Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
J Histochem Cytochem ; 69(8): 511-522, 2021 08.
Article En | MEDLINE | ID: mdl-34291686

Induction of severe inflammatory arthritis in the collagen antibody-induced arthritis (CAIA) murine model causes extensive joint damage and pain-like behavior compromising analysis. While mild models are less severe, their reduced, variable penetrance makes assessment of treatment efficacy difficult. This study aimed to compare macroscopic and microscopic changes in the paws, along with central nervous system activation between a mild and moderate CAIA model. Balb/c mice (n=18) were allocated to control, mild, and moderate CAIA groups. Paw inflammation, bone volume (BV), and paw volume (PV) were assessed. Histologically, the front paws were assessed for joint inflammation, cartilage damage, and pre/osteoclast-like cells and the lumbar spinal cord and the periaqueductal gray (PAG) region of the brain for glial reactivity. A moderate CAIA dose induced (1) significantly greater local paw inflammation, inflammatory cell infiltration, and PV; (2) significantly more osteoclast-like cells on the bone surface and within the surrounding soft tissue; and (3) significantly greater glial reactivity within the PAG compared with the mild CAIA model. These findings support the use of a moderate CAIA model (higher dose of monoclonal antibodies with low-dose lipopolysaccharide) to induce more consistent histopathological features, without excessive joint destruction.


Arthritis, Experimental/pathology , Bone Resorption/pathology , Cartilage, Articular/pathology , Disease Models, Animal , Edema/pathology , Animals , Antibodies, Monoclonal/administration & dosage , Arthritis, Experimental/chemically induced , Arthritis, Experimental/diagnosis , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/pathology , Bone Resorption/chemically induced , Bone Resorption/diagnosis , Cartilage, Articular/drug effects , Edema/chemically induced , Edema/diagnosis , Female , Forelimb/drug effects , Forelimb/pathology , Histocytochemistry , Lipopolysaccharides/administration & dosage , Mice , Mice, Inbred BALB C , Neuroglia/drug effects , Neuroglia/pathology , Osteoclasts/drug effects , Osteoclasts/pathology , Periaqueductal Gray/drug effects , Periaqueductal Gray/pathology , Severity of Illness Index , Spinal Cord/drug effects , Spinal Cord/pathology
2.
Acta Biomater ; 87: 296-306, 2019 03 15.
Article En | MEDLINE | ID: mdl-30690207

Periprosthetic osteolysis is a major cause of implant failure in total hip replacements. Aseptic loosening caused by osteolytic lesions is associated with the production of bioactive wear particles from the articulations of implants. Wear particles infiltrate the surrounding tissue of implants, promoting inflammation as well as bone resorption. Osteocytes have been shown to both regulate physiological osteoclastogenesis and directly remodel their perilacunar bone matrix by the process of osteocytic osteolysis. We hypothesise that osteocytes respond to wear debris of orthopaedic implant materials by adopting a pro-catabolic phenotype and thus contribute to periprosthetic osteolysis through the known pathways of bone loss. Osteocyte responses to particles derived from clinically relevant materials, ultra-high molecular weight polyethylene (UHMWPE), highly cross-linked polyethylene (XLPE) and metal alloys, Ti6Al4V and CoCrMo, were examined in vitro in human primary osteocyte-like cultures. Osteocyte-like cells exposed to both polyethylene and metal wear particle types showed upregulated expression of catabolic markers associated with osteocytic osteolysis, MMP13, carbonic anhydrase 2 (CA2) and cathepsin K (CTSK). In addition, pro-osteoclastogenesis markers RANKL and M-CSF were induced, as well as the expression of pro-inflammatory cytokines, IL-6 and TNFα, albeit with different kinetics. These findings suggest a previously unrecognised action of wear particles of multiple orthopaedic materials on osteocytes, and suggest a multifaceted role for osteocytes in periprosthetic osteolysis. STATEMENT OF SIGNIFICANCE: This study addresses periprosthetic osteolysis, a major clinical problem leading to aseptic loosening of orthopaedic implants. It is well accepted that wear particles of polyethylene and of other implant materials stimulate the activity of bone resorbing osteoclasts. Our recent work provided evidence that commercial particles of ultra-high molecular weight polyethylene (UHMWPE) stimulated osteocytes to adopt a bone catabolic state. In this study we demonstrate for the first time that particles derived from materials in clinical use, conventional UHMWPE, highly cross-linked polyethylene (XLPE), and Ti6Al4V and CoCrMo metal alloys, all stimulate human osteocyte activities of osteocyte-regulated osteoclastogenesis, osteocytic osteolysis, proinflammatory responses, osteocyte apoptosis, albeit to varying extents. This study provides further mechanistic insight into orthopaedic wear particle mediated bone disease in terms of the osteocyte, the most abundant and key controlling cell type in bone.


