Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Osteoarthritis Cartilage ; 25(8): 1353-1361, 2017 08.
Article En | MEDLINE | ID: mdl-28365462

OBJECTIVE: Arthroscopy with lavage and synovectomy can remove tissue debris from the joint space and the synovial lining to provide pain relief to patients with osteoarthritis (OA). Here, we developed an in vitro model to study the interaction of cartilage wear particles with fibroblast-like synoviocytes (FLS) to better understand the interplay of cartilage particulates with cytokines on cells of the synovium. METHOD: In this study sub-10 µm cartilage particles or 1 µm latex particles were co-cultured with FLS ±10 ng/mL interleukin-1α (IL-1α) or tumor necrosis factor-α (TNF-α). Samples were analyzed for DNA, glycosaminoglycan (GAG), and collagen, and media samples were analyzed for media GAG, nitric oxide (NO) and prostaglandin-E2 (PGE2). The nature of the physical interaction between the particles and FLS was determined by microscopy. RESULTS: Both latex and cartilage particles could be phagocytosed by FLS. Cartilage particles were internalized and attached to the surface of both dense monolayers and individual cells. Co-culture of FLS with cartilage particulates resulted in a significant increase in cell sheet DNA and collagen content as well as NO and PGE2 synthesis compared to control and latex treated groups. CONCLUSION: The proliferative response of FLS to cartilage wear particles resulted in an overall increase in extracellular matrix (ECM) content, analogous to the thickening of the synovial lining observed in OA patients. Understanding how cartilage particles interface with the synovium may provide insight into how this interaction contributes to OA progression and may guide the role of lavage and synovectomy for degenerative disease.


Cartilage , Latex , Synovial Membrane/chemistry , Synovitis/pathology , Animals , Cattle , Cells, Cultured , Cytokines/pharmacology , Fibroblasts/physiology , Models, Biological , Phagocytosis/physiology
2.
Curr Issues Mol Biol ; 4(4): 129-46, 2002 Oct.
Article En | MEDLINE | ID: mdl-12432964

Human and other annotated genome sequences have facilitated generation of vast amounts of correlative data, from human/animal genetics, normal and disease-affected tissues from complex diseases such as arthritis using gene/protein chips and SNP analysis. These data sets include genes/proteins whose functions are partially known at the cellular level or may be completely unknown (e.g. ESTs). Thus, genomic research has transformed molecular biology from "data poor" to "data rich" science, allowing further division into subpopulations of subcellular fractions, which are often given an "-omic" suffix. These disciplines have to converge at a systemic level to examine the structure and dynamics of cellular and organismal function. The challenge of characterizing ESTs linked to complex diseases is like interpreting sharp images on a blurred background and therefore requires a multidimensional screen for functional genomics ("functionomics") in tissues, mice and zebra fish model, which intertwines various approaches and readouts to study development and homeostasis of a system. In summary, the post-genomic era of functionomics will facilitate to narrow the bridge between correlative data and causative data by quaint hypothesis-driven research using a system approach integrating "intercoms" of interacting and interdependent disciplines forming a unified whole as described in this review for Arthritis.


Computational Biology , Gene Expression Profiling , Genomics , Osteoarthritis/genetics , Animals , Cartilage/metabolism , Cluster Analysis , Collagen/genetics , Collagen/metabolism , Cytokines/metabolism , Endopeptidases/metabolism , Fibronectins/metabolism , Genome , Humans , Oligonucleotide Array Sequence Analysis , Osteopontin , Risk Factors , Sialoglycoproteins/metabolism
4.
Arthritis Rheum ; 44(3): 578-84, 2001 Mar.
Article En | MEDLINE | ID: mdl-11263772

