment of bone injury in patients with RA. ETHICS AND DISSEMINATION: This study is a secondary study based on the published clinical research; therefore, approval from an ethics committee is not required for this study. In accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Protocol (PRISMA-P), the results of this study will be published in peer-reviewed scientific journals and conference papers. REGISTRATION NUMBER:: is INPLASY202090064.


Subject(s)
Antirheumatic Agents/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Matrix Metalloproteinase 3/drug effects , Methotrexate/therapeutic use , Bone and Bones/drug effects , Disease Progression , Down-Regulation/drug effects , Humans , Matrix Metalloproteinase 3/blood , Medicine, Chinese Traditional/methods , Systematic Reviews as Topic
5.
Aging (Albany NY) ; 12(3): 2246-2260, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32023553

ABSTRACT

The treatment for intervertebral disc degeneration (IDD) has drawn great attention and recent studies have revealed that the p38 MAPK pathway is a potential therapeutic target for delaying the degeneration of intervertebral discs. In this study, we analyzed a nature-derived protein tyrosine kinase inhibitor, Genistein, and its function in delaying IDD in rats both in vitro and in vivo via the p38 MAPK pathway. Nucleus pulposus cells treated with Genistein showed better function compared with untreated cells. Further study revealed that Genistein could play a protective role in IDD by inhibiting phosphorylation of p38, consequently inhibiting the p38 pathway-mediated inflammatory response. The rat IDD model also demonstrated that Genistein could effectively delay the degeneration of intervertebral disc tissue. The current study reveals new biological functions of Genistein, further demonstrates the effects of the p38 MAPK pathway on intervertebral disc degeneration, and deepens our understanding of the treatment and prevention of IDD.


Subject(s)
Genistein/pharmacology , Intervertebral Disc Degeneration/metabolism , Nucleus Pulposus/drug effects , Protein Kinase Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases/drug effects , Aggrecans/drug effects , Aggrecans/genetics , Animals , Cell Survival/drug effects , Collagen Type II/drug effects , Collagen Type II/genetics , Collagen Type X/drug effects , Collagen Type X/genetics , Inflammation , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Intervertebral Disc/cytology , Intervertebral Disc/drug effects , Intervertebral Disc/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Nucleus Pulposus/cytology , Nucleus Pulposus/metabolism , Phosphorylation , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Osteoarthritis Cartilage ; 28(5): 658-668, 2020 05.
Article in English | MEDLINE | ID: mdl-31734268

ABSTRACT

OBJECTIVE: Links between pain and joint degradation are poorly understood. We investigated the role of activation of Toll-like receptors (TLR) by cartilage metabolites in initiating and maintaining the inflammatory loop in OA causing joint destruction. METHODS: Synovial membrane explants (SMEs) were prepared from OA patients' synovial biopsies. SMEs were cultured for 10 days under following conditions: culture medium alone, OSM + TNFα, TLR2 agonist - Pam2CSK4, Pam3CSK4 or synthetic aggrecan 32-mer, TLR4 agonist - Lipid A. Release of pro-inflammatory and degradation biomarkers (acMMP3 and C3M) were measured by ELISA in conditioned media along with IL-6. Additionally, human cartilage was digested with ADAMTS-5, with or without the ADAMTS-5 inhibiting nanobody - M6495. Digested cartilage solution (DCS) and synthetic 32-mer were tested for TLR activation in SEAP based TLR reporter assay. RESULTS: Western blotting confirmed TLR2 and TLR4 in untreated OA synovial biopsies. TLR agonists showed an increase in release of biomarkers - acMMP3 and C3M in SME. Synthetic 32-mer showed no activation in the TLR reporter assay. ADAMTS-5 degraded cartilage fragments activated TLR2 in vitro. Adding M6495 - an anti-ADAMTS-5 inhibiting nanobody®, blocked ADAMTS-5-mediated DCS TLR2 activation. CONCLUSION: TLR2 is expressed in synovium of OA patients and their activation by synthetic ligands causes increased tissue turnover. ADAMTS-5-mediated cartilage degradation leads to release of aggrecan fragments which activates the TLR2 receptor in vitro. M6495 suppressed cartilage degradation by ADAMTS-5, limiting the activation of TLR2. In conclusion, pain and joint destruction may be linked to generation of ADAMTS-5 cartilage metabolites.