OBJECTIVE: To identify extracellular and intraarticular matrix components that are differentially expressed in normal and osteoarthritis (OA)-affected cartilage and to investigate their functions with respect to regulation of mediators of inflammation. METHODS: Differential-display reverse transcriptase-polymerase chain reaction (RT-PCR) analysis of a pool of messenger RNA (mRNA) from 10 human OA cartilage samples and 5 normal cartilage samples was performed using arbitrary primers. Confirmatory analysis of the up-regulated transcripts of fibronectin (FN) and osteopontin (OPN) was performed by RT-PCR of individual RNA samples from a separate set of donors. The effect of recombinant OPN (or anti-OPN antiserum) on chondrocyte function was examined by analyzing the spontaneous or interleukin-1 (IL-1)-induced release of nitric oxide (NO) and prostaglandin E2 (PGE2) from human OA-affected cartilage under ex vivo conditions. RESULTS: Up-regulation (300-700%) of FN and OPN mRNA was observed in human OA-affected cartilage as compared with normal cartilage. Functional analysis of the role of OPN in OA cartilage showed that 1) Addition of 1 microg/ml (20 nM) of recombinant OPN to human OA-affected cartilage under ex vivo conditions inhibited spontaneous and IL-1beta-induced NO and PGE2 production, and 2) neutralization of intraarticular OPN with anti-OPN antiserum augmented NO production. CONCLUSION: The data indicate that one of the functions of intraarticular OPN, which is overexpressed in OA cartilage, is to act as an innate inhibitor of IL-1, NO, and PGE2 production. These findings suggest that the production of pleiotropic mediators of inflammation that influence cartilage homeostasis, such as NO and PGE2, is regulated by the interaction of chondrocytes with differentially expressed proteins within the extracellular matrix.


Sialoglycoproteins/pharmacology , Aged , Cartilage, Articular/chemistry , Cartilage, Articular/drug effects , Humans , Inflammation Mediators/antagonists & inhibitors , Middle Aged , Osteoarthritis/metabolism , Osteopontin , RNA, Messenger/isolation & purification
5.
J Biol Chem ; 275(51): 40307-15, 2000 Dec 22.
Article En | MEDLINE | ID: mdl-11007768

Interleukin 1 (IL-1), produced by both synovial cells and chondrocytes, plays a pivotal role in the pathogenesis of cartilage destruction in osteoarthritis (OA). We examined the specific expression and function of IL-1 receptor family-related genes in human joint tissues. Gene array analysis of human normal and OA-affected cartilage showed mRNA expression of IL-1 receptor accessory protein (IL-1RAcp) and IL-1 type I receptor (IL-1RI), but not IL-1 antagonist (IL-1ra) and IL-1 type II decoy receptor (IL-1RII). Similarly, human synovial and epithelial cells showed an absence of IL-1RII mRNA. Functional genomic analyses showed that soluble (s) IL-1RII, at picomolar concentrations, but not soluble TNF receptor:Fc, significantly inhibited IL-1beta-induced nitric oxide (NO) and/or prostaglandin E(2) production in chondrocytes, synovial and epithelial cells. In OA-affected cartilage, the IC(50) for inhibition of NO production by sIL-1RII was 2 log orders lower than that for sIL-1RI. Human chondrocytes that overexpressed IL-1RII were resistant to IL-1-induced IL-1beta mRNA accumulation and inhibition of proteoglycan synthesis. In osteoarthritis, deficient expression by chondrocytes of innate regulators or antagonists of IL-1 such as IL-1ra and IL-1RII (soluble or membrane form) may allow the catabolic effects of IL-1 to proceed unopposed. The sensitivity of IL-1 action to inhibition by sIL-1RII has therapeutic implications that could be directed toward correcting this unfavorable tissue(s) dependent imbalance.


Interleukin-1/physiology , Osteoarthritis/physiopathology , Receptors, Interleukin-1/physiology , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/physiopathology , Cattle , Chondrocytes/metabolism , Chondrocytes/pathology , Dinoprostone/biosynthesis , Gene Expression Regulation , Humans , Interleukin-1/genetics , Nitric Oxide/biosynthesis , Osteoarthritis/drug therapy , Proteoglycans/biosynthesis , Synovial Membrane/metabolism
6.
Inflamm Res ; 49(1): 20-6, 2000 Jan.
Article En | MEDLINE | ID: mdl-10778917

OBJECTIVE AND DESIGN: Cyclosporin, FK-506 and rapamycin have similar but distinct modes of interaction with cyclophilins, calcineurins and transcription factors. These immunosuppressive drugs have also been shown to inhibit cytotoxic and inflammatory responses in macrophage. Therefore, we evaluated the mechanism of action of these drugs on iNOS and COX-2 expression by macrophages, the products of which (NO and PGE2) have cytotoxic and proinflammatory activities. MATERIALS AND METHODS: The murine macrophage cell line RAW 264.7 was grown as monolayer cultures. The effects of pharmacologically relevant concentrations of cyclosporin, rapamycin and FK-506 were evaluated in the presence and absence of lipopolysaccharide (LPS) which is a known inducer of iNOS and COX-2. Subsequently the expression of iNOS and COX-2 were analyzed by Western and Northern analysis. The production of NO and PGE2 were assayed by Greiss and RIA respectively. RESULTS: Cyclosporin (1-5 microg/ml) and rapamycin (1.0-10 nM) but not FK-506 (5-10 nM) inhibited both iNOS and COX-2 expression at mRNA level which led to significant inhibition of NO and PGE2 production. CONCLUSION: These studies characterize differential mechanistic capacity of the immunophilin-binding immunosuppressive drugs (comparable to hydrocortisone) to inhibit both iNOS and COX-2 expression. Inhibition of iNOS and COX-2 mRNA accumulation by cyclosporin and rapamycin seem to be distinct. These studies also highlight potential anti-inflammatory properties of these drugs in addition to their known immunosuppressive activity.


Anti-Inflammatory Agents/pharmacology , Dinoprostone/biosynthesis , Immunosuppressive Agents/pharmacology , Isoenzymes/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Cell Line , Cyclooxygenase 2 , Cyclosporine/pharmacology , Gene Expression/drug effects , Hydrocortisone/pharmacology , Isoenzymes/genetics , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/metabolism , Sirolimus/pharmacology , Tacrolimus/pharmacology
7.
J Immunol ; 164(5): 2684-91, 2000 Mar 01.
Article En | MEDLINE | ID: mdl-10679109

Osteoarthritis-affected cartilage exhibits enhanced expression of fibronectin (FN) and osteopontin (OPN) mRNA in differential display and bioinformatics screen. Functional genomic analysis shows that the engagement of the integrin receptors alpha 5 beta 1 and alpha v beta 3 of FN and OPN, respectively, have profound effects on chondrocyte functions. Ligation of alpha 5 beta 1 using activating mAb JBS5 (which acts as agonist similar to FN N-terminal fragment) up-regulates the inflammatory mediators such as NO and PGE2 as well as the cytokines, IL-6 and IL-8. Furthermore, up-regulation of these proinflammatory mediators by alpha 5 beta1 integrin ligation is mediated via induction and autocrine production of IL-1 beta, because type II soluble IL-1 decoy receptor inhibits their production. In contrast, alpha v beta 3 complex-specific function-blocking mAb (LM609), which acts as an agonist similar to OPN, attenuates the production of IL-1 beta, NO, and PGE2 (triggered by alpha 5 beta 1, IL-1 beta, IL-18, or IL-1 beta, TNF-alpha, plus LPS) in a dominant negative fashion by osteoarthritis-affected cartilage and activated bovine chondrocytes. These data demonstrate a cross-talk in signaling mechanisms among integrins and show that integrin-mediated "outside in" and "inside out" signaling very likely influences cartilage homeostasis, and its deregulation may play a role in the pathogenesis of osteoarthritis.


Cartilage, Articular/immunology , Inflammation Mediators/metabolism , Osteoarthritis/genetics , Osteoarthritis/immunology , Receptors, Fibronectin/physiology , Receptors, Vitronectin/physiology , Adult , Aged , Animals , Antibodies, Monoclonal/pharmacology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cattle , Chondrocytes/metabolism , Dinoprostone/antagonists & inhibitors , Dinoprostone/biosynthesis , Humans , Interleukin-1/antagonists & inhibitors , Interleukin-1/biosynthesis , Interleukin-1/genetics , Interleukin-18/physiology , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Ligands , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Middle Aged , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Osteoarthritis/metabolism , RNA, Messenger/metabolism , Receptors, Fibronectin/antagonists & inhibitors , Receptors, Fibronectin/immunology , Receptors, Fibronectin/metabolism , Receptors, Vitronectin/immunology , Receptors, Vitronectin/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Up-Regulation/immunology
8.
J Immunol ; 163(6): 3459-67, 1999 Sep 15.
Article En | MEDLINE | ID: mdl-10477618