Subject(s)
ADAMTS5 Protein/metabolism , Cartilage, Articular/metabolism , Inflammation/metabolism , Synovial Membrane/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , ADAMTS5 Protein/drug effects , Aged , Aged, 80 and over , Aggrecans/metabolism , Blotting, Western , Cartilage, Articular/drug effects , Female , Humans , In Vitro Techniques , Interleukin-6/metabolism , Lipid A/pharmacology , Lipopeptides/pharmacology , Male , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Middle Aged , Oligopeptides/pharmacology , Single-Domain Antibodies/pharmacology , Synovial Membrane/drug effects , Toll-Like Receptor 2/agonists , Toll-Like Receptor 4/agonists , Toll-Like Receptor 9/agonists , Tumor Necrosis Factor-alpha/pharmacology
7.
Inflammation ; 42(4): 1456-1462, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30997585

ABSTRACT

Sudachitin, a polymethoxylated flavonoid found in the skin of Citrus sudachi, is a biologically active substance. The aim of this study was to examine whether sudachitin could be used to inhibit the expression of matrix metalloproteinase (MMP)-1 and MMP-3, which are involved in the destruction of periodontal tissues in periodontal lesions, in tumor necrosis factor (TNF)-α-stimulated human periodontal ligament cells (HPDLC). Sudachitin suppressed TNF-α-induced MMP-1 and MMP-3 production in HPDLC. On the other hand, it enhanced tissue inhibitor of metalloproteinase (TIMP)-1 expression. The level of Akt phosphorylation in the TNF-α-stimulated HPDLC was decreased by sudachitin treatment. Moreover, an Akt inhibitor reduced MMP-1 and MMP-3 production and increased TIMP-1 production. These findings indicate that sudachitin reduces MMP-1 and MMP-3 production in TNF-α-stimulated HPDLC by inhibiting the Akt pathway.


Subject(s)
Flavonoids/pharmacology , Glycosides/pharmacology , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 3/drug effects , Periodontal Ligament/cytology , Tumor Necrosis Factor-alpha/pharmacology , Anti-Infective Agents/pharmacology , Cells, Cultured , Humans , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 3/biosynthesis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Tissue Inhibitor of Metalloproteinase-1/drug effects
8.
Arthritis Rheumatol ; 71(2): 222-231, 2019 02.
Article in English | MEDLINE | ID: mdl-30079992

ABSTRACT

OBJECTIVE: To elucidate the role of the fractalkine (FKN)/CX3 CR1 pathway in joint destruction in rheumatoid arthritis. METHODS: We examined the effect of treatment with anti-mouse FKN (anti-mFKN) monoclonal antibody (mAb) on joint destruction and the migration of osteoclast precursors (OCPs) into the joint, using the collagen-induced arthritis (CIA) model. DBA/1 mice were immunized with bovine type II collagen to induce arthritis, and then treated with anti-mFKN mAb. Disease severity was monitored by arthritis score, and joint destruction was evaluated by soft x-ray and histologic analyses. Plasma levels of joint destruction markers were assessed by enzyme-linked immunosorbent assay. FKN expression on endothelial cells was detected by immunohistochemistry. Bone marrow-derived OCPs were labeled with fluorescein and transferred to mice with CIA, and the migration of the OCPs to the joints was then analyzed. RESULTS: Both prophylactic and therapeutic treatment with anti-mFKN mAb significantly decreased the arthritis and soft x-ray scores. Plasma levels of cartilage oligomeric matrix protein and matrix metalloproteinase 3 decreased after treatment with anti-mFKN mAb. Histologic analysis revealed that anti-mFKN mAb inhibited synovitis, pannus formation, and cartilage destruction, as well as suppressed bone damage, with a marked reduction in the number of tartrate-resistant acid phosphatase-positive osteoclasts. Anti-mFKN mAb strongly inhibited the migration of bone marrow-derived OCPs into the affected synovium. CONCLUSION: Anti-mFKN mAb notably ameliorates arthritis and joint destruction in the CIA model, as well as inhibits migration of OCPs into the synovium. These results suggest that inhibition of the FKN/CX3 CR1 pathway could be a novel strategy for treatment of both synovitis and joint destruction in rheumatoid arthritis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , CX3C Chemokine Receptor 1/immunology , Cell Movement/drug effects , Chemokine CX3CL1/antagonists & inhibitors , Osteoclasts/drug effects , Stem Cells/drug effects , Animals , Cartilage Oligomeric Matrix Protein/drug effects , Cartilage Oligomeric Matrix Protein/metabolism , Cartilage, Articular/drug effects , Cartilage, Articular/immunology , Cartilage, Articular/pathology , Chemokine CX3CL1/immunology , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Inbred DBA , Osteoclasts/metabolism , Synovial Membrane/drug effects , Synovial Membrane/immunology , Synovial Membrane/pathology , Synovitis/pathology , Tartrate-Resistant Acid Phosphatase/metabolism
9.
Cartilage ; 9(3): 293-303, 2018 07.
Article in English | MEDLINE | ID: mdl-29986604