Tetracyclines (doxycycline and minocycline) inhibit inducible NO synthase expression and augment cyclooxygenase (COX)-2 expression and PGE2 production. In contrast, chemically modified tetracyclines (CMTs), such as CMT-3 and -8 (but not CMT-1, -2, and -5), that lack antimicrobial activity, inhibit both NO and PGE2 production in LPS-stimulated murine macrophages, bovine chondrocytes, and human osteoarthritis-affected cartilage, which spontaneously produces NO and PGE2 in ex vivo conditions. Furthermore, CMT-3 augments COX-2 protein expression but inhibits net PGE2 accumulation. This coincides with the ability of CMT-3 and -8 to inhibit COX-2 enzyme activity in vitro. The action of CMTs is distinct from that observed with tetracyclines because 1) CMT-3-mediated inhibition of PGE2 production coincides with modification of COX-2 protein, which is distinct from the nonglycosylated COX-2 protein generated in the presence of tunicamycin, as observed by Western blot analysis and 2) CMT-3 and -8 have no significant effect on COX-2 mRNA accumulation. In contrast, CMT-3 and -8 do not inhibit COX-1 expression in A549 human epithelial cells at the level of protein and mRNA accumulation or modification of COX-1 protein. CMT-3 and -8 inhibit the sp. act. of COX-2 (but not COX-1) in cell-free extracts. These results demonstrate differential action of CMT-3 (Metastat) on COX-1 and -2 expression, which is distinct from other tetracyclines.


Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/antagonists & inhibitors , Dinoprostone/biosynthesis , Isoenzymes/physiology , Prostaglandin-Endoperoxide Synthases/physiology , Tetracyclines/pharmacology , Animals , Blotting, Western , Cattle , Cell Line , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/chemistry , Dinoprostone/metabolism , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Glycosylation/drug effects , Humans , Isoenzymes/biosynthesis , Isoenzymes/isolation & purification , Isoenzymes/metabolism , Membrane Proteins , Mice , Nitric Oxide/metabolism , Nitric Oxide/physiology , Organ Culture Techniques , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/isolation & purification , Prostaglandin-Endoperoxide Synthases/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Tetracyclines/chemistry , Tumor Cells, Cultured , Tunicamycin/pharmacology
9.
Inflamm Res ; 48(6): 337-43, 1999 Jun.
Article En | MEDLINE | ID: mdl-10442486

OBJECTIVE AND DESIGN: To compare two anti-inflammatory drugs: CSAIDS (SB203580) and hydrocortisone on iNOS and COX-2 expression. MATERIAL OR SUBJECTS: Murine macrophages and bovine chondrocytes stimulated with LPS and human OA-affected cartilage were used in this study. TREATMENT: The macrophages and chondrocytes were preincubated (30 min) with 0.1-1.0 microM CSAIDS or 10 microM of hydrocortisone before stimulating them with 1-100 microg/ml LPS. METHODS: The end products of iNOS and COX-2: nitric oxide (NO) and PGE2 were estimated by Greiss method and RIA, respectively. RESULTS: CSAIDS (1 microM) inhibited the production of NO and PGE2 (p< or =0.01) in bovine chondrocytes, but not in murine macrophages (RAW 264.7) (p< or =0.1). In fact, CSAIDS (in murine macrophages) marginally augmented nitrite accumulation (approximately 20%) at 14-24 h of LPS stimulation. Western blot analysis of COX-2 in bovine chondrocytes show decrease in COX-2 expression by hydrocortisone but not CSAIDS, although hydrocortisone and CSAIDS inhibit PGE2 accumulation. Hydrocortisone inhibited both PGE2 and NO production significantly (p< or =0.01) in murine macrophages. Furthermore, hydrocortisone significantly inhibited (p< or =0.01) PGE2 but marginally (p< or =0.05) NO in bovine chondrocytes. CONCLUSION: These experiments demonstrate differential action of CSAIDS and hydrocortisone on NO and PGE2 production in bovine chondrocytes and RAW 264.7 cells.


Anti-Inflammatory Agents/pharmacology , Chondrocytes/drug effects , Dinoprostone/biosynthesis , Hydrocortisone/pharmacology , Imidazoles/pharmacology , Macrophages/drug effects , Nitric Oxide/metabolism , Pyridines/pharmacology , Animals , Cattle , Cell Line , Chondrocytes/metabolism , Cyclooxygenase 2 , Humans , Isoenzymes/biosynthesis , Isoenzymes/drug effects , Lipopolysaccharides/immunology , Macrophages/metabolism , Membrane Proteins , Mice , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/drug effects
10.
J Immunol ; 162(7): 4191-7, 1999 Apr 01.
Article En | MEDLINE | ID: mdl-10201946