ABSTRACT

Objective Early intervention therapies targeting inflammation and cell death during the acute phase of cartilage injury have the potential to prevent posttraumatic osteoarthritis. The objective of this study was to investigate the effects of interleukin receptor antagonist protein (IRAP), hyaluronan (HA), dexamethasone (DEX), and mesenchymal stem cell (MSC) treatment on the expression of established genetic markers for matrix degradation, apoptosis, and inflammation in articular cartilage during the acute phase of injury. Design A custom impact device was used to create replicable injury ex vivo to intact porcine knee joint. One hour after impact, IRAP, HA, DEX, or MSCs was intra-articularly injected. At 8 hours postinjury, cartilage and meniscus samples were harvested for genetic expression analysis. Expression of miR-27b, miR-140, miR-125b, miR-16, miR-34a, miR-146a, miR-22, ADAMTS-4, ADAMTS-5, MMP-3, IL-1ß, and TNF-α was analyzed by real-time polymerase chain reaction. Results At 8 hours postinjury, expression of ADAMTS-4, ADAMTS-5, MMP-3, IL-1ß, and TNF-α in cartilage was significantly decreased in IRAP- and DEX-treated joints as compared to nontreated injured joints, whereas only IRAP upregulated expression of miR-140, miR-125b, miR-27b, miR-146a, and miR-22 in cartilage. HA and MSC treatments had no significant effects on catabolic and inflammatory gene expression in cartilage. However, HA treatment significantly upregulated expression of all miRNAs except miR-16. In addition, the treatments tested also exhibited significant influences on meniscus. Conclusions This study provides a valuable starting point for further research into potential targets for and efficacy of various early intervention strategies that may delay or prevent the progression of posttraumatic osteoarthritis after acute cartilage injury.


Subject(s)
Anti-Inflammatory Agents/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Osteoarthritis, Knee/genetics , ADAMTS4 Protein/drug effects , ADAMTS4 Protein/genetics , ADAMTS5 Protein/drug effects , ADAMTS5 Protein/genetics , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/injuries , Cell Death/drug effects , Cells, Cultured/metabolism , Chondrocytes/drug effects , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Gene Expression , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/therapeutic use , Inflammation/metabolism , Injections, Intra-Articular/methods , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/genetics , Meniscus/drug effects , Meniscus/metabolism , Mesenchymal Stem Cell Transplantation/methods , MicroRNAs/genetics , Models, Animal , Osteoarthritis, Knee/prevention & control , Receptors, Interleukin/antagonists & inhibitors , Receptors, Interleukin/therapeutic use , Swine , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics
10.
Chin J Dent Res ; 20(3): 125-135, 2017.
Article in English | MEDLINE | ID: mdl-28808696

ABSTRACT

OBJECTIVE: To explore the effect of long-term stress on the temporomandibular joint (TMJ) condyle and its possible underlying mechanism. METHODS: A 12-week, chronic unpredictable mild stress (CUMS) model was used to induce long-term psychological stress in rats. Rats were randomly divided into control group (CONT), chronic unpredictable mild stress group (CUMS) and chronic unpredictable mild stress with fluoxetine treatment group (CUMS + DT) (n = 30 per group). A 5 mg/kg dose of fluoxetine was intraperitoneally injected daily 0.5 h before stress. A sucrose preference test, plasma corticosterone test and open-field test were performed to verify the feasibility of the CUMS model. Histopathology was used to observe the pathological changes of condyle. The expression levels of inflammatory cytokines, matrix metalloproteases (MMPs) and extracellular matrix (ECM) were measured by real-time polymerase chain reaction, western blotting and immunohistochemistry. RESULTS: At 8 and 12 weeks after exposure to CUMS, the rats showed higher plasma corticosterone than the control rats. Additionally, for the open-field test, the rats exposed to CUMS spent more time in the centre zone and moved a shorter distance than the control and drug treatment rats. In addition, pathological changes in the condylar cartilage occurred in the 8-week CUMS subgroup and were more obvious in the 12-week CUMS subgroup. The CUMS caused an increase in the secretion of inflammatory cytokines, imbalanced expression of MMPs and tissue inhibitor of metalloproteinase-1 and accelerated degradation of ECM in condylar cartilage in a time-dependent manner. CONCLUSION: Osteoarthritis-like lesions can be caused by long-term CUMS in the mandibular condyles, which suggests that the imbalance in chondrocyte-secreted regulatory factors within the cartilage of the TMJ may play an important role in cartilage injury induced by psychological stress.