Murine macrophages (RAW 264.7) when stimulated with LPS show 90% distribution of cyclooxygenase-2 (COX-2) in the nuclear fraction and approximately 10% in the cytosolic fraction. Further analysis of this cytosolic fraction at 100,000 x g indicates that the COX-2 is distributed both in the 100,000 x g soluble fraction and membrane fraction. Stimulation of RAW 264.7 cells with LPS in the presence of inducible nitric oxide synthase inhibitor L-NMMA at concentrations that inhibit nitrite accumulation by /=85% with higher concentrations of L-NMMA shows 1) up-regulation of PGE2 production, 2) accumulation of COX-2 protein in the 100,000 x g soluble and membrane fractions of the cytosolic fraction, and 3) with no significant effects on the accumulation of COX-2 mRNA. These experiments suggest that low concentrations of nitric oxide (10-15% of the total) attenuate PGE2 production in response to LPS in RAW 264.7 cells. This inhibition is, in part, due to decreased expression of cytosolic COX-2 protein.


Cytosol/enzymology , Dinoprostone/biosynthesis , Isoenzymes/metabolism , Macrophage Activation , Macrophages/metabolism , Nitric Oxide/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , 3T3 Cells , Animals , Blotting, Western , Cell Line , Cycloheximide/pharmacology , Cyclooxygenase 2 , Enzyme Induction/immunology , Isoenzymes/biosynthesis , Isoenzymes/genetics , Macrophage Activation/drug effects , Macrophages/enzymology , Macrophages/immunology , Mice , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/genetics
11.
J Immunol ; 162(6): 3160-7, 1999 Mar 15.
Article En | MEDLINE | ID: mdl-10092766

Tetracyclines (doxycycline and minocycline) augmented (one- to twofold) the PGE2 production in human osteoarthritis-affected cartilage (in the presence or absence of cytokines and endotoxin) in ex vivo conditions. Similarly, bovine chondrocytes stimulated with LPS showed (one- to fivefold) an increase in PGE2 accumulation in the presence of doxycycline. This effect was observed at drug concentrations that did not affect nitric oxide (NO) production. In murine macrophages (RAW 264.7) stimulated with LPS, tetracyclines inhibited NO release and increased PGE2 production. Tetracycline(s) and L-N-monomethylarginine (L-NMMA) (NO synthase inhibitor) showed an additive effect on inhibition of NO and PGE2 accumulation, thereby uncoupling the effects of tetracyclines on NO and PGE2 production. The enhancement of PGE2 production in RAW 264.7 cells by tetracyclines was accompanied by the accumulation of both cyclooxygenase (COX)-2 mRNA and cytosolic COX-2 protein. In contrast to tetracyclines, L-NMMA at low concentrations (< or = 100 microM) inhibited the spontaneous release of No in osteoarthritis-affected explants and LPS-stimulated macrophages but had no significant effect on the PGE2 production. At higher concentrations, L-NMMA (500 microM) inhibited NO release but augmented PGE2 production. This study indicates a novel mechanism of action of tetracyclines to augment the expression of COX-2 and PGE2 production, an effect that is independent of endogenous concentration of NO.


Dinoprostone/biosynthesis , Isoenzymes/biosynthesis , Nitric Oxide/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Tetracycline/pharmacology , Up-Regulation/drug effects , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/enzymology , Cartilage, Articular/metabolism , Cattle , Cell Line , Cyclooxygenase 2 , Cytokines/pharmacology , Doxycycline/pharmacology , Endotoxins/pharmacology , Humans , Isoenzymes/genetics , Macrophages/drug effects , Macrophages/enzymology , Macrophages/metabolism , Membrane Proteins , Mice , Minocycline/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Organ Culture Techniques , Osteoarthritis/metabolism , Prostaglandin-Endoperoxide Synthases/genetics , RNA, Messenger/biosynthesis , omega-N-Methylarginine/pharmacology
12.
Cell Mol Life Sci ; 56(3-4): 305-12, 1999 Oct 15.
Article En | MEDLINE | ID: mdl-11212358

Recent studies have suggested that aspirin and aspirin-like compounds have a variety of actions in addition to their well-studied ability to inhibit cyclooxygenases. These actions include inhibition of the uncoupling of oxidative phosphorylation, decreases in adenosine triphosphate stores. increases in extracellular adenosine, downregulation of the expression and activity of inducible nitric oxide synthetase, inhibition and/or stimulation of various mitogen-activated protein kinase activities and inhibition of nuclear factor binding kappaB site (NF-kappaB) activation. Moreover, aspirin-like compounds have recently been shown to have previously unappreciated clinical and biological effects, some apparently independent of cyclooxygenase. In this review we discuss the various mechanisms of action of aspirin-like compounds and their relevance to clinical disease and therapy.


Aspirin/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Fibrinolytic Agents/pharmacology , Humans , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II
13.
J Immunol ; 160(9): 4570-9, 1998 May 01.
Article En | MEDLINE | ID: mdl-9574564

A snake venom-like protease isolated by a differential display screen between normal and osteoarthritis (OA)-affected cartilage (designated as cSVP) has a cDNA sequence identical to TNF-alpha convertase enzyme (TACE). TACE shows the presence of an unknown prodomain, a cysteine switch, a catalytic domain, a zinc binding region, a disintegrin region, an EGF-like domain, a transmembrane domain, and a unique cytoplasmic region. A TACE construct harboring the signal + prodomain + catalytic region (TACE-SPCdeltaDETCy), expressed in baculovirus could cleave preferentially (approximately 12-fold) the TNF-specific peptide over the matrix metalloproteases peptide in vitro. This recombinant protein also cleaved the natural substrate GST-ProTNF-alpha to TNF-alpha (17 kDa) in vitro. The mRNA for TACE, which is broadly distributed and differentially expressed in a variety of human tissues, is up-regulated in arthritis-affected cartilage, but not normal cartilage. OA-affected cartilage also expressed TNF-alpha mRNA that was not detected in normal cartilage. The OA-affected cartilage (in explant assays) spontaneously released TNF-alpha and IL-8 in ex vivo conditions. Addition of TNF-alphaR fused to IgG Fc fragment (TNF-alphaR:Fc) in the presence or absence of soluble IL-1R (with which it acted additively) significantly attenuated the spontaneous/autocrine release of articular IL-8 in this assay. These experiments demonstrate a functional paracrine/autocrine role of TNF-alpha in OA-affected cartilage that may depend, in part, on up-regulated levels of chondrocyte-derived TACE.


Arthritis, Rheumatoid/genetics , Cartilage/enzymology , DNA, Complementary/genetics , Gene Expression Regulation, Enzymologic/drug effects , Metalloendopeptidases/genetics , Tumor Necrosis Factor-alpha/pharmacology , ADAM Proteins , ADAM17 Protein , Arthritis, Rheumatoid/enzymology , Cloning, Molecular , DNA, Complementary/isolation & purification , Humans , Metalloendopeptidases/biosynthesis , Molecular Sequence Data , Organ Culture Techniques , Sequence Analysis
14.
Proc Assoc Am Physicians ; 110(1): 65-72, 1998.
Article En | MEDLINE | ID: mdl-9460084

Interleukin-1 beta (IL-1 beta) plays a central role in the pathophysiology of cartilage damage and degradation in arthritis. In noninflammatory arthropathies such as osteoarthritis (OA), the synovial-derived IL-1 beta has been implicated in the disease process. In this study, we report that human OA-affected cartilage demonstrates upregulated IL-1 beta mRNA not seen in normal cartilage. The OA-affected cartilage in ex vivo conditions spontaneously releases detectable amounts of autocrine IL-1 beta, nitric oxide (NO), and prostaglandin E2 (PGE2), known to be involved in cartilage damage and inflammation, that cannot be detected in normal cartilage. The autocrine IL-1 beta released by the OA-affected cartilage (for at least 72 hr in ex vivo conditions) is present in sufficient quantities to modulate NO and PGE2 production because addition of recombinant soluble IL-1 beta receptor (but not soluble tumor necrosis factor-alpha receptor) and cytokine-suppressive antiinflammatory drugs (CSAIDs) significantly attenuates the spontaneous release of NO and PGE2. Furthermore, OA-affected cartilage releases significant amounts of IL-6 and IL-8 in ex vivo conditions. Addition of CSAIDs to OA-affected cartilage differentially regulates IL-6 and IL-8 production by inhibiting the spontaneous release of IL-6 but not IL-8 in ex vivo conditions. These experiments demonstrate that the human OA-affected cartilage itself releases sufficient amounts of functionally active autocrine IL-1 beta that can modulate endogenous NO, PGE2, and IL-6, but not IL-8, all of which are known to be stimulated by IL-1 beta in vitro. These IL-1 beta induced pleotropic inflammatory mediators in OA-affected cartilage may be sufficient to facilitate or augment cartilage degradation and inhibit cartilage repair, and therefore lead the cartilage into an autodestructive pathway in osteoarthritis.