Subject(s)
Cartilage, Articular/metabolism , Extracellular Matrix/metabolism , Mandibular Condyle/metabolism , Stress, Psychological/metabolism , Aggrecans/drug effects , Aggrecans/genetics , Aggrecans/metabolism , Animals , Behavior, Animal , Blotting, Western , Cartilage, Articular/drug effects , Collagen Type II/drug effects , Collagen Type II/genetics , Collagen Type II/metabolism , Corticosterone/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/drug effects , Fluoxetine/pharmacology , Immunohistochemistry , Male , Mandibular Condyle/drug effects , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Psychological/genetics , Temporomandibular Joint , Tissue Inhibitor of Metalloproteinase-1/drug effects , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism
11.
Rheumatology (Oxford) ; 56(8): 1417-1427, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28431044

ABSTRACT

Objective: To explore the role of leonurine in the regulation of synovial inflammation and joint destruction inRA. Methods: Fibroblast-like synoviocytes were isolated from synovial tissue from RA patients. Pro-inflammatory cytokine and MMP expression was evaluated using real-time PCR and a cytometric bead array. Cell migration and invasion in vitro were measured using the Boyden chamber method and the scratch assay, respectively. Protein expression was measured by western blotting. Nuclear factor kappa B (NF-κB) nuclear translocation was detected by immunofluorescence. The in vivo effect of leonurine was evaluated in mice with CIA. Results: Leonurine treatment significantly decreased the production of pro-inflammatory cytokines (IL-1ß, IL-6, IL-8 and TNFα) and MMPs (MMP-1 and MMP-3) and suppressed the migration and invasion of RA fibroblast-like synoviocytes. The molecular analysis revealed that leonurine impaired TNFα-induced NF-κB signalling by inhibiting the phosphorylation and degradation of inhibitor of NF-κB alpha (IκBα) and subsequently preventing the nuclear translocation of the NF-κB p65 subunit. Leonurine also inhibited the p38 and Jun N-terminal kinase mitogen-activated protein kinases signalling pathways without affecting ERK signalling. Intraperitoneal injection of leonurine reduced synovial inflammation, joint destruction and the serum IL-1ß, IL-6 and TNFα levels in mice with CIA. Conclusion: Our findings show that leonurine reduces synovial inflammation and joint destruction in RA through the NF-κB and mitogen-activated protein kinases pathways. Leonurine has potential as a therapeutic agent for RA.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Gallic Acid/analogs & derivatives , Adult , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Cytokines/drug effects , Female , Fibroblasts/metabolism , Gallic Acid/pharmacokinetics , Humans , Inflammation Mediators/metabolism , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 3/drug effects , Mice , Middle Aged , Mitogen-Activated Protein Kinases/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects , Synovial Membrane/drug effects , Synoviocytes/drug effects , Synoviocytes/pathology , Transcription Factor RelA/drug effects
12.
Rheumatology (Oxford) ; 56(3): 351-361, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27940589

ABSTRACT

Objective: A crucial feature of OA is cartilage degradation. This process is mediated by pro-inflammatory cytokines, among other factors, via induction of matrix-degrading enzymes. Interleukin 37 (IL37) is an anti-inflammatory cytokine and is efficient in blocking the production of pro-inflammatory cytokines during innate immune responses. We hypothesize that IL37 is therapeutic in treating the inflammatory cytokine cascade in human OA chondrocytes and can act as a counter-regulatory cytokine to reduce cartilage degradation in OA. Methods: Human OA cartilage was obtained from patients undergoing total knee or hip arthroplasty. Immunohistochemistry was applied to study IL37 protein expression in cartilage biopsies from OA patients. Induction of IL37 expression by IL1ß, OA synovium-conditioned medium and TNFα was investigated in human OA chondrocytes. Adenoviral overexpression of IL37 followed by IL1ß stimulation was performed to investigate the anti-inflammatory potential of IL37. Results: IL37 expression was detected in cartilage biopsies of OA patients and induced by IL1ß. After IL1ß stimulation, increased IL1ß, IL6 and IL8 expression was observed in OA chondrocytes. Elevated IL37 levels diminished the IL1ß-induced IL1ß , IL6 and IL8 gene levels and IL1ß and IL8 protein levels. In addition to the reduction in pro-inflammatory cytokine expression, IL37 reduced MMP1 , MMP3 , MMP13 and disintegrin and metalloproteinase with thrombospondin motifs 5 gene levels and MMP3 and MMP13 protein levels. Conclusion: IL37 is induced by IL1ß, and IL37 itself reduced IL1ß, IL6 and IL8 production, indicating that IL37 is able to induce a counter-regulatory anti-inflammatory feedback loop in chondrocytes. In addition, IL37 dampens catabolic enzyme expression. This supports IL37 as a potential therapeutic target in OA.


Subject(s)
Chondrocytes/metabolism , Interleukin-1/metabolism , Interleukin-1beta/pharmacology , Osteoarthritis , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Adenoviridae , Blotting, Western , Chondrocytes/drug effects , Disintegrins/drug effects , Disintegrins/genetics , Disintegrins/metabolism , Humans , Immunohistochemistry , Interleukin-1/genetics , Interleukin-1beta/drug effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/drug effects , Interleukin-8/genetics , Interleukin-8/metabolism , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 13/drug effects , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , RNA, Messenger/drug effects , Real-Time Polymerase Chain Reaction
13.
Arch Oral Biol ; 75: 89-99, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27825679

ABSTRACT

OBJECTIVES: Elevated levels of matrix metalloproteinases (MMPs) have been associated with the active phases of tissue and bone destruction in periodontitis, an inflammatory disease characterized by a significant breakdown of tooth support. In the present study, we used a three-dimensional (3D) co-culture model of macrophages and gingival fibroblasts to investigate the ability of a green tea extract and its major constituent epigallocatechin-3-gallate (EGCG) to regulate the secretion of MMP-3, -8, and -9. METHODS: The 3D co-culture model was composed of gingival fibroblasts embedded in a type I collagen matrix overlaid with macrophages. Two arbitrary ratios were tested. The ratio composed of 1 macrophage to 10 fibroblasts was used to mimic a slightly inflamed periodontal site while the ratio composed of 10 macrophages to 1 fibroblast was used to mimic a severely inflamed periodontal site. The 3D co-culture model was pre-treated for 2h with either the green tea extract or EGCG. It was then stimulated with Aggregatibacter actinomycetemcomitans lipopolysaccharide (LPS). The model was also first stimulated with LPS for 2h and then incubated with the green tea extract or EGCG. The concentrations of secreted MMP-3, -8, and -9 were quantified by enzyme-linked immunoassays. RESULTS: When the 3D co-culture model was stimulated with A. actinomycetemcomitans LPS, the 10:1 ratio of macrophages to gingival fibroblasts was associated with a highest secretion of MMP-3 and -9 and, to a lesser extent, MMP-8, than the 1:10 ratio. Non-cytotoxic concentrations of the green tea extract or EGCG reduced the basal secretion levels of all three MMPs. A 2-h treatment with the green tea extract or EGCG prior to the stimulation with LPS resulted in a dose-dependent decrease in MMP secretion, with MMP-9 showing the most significant decrease. A decrease in MMP secretion was also observed when the green tea extract or EGCG was added following a 2-h stimulation with LPS. CONCLUSIONS: Our results suggested that green tea catechins, and more specifically EGCG, offer promising prospects for the development of a novel adjunctive treatment for periodontitis because of their ability to decrease the secretion of MMPs, which are important tissue-destructive enzymes produced by mucosal and immune cells.


Subject(s)
Catechin/pharmacology , Coculture Techniques/methods , Fibroblasts/metabolism , Macrophages/metabolism , Matrix Metalloproteinases/drug effects , Matrix Metalloproteinases/metabolism , Plant Extracts/pharmacology , Tea/chemistry , Aggregatibacter actinomycetemcomitans/chemistry , Catechin/analogs & derivatives , Cell Line , Cell Survival/drug effects , Collagen Type I/chemistry , Humans , Lipopolysaccharides/isolation & purification , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 8/drug effects , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Periodontitis/drug therapy , Plant Extracts/chemistry , U937 Cells
14.
J Diabetes Res ; 2016: 3765173, 2016.
Article in English | MEDLINE | ID: mdl-27635402

ABSTRACT

Objectives. To investigate whether high glucose-induced oxidative stress is implicated in apoptosis of rat nucleus pulposus cells (NPCs) and abnormal expression of critical genes involved in the metabolic balance of extracellular matrix (ECM). Methods. NPCs were cultured with various concentrations of glucose to detect cell viability and apoptosis. Cells cultured with high glucose (25 mM) were untreated or pretreated with N-acetylcysteine or a p38 MAPK inhibitor SB 202190. Reactive oxygen species (ROS) production was evaluated. Activation of p38 MAPK was measured by Western blot. The expression of ECM metabolism-related genes, including type II collagen, aggrecan, SRY-related high-mobility-group box 9 (Sox-9), matrix metalloproteinase 3 (MMP-3), and tissue inhibitor of metalloproteinase 1 (TIMP-1), was analyzed by semiquantitative RT-PCR. Results. High glucose reduced viability of NPCs and induced apoptosis. High glucose resulted in increased ROS generation and p38 MAPK activation. In addition, it negatively regulated the expression of type II collagen, aggrecan, Sox-9, and TIMP-1 and positively regulated MMP-3 expression. These results were changed by pretreatment with N-acetylcysteine or SB 202190. Conclusions. High glucose might promote apoptosis of NPCs, trigger ECM catabolic pathways, and inhibit its anabolic activities, possibly through a p38 MAPK-dependent oxidative stress mechanism.