Cartilage, Articular/metabolism , Dinoprostone/biosynthesis , Interleukin-1/metabolism , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Nitric Acid/metabolism , Osteoarthritis/metabolism , Anti-Inflammatory Agents/pharmacology , Cartilage, Articular/drug effects , Cell Line , Humans , Imidazoles/pharmacology , Interleukin-1/biosynthesis , Interleukin-1/genetics , Organ Culture Techniques , Osteoarthritis/pathology , Pyridines/pharmacology
15.
Osteoarthritis Cartilage ; 6(4): 269-77, 1998 Jul.
Article En | MEDLINE | ID: mdl-9876396

OBJECTIVE: To examine the effects of non-steroidal anti-inflammatory drugs (NSAIDS) on nitric oxide (NO) and prostaglandin E2 (PGE2) production in chondrocytes from three different species. METHODS: We have estimated NO production by Griess method, and PGE2 by RIA from the supernatants of articular cartilage obtained from osteoarthritis joints (OA-affected cartilage), rat chondrosarcomas (in ex vivo conditions) and bovine chondrocytes (stimulated with cytokines + endotoxin in vitro conditions) in the presence or absence of aspirin, indomethacin, sodium salicylate, tenidap and glucocorticoids. RESULTS: NO, which was spontaneously released in ex vivo conditions by OA-affected cartilage and rat chondrosarcomas (maintained in vivo), was susceptible to inhibition by pharmacologically relevant concentrations of aspirin, sodium salicylate and tenidap, but not to concentrations of indomethacin or glucocorticoids that significantly inhibited PGE2 production under the same conditions. Similarly, the production of NO by bovine chondrocytes grown in monolayer cultures that had been stimulated with cytokines + endotoxins (in vitro) to release both NO and PGE2 (at 48-72 h post stimulation), were inhibited by aspirin, sodium salicylate and tenidap, but not by indomethacin or glucocorticoids at concentrations sufficient to PGE2 production. Inhibition of NO in the cytokines + endotoxin stimulated bovine chondrocytes (like the human OA-affected cartilage) augmented PGE2 production. CONCLUSION: These experiments demonstrate that NO production by chondrocytes across species show a similar profile of susceptibility to inhibition by selected anti-inflammatory drugs. The insensitivity of NO production to glucocorticoids is an important characteristics of these cells that merits further investigation.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cartilage, Articular/drug effects , Chondrocytes/drug effects , Nitric Oxide/biosynthesis , Osteoarthritis, Knee/metabolism , Aged , Animals , Cartilage, Articular/metabolism , Cattle , Chondrocytes/metabolism , Chondrosarcoma/metabolism , Culture Techniques , Cytokines/pharmacology , Endotoxins/pharmacology , Humans , Indoles/pharmacology , Middle Aged , Nitric Oxide Synthase/metabolism , Osteoarthritis, Knee/pathology , Oxindoles , Rats , Rats, Sprague-Dawley , Salicylates/pharmacology , Species Specificity
16.
Arthritis Rheum ; 40(6): 1050-3, 1997 Jun.
Article En | MEDLINE | ID: mdl-9182915

OBJECTIVE: To examine the effect of human interleukin-17 (IL-17) on nitric oxide (NO) production in human osteoarthritis (OA) cartilage under ex vivo conditions. METHODS: OA cartilage from patients undergoing knee replacement surgery was used in explant assays to assess the effect of IL-17. NO production was measured by estimating the stable NO metabolite, nitrite, in conditioned medium. RESULTS: IL-17 augmented the spontaneous production of nitric oxide. This augmentation was sensitive to cycloheximide and pyrrolidine dithiocarbamate, but not to dexamethasone or soluble IL-1 receptor. CONCLUSION: IL-17 augments nitric oxide production in OA cartilage via nuclear factor kappaB activation, but independently of IL-1beta signaling.