Subject(s)
Apoptosis/drug effects , Extracellular Matrix/drug effects , Glucose/pharmacology , Nucleus Pulposus/drug effects , Oxidative Stress/drug effects , RNA, Messenger/drug effects , p38 Mitogen-Activated Protein Kinases/drug effects , Acetylcysteine/pharmacology , Aggrecans/drug effects , Aggrecans/genetics , Animals , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Collagen Type II/drug effects , Collagen Type II/genetics , Extracellular Matrix/metabolism , Free Radical Scavengers/pharmacology , Humans , Hyperglycemia/genetics , Hyperglycemia/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/genetics , Nucleus Pulposus/cytology , Nucleus Pulposus/metabolism , RNA, Messenger/metabolism , Rats , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SOX9 Transcription Factor/drug effects , SOX9 Transcription Factor/genetics , Tissue Inhibitor of Metalloproteinase-1/drug effects , Tissue Inhibitor of Metalloproteinase-1/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
J Cosmet Dermatol ; 15(1): 72-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26799467

ABSTRACT

Sulforaphane (SFN), a natural compound occurring in cruciferous vegetables, has been known for years as a chemopreventive agent against many types of cancer. Recently, it has been investigated as an antioxidant and anti-aging agent, and interesting conclusions have been made over the last decade. SFN demonstrated protective effects against ultraviolet (UV)-induced skin damage through several mechanisms of action, for example, decrease of reactive oxygen species production, inhibition of matrix metalloproteinase expression, and induction of phase 2 enzymes. SFN used as a protective agent against UV damage is a whole new matter, and it seems to be a very promising ingredient in upcoming anti-aging drugs and cosmetics.


Subject(s)
Anticarcinogenic Agents/pharmacology , Isothiocyanates/pharmacology , Signal Transduction/drug effects , Skin Aging/drug effects , Skin/drug effects , Humans , Kelch-Like ECH-Associated Protein 1/drug effects , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 3/drug effects , NF-E2-Related Factor 2/drug effects , NF-kappa B/drug effects , Radiation-Protective Agents/pharmacology , Skin/radiation effects , Skin Aging/radiation effects , Sulfoxides , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/radiation effects , Ultraviolet Rays/adverse effects
16.
Arthritis Rheumatol ; 68(1): 152-63, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26360647

ABSTRACT

OBJECTIVE: Both alarmins S100A8/A9 and canonical Wnt signaling have been found to play active roles in the development of experimental osteoarthritis (OA). However, what activates canonical Wnt signaling remains unknown. This study was undertaken to investigate whether S100A8 induces canonical Wnt signaling and whether S100 proteins exert their effects via activation of Wnt signaling. METHODS: Expression of the genes for S100A8/A9 and Wnt signaling pathway members was measured in an experimental OA model. Selected Wnt signaling pathway members were overexpressed, and levels of S100A8/A9 were measured. Activation of canonical Wnt signaling was determined after injection of S100A8 into naive joints and induction of collagenase-induced OA in S100A9-deficient mice. Expression of Wnt signaling pathway members was tested in macrophages and fibroblasts after S100A8 stimulation. Canonical Wnt signaling was inhibited in vivo to determine if the effects of S100A8 injections were dependent on Wnt signaling. RESULTS: The alarmins S100A8/A9 and members of the Wnt signaling pathway showed coinciding expression in synovial tissue in an experimental OA model. Synovial overexpression of selected Wnt signaling pathway members did not result in increased expression of S100 proteins. In contrast, intraarticular injection of S100A8 increased canonical Wnt signaling, whereas canonical Wnt signaling was decreased after induction of experimental OA in S100A9-deficient mice. S100A8 stimulation of macrophages, but not fibroblasts, resulted in increased expression of canonical Wnt signaling members. Overexpression of Dkk-1 to inhibit canonical Wnt signaling decreased the induction of matrix metalloproteinase 3, interleukin-6, and macrophage inflammatory protein 1α after injection of S100A8. CONCLUSION: Our findings indicate that the alarmin S100A8 induces canonical Wnt signaling in macrophages and murine knee joints. The effects of S100A8 are partially dependent on activation of canonical Wnt signaling.


Subject(s)
Arthritis, Experimental/genetics , Calgranulin A/genetics , Calgranulin B/genetics , Macrophages/metabolism , Osteoarthritis, Knee/genetics , Stifle/metabolism , Synovial Membrane/metabolism , Wnt Signaling Pathway/genetics , Alarmins/pharmacology , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/metabolism , Chemokine CCL3/drug effects , Chemokine CCL3/metabolism , Collagenases/toxicity , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Interleukin-6/metabolism , Macrophages/drug effects , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Knockout , Osteoarthritis, Knee/metabolism , Real-Time Polymerase Chain Reaction
17.
Surg Endosc ; 30(5): 2016-21, 2016 05.
Article in English | MEDLINE | ID: mdl-26264696

ABSTRACT

BACKGROUND: Ventral hernia is a commonly occurring surgical problem. Our earlier studies have shown that a 30 mg/kg dose of doxycycline can significantly impact the strength of polypropylene (PP) mesh in a rat hernia repair model at 6 and 12 weeks. The objective of the present study was to investigate the dose dependence of doxycycline treatment on hernia repair strengths in rats. STUDY DESIGN: Fifty-six Sprague-Dawley rats underwent hernia repair with either PP mesh (n = 28) or sutures only (primary; n = 28); both groups were further divided into four doxycycline groups of seven animals each: control (0 mg/kg), low (3 mg/kg), medium (10 mg/kg), and high (30 mg/kg). One day before hernia repair surgery, animals received doxycycline doses by gavage and continued receiving daily until euthanasia. After 8 weeks, rats were euthanized and tissue samples from hernia repaired area were collected and analyzed for tensile strength using a tensiometer (Instron, Canton, MA, USA), while MMPs 2, 3, and 9, and collagen type 1 and 3 were analyzed by western blotting. RESULTS: In mesh-repaired animals, medium and high doxycycline dose repaired mesh fascia interface (MFI) showed significant increase in tensile strength when compared to control. In the primary repaired animals, there was no significant difference in MFI tensile strength in any dose group. In medium-dose MFI, there was a significant reduction in MMPs 2, 3, and 9. In this animal group, MFI showed significant increase in collagen 1 and significant reduction in collagen type 3 when compared to control. CONCLUSION: It is possible to improve the strength of mesh-repaired tissue by administering a significantly lower dose of the drug, which has implications for translation of the findings.


Subject(s)
Anti-Bacterial Agents/pharmacology , Doxycycline/pharmacology , Fascia/drug effects , Hernia, Ventral/surgery , Herniorrhaphy/methods , Surgical Mesh , Tensile Strength/drug effects , Animals , Blotting, Western , Collagen Type I/drug effects , Collagen Type I/metabolism , Collagen Type III/drug effects , Collagen Type III/metabolism , Dose-Response Relationship, Drug , Fascia/metabolism , Male , Matrix Metalloproteinase 2/drug effects , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/metabolism , Polypropylenes , Prostheses and Implants , Rats , Rats, Sprague-Dawley , Sutures
18.
Chin J Nat Med ; 13(4): 283-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25908625

ABSTRACT

This study investigated effects of Ginsenoside Ro (Ro) on interleukin-1ß (IL-1ß)-induced apoptosis and inflammation in rat chondrocytes. The rat chondrocytes were co-treated with IL-1ß (10 ng·kg(-1)) and Ro (50, 100 and 200 µmol·L(-1)) for 48 h. Chondrocytes viability was detected by the MTT assay and Annexin V-FITC/PI dual staining assay. Caspase 3 activity was measured by using caspase 3 colorimetric assay kit. Apoptosis related proteins Bax, Bad, Bcl-xL, PCNA, p53 and phospho-p53, along with inflammation related protein MMP 3, MMP 9 and COX-2, and the expression of phospho-NF-κB p65 were assayed by western blotting analyses. Ro could improve IL-1ß-induced chondrocytes viability. Ro could suppress IL-1ß-induced apoptosis by inhibiting levels of Bax and Bad, decreasing p53 phosphorylation and promoting the expression of Bcl-xL and PCNA. Ro inhibited caspase 3 activity. IL-1ß-induced inflammation and matrix degration were also alleviated by Ro with down-regulating the expression of MMP 3, MMP 9 and COX-2. Moreover, Ro inhibited NF-κB p65 phosphorylation induced by IL-1ß. In conclusion, these results suggested Ro exerted anti-apoptosis and anti-inflammation in IL-1ß-induced rat chondrocytes, which might be related to NF-κB signal pathway. Therefore, we propose that Ro might be a potential novel drug for the treatment of osteoarthritis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Chondrocytes/drug effects , Ginsenosides/pharmacology , Inflammation/drug therapy , Interleukin-1beta/pharmacology , NF-kappa B/antagonists & inhibitors , Animals , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Cell Survival/drug effects , Chondrocytes/cytology , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Inflammation/chemically induced , Interleukin-1beta/antagonists & inhibitors , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 9/drug effects , NF-kappa B/drug effects , NF-kappa B/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
19.
Ann Rheum Dis ; 74(6): 1311-6, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25398374