Cartilage, Articular/metabolism , Interleukins/physiology , Nitric Oxide/biosynthesis , Osteoarthritis/metabolism , Aged , Cytokines/physiology , Humans , Interleukin-17 , Knee Joint , Knee Prosthesis , Middle Aged , Up-Regulation
17.
FEBS Lett ; 410(2-3): 259-64, 1997 Jun 30.
Article En | MEDLINE | ID: mdl-9237641

Chemically modified tetracyclines [CMT-3 (IC50 approximately 6-13 microM = approximately 2.5-5 microg/ml) and CMT-8 (IC50 approximately 26 microM = 10 microg/ml), but not CMT-1, -2 or -5], which lack anti-microbial activity, inhibited nitrite production in LPS-stimulated macrophages. Unlike competitive inhibitors of L-arginine which inhibited the specific activity of inducible nitric oxide synthase (iNOS) in cell-free extracts, CMTs exerted no such direct effect on the enzyme. CMTs could, however, be shown to inhibit both iNOS mRNA accumulation and protein expression in LPS-stimulated cells. Tetracyclines (doxycycline and CMT-3) unlike hydrocortisone had no significant effect on murine macrophages transfected with iNOS promoter (tagged to a luciferase reporter gene) in the presence of LPS. However, doxycycline and CMT-3 augmented iNOS mRNA degradation, in LPS-stimulated murine macrophages. These studies show a novel mechanism of action of tetracyclines which harbours properties to increase iNOS mRNA degradation and decrease iNOS protein expression and nitric oxide production in macrophages. This property of tetracyclines may have beneficial effects in the treatment of various diseases where excess nitric oxide has been implicated in the pathophysiology of these diseases.


Macrophages/drug effects , Nitric Oxide Synthase/genetics , RNA Processing, Post-Transcriptional/drug effects , RNA, Messenger/metabolism , Tetracyclines/pharmacology , Animals , Arginine/metabolism , Citrulline/metabolism , Doxycycline/chemistry , Doxycycline/pharmacology , Hydrocortisone/pharmacology , Macrophages/metabolism , Mice , Minocycline/chemistry , Minocycline/pharmacology , Molecular Structure , Nitrites/metabolism , RNA, Messenger/drug effects , Tetracyclines/chemistry
18.
Proc Natl Acad Sci U S A ; 93(24): 14014-9, 1996 Nov 26.
Article En | MEDLINE | ID: mdl-8943052

Tetracyclines have recently been shown to have "chondroprotective" effects in inflammatory arthritides in animal models. Since nitric oxide (NO) is spontaneously released from human cartilage affected by osteoarthritis (OA) or rheumatoid arthritis in quantities sufficient to cause cartilage damage, we evaluated the effect of tetracyclines on the expression and function of human OA-affected nitric oxide synthase (OA-NOS) and rodent inducible NOS (iNOS). Among the tetracycline group of compounds, doxycycline > minocycline blocked and reversed both spontaneous and interleukin 1 beta-induced OA-NOS activity in ex vivo conditions. Similarly, minocycline > or = doxycycline inhibited both lipopolysaccharide- and interferon-gamma-stimulated iNOS in RAW 264.7 cells in vitro, as assessed by nitrite accumulation. Although both these enzyme isoforms could be inhibited by doxycycline and minocycline, their susceptibility to each of these drugs was distinct. Unlike acetylating agents or competitive inhibitors of L-arginine that directly inhibit the specific activity of NOS, doxycycline or minocycline has no significant effect on the specific activity of iNOS in cell-free extracts. The mechanism of action of these drugs on murine iNOS expression was found to be, at least in part, at the level of RNA expression and translation of the enzyme, which would account for the decreased iNOS protein and activity of the enzyme. Tetracyclines had no significant effect on the levels of mRNA for beta-actin and glyceraldehyde-3-phosphate dehydrogenase nor on levels of protein of beta-actin and cyclooxygenase 2 expression. These studies indicate that a novel mechanism of action of tetracyclines is to inhibit the expression of NOS. Since the overproduction of NO has been implicated in the pathogenesis of arthritis, as well as other inflammatory diseases, these observations suggest that tetracyclines should be evaluated as potential therapeutic modulators of NO for various pathological conditions.


Cartilage/enzymology , Doxycycline/pharmacology , Minocycline/pharmacology , Nitric Oxide Synthase/biosynthesis , Osteoarthritis/enzymology , Tetracyclines/pharmacology , Animals , Arthritis, Rheumatoid/enzymology , Cell Line , DNA Primers , Enzyme Induction , Humans , Interferon-gamma/pharmacology , Kinetics , Lipopolysaccharides/pharmacology , Macrophages , Mice , Organ Culture Techniques , Polymerase Chain Reaction
...