ABSTRACT

OBJECTIVE: Tofacitinib is an oral Janus kinase (JAK) inhibitor for the treatment of rheumatoid arthritis (RA). The pathways affected by tofacitinib and the effects on gene expression in situ are unknown. Therefore, tofacitinib effects on synovial pathobiology were investigated. METHODS: A randomised, double-blind, phase II serial synovial biopsy study (A3921073; NCT00976599) in patients with RA with an inadequate methotrexate response. Patients on background methotrexate received tofacitinib 10 mg twice daily or placebo for 28 days. Synovial biopsies were performed on Days -7 and 28 and analysed by immunoassay or quantitative PCR. Clinical response was determined by disease activity score and European League Against Rheumatism (EULAR) response on Day 28 in A3921073, and at Month 3 in a long-term extension study (A3921024; NCT00413699). RESULTS: Tofacitinib exposure led to EULAR moderate to good responses (11/14 patients), while placebo was ineffective (1/14 patients) on Day 28. Tofacitinib treatment significantly reduced synovial mRNA expression of matrix metalloproteinase (MMP)-1 and MMP-3 (p<0.05) and chemokines CCL2, CXCL10 and CXCL13 (p<0.05). No overall changes were observed in synovial inflammation score or the presence of T cells, B cells or macrophages. Changes in synovial phosphorylation of signal transducer and activator of transcription 1 (STAT1) and STAT3 strongly correlated with 4-month clinical responses (p<0.002). Tofacitinib significantly decreased plasma CXCL10 (p<0.005) at Day 28 compared with placebo. CONCLUSIONS: Tofacitinib reduces metalloproteinase and interferon-regulated gene expression in rheumatoid synovium, and clinical improvement correlates with reductions in STAT1 and STAT3 phosphorylation. JAK1-mediated interferon and interleukin-6 signalling likely play a key role in the synovial response. TRIAL REGISTRATION NUMBER: NCT00976599.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Janus Kinase 1/drug effects , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , RNA, Messenger/drug effects , STAT Transcription Factors/drug effects , Synovial Membrane/drug effects , Adult , Aged , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/metabolism , Chemokines/drug effects , Chemokines/genetics , Chemokines/metabolism , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Janus Kinase 1/metabolism , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Methotrexate/therapeutic use , Middle Aged , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , RNA, Messenger/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Synovial Membrane/metabolism , Treatment Outcome
20.
Rheumatology (Oxford) ; 54(6): 1087-92, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25389358

ABSTRACT

OBJECTIVE: Glucocorticoids are powerful anti-inflammatory compounds that also induce the expression of leptin and leptin receptor (Ob-R) in synovial fibroblasts through TGF-ßsignalling and Smad1/5 phosphorylation. Compound A (CpdA), a selective glucocorticoid receptor agonist, reduces inflammation in murine arthritis models and does not induce diabetes or osteoporosis, thus offering an improved risk:benefit ratio in comparison with glucocorticoids. Due to the detrimental role of leptin in OA pathogenesis, we sought to determine whether CpdA also induced leptin and Ob-R protein expression as observed with prednisolone. METHODS: Human synovial fibroblasts and chondrocytes were isolated from the synovium and cartilage of OA patients after joint surgery. The cells were treated with prednisolone, TGF-ß1, TNF-α and/or CpdA. Levels of leptin, IL-6, IL-8, MMP-1 and MMP-3 were measured by ELISA and expression levels of Ob-R phospho-Smad1/5, phospho-Smad2, α-tubulin and glyceraldehyde 3-phosphate dehydrogenase were analysed by western blotting. RESULTS: CpdA, unlike prednisolone, did not induce leptin secretion or Ob-R protein expression in OA synovial fibroblasts. Moreover, CpdA decreased endogenous Ob-R expression and down-regulated prednisolone-induced leptin secretion and Ob-R expression. Mechanistically, CpdA, unlike prednisolone, did not induce Smad1/5 phosphorylation. CpdA, similarly to prednisolone, down-regulated endogenous and TNF-α-induced IL-6, IL-8, MMP-1 and MMP-3 protein secretion. The dissociative effect of CpdA was confirmed using chondrocytes with no induction of leptin secretion, but with a significant decrease in IL-6, IL-8, MMP-1 and MMP-3 protein secretion. CONCLUSION: CpdA, unlike prednisolone, did not induce leptin or Ob-R in human OA synovial fibroblasts, thereby demonstrating an improved risk:benefit ratio.


Subject(s)
Chondrocytes/metabolism , Fibroblasts/metabolism , Osteoarthritis/metabolism , Prednisolone/pharmacology , Receptors, Glucocorticoid/agonists , Synovial Membrane/metabolism , Aged , Aged, 80 and over , Blotting, Western , Chondrocytes/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/drug effects , Interleukin-8/metabolism , Leptin/metabolism , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Middle Aged , Receptors, Leptin/drug effects , Receptors, Leptin/metabolism , Smad Proteins, Receptor-Regulated/drug effects , Smad Proteins, Receptor-Regulated/metabolism , Synovial Membrane/drug effects , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/metabolism , Tubulin/drug effects , Tubulin/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...