Antigens, Differentiation/biosynthesis , Bone-Implant Interface , Osteocytes/metabolism , Osteolysis/metabolism , Polyethylenes/adverse effects , Titanium/adverse effects , Up-Regulation/drug effects , Alloys , Humans , Osteocytes/pathology , Osteolysis/chemically induced , Osteolysis/pathology , Polyethylenes/chemistry , Polyethylenes/pharmacology , Titanium/chemistry , Titanium/pharmacology
3.
Inflammation ; 40(5): 1566-1575, 2017 Oct.
Article En | MEDLINE | ID: mdl-28555364

Osteoclast-associated receptor (OSCAR) is a co-stimulatory receptor in osteoclastogenesis. Synovial tissues from active rheumatoid arthritis (RA) patients express higher levels of OSCAR compared with osteoarthritic and normal patients; however, the comparison of OSCAR levels in different regions of active RA synovium has not been reported. The regulation of OSCAR by TNF-α and receptor activator of NF kappa ß ligand (RANKL) in pre-osteoclasts/osteoclasts in vitro is unclear. OSCAR and tartrate-resistant acid phosphatase (TRAP) expression levels did not differ between the cartilage pannus junction (CPJ) and non-CPJ regions in active RA. We demonstrate a similar pattern of OSCAR expression in the CPJ and non-CPJ synovial tissue from patients with active RA. OSCAR was associated with mononuclear cells in both the lining and sub-lining and endothelial cells (von Willebrand factor positive). Pre-osteoclasts (TRAP-positive cells) were present in the lining and sub-lining of both regions. OSCAR messenger RNA (mRNA) expression and release by pre-oscteoclasts/osteoclasts was modulated by RANKL with/without TNF-α in vitro. Osteoclast resorption on dentine slices was significantly greater with TNF-α pre-treatment and RANKL (10 ng/ml) than RANKL 10 or 50 ng/ml alone or RANKL 10 ng/ml with TNF-α given from day 3 post-RANKL. The lower levels of OSCAR mRNA expression corresponded with high osteoclast activity levels.


Arthritis, Rheumatoid/metabolism , Osteoclasts/metabolism , Receptors, Cell Surface/metabolism , Synovial Membrane/chemistry , Endothelial Cells/chemistry , Humans , Leukocytes, Mononuclear/chemistry , RANK Ligand/physiology , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Synovial Membrane/metabolism , Tartrate-Resistant Acid Phosphatase/metabolism , Tumor Necrosis Factor-alpha/physiology
4.
Mediators Inflamm ; 2015: 564042, 2015.
Article En | MEDLINE | ID: mdl-26347311

OBJECTIVE: To investigate the effect of Embelin, an inhibitor of X-Linked Inhibitor of Apoptosis Protein (XIAP), on inflammation and bone erosion in a collagen antibody induced arthritis (CAIA) in mice. METHODS: Four groups of mice (n = 6 per group) were allocated: CAIA untreated mice, CAIA treated with Prednisolone (10 mg/kg/day), CAIA treated with low dose Embelin (30 mg/kg/day), and CAIA treated with high dose Embelin (50 mg/kg/day). Joint inflammation was evaluated using clinical paw score and histological assessments. Bone erosion was assessed using micro-CT, tartrate resistant acid phosphatase (TRAP) staining, and serum carboxy-terminal collagen crosslinks (CTX-1) ELISA. Immunohistochemistry was used to detect XIAP protein. TUNEL was performed to identify apoptotic cells. RESULTS: Low dose, but not high dose Embelin, suppressed inflammation as reflected by lower paw scores (P < 0.05) and lower histological scores for inflammation. Low dose Embelin reduced serum CTX-1 (P < 0.05) and demonstrated lower histological score and TRAP counting, and slightly higher bone volume as compared to CAIA untreated mice. XIAP expression was not reduced but TUNEL positive cells were more abundant in Embelin treated CAIA mice. CONCLUSION: Low dose Embelin suppressed inflammation and serum CTX-1 in CAIA mice, indicating a potential use for Embelin to treat pathological bone loss.


Arthritis, Experimental/drug therapy , Benzoquinones/therapeutic use , Bone Resorption/drug therapy , Inflammation/drug therapy , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Acid Phosphatase/metabolism , Animals , Arthritis, Experimental/immunology , Bone Resorption/immunology , Isoenzymes/metabolism , Mice , Tartrate-Resistant Acid Phosphatase
5.
J Immunol Res ; 2015: 281287, 2015.
Article En | MEDLINE | ID: mdl-26064999

The field of osteoimmunology has emerged in response to the range of evidences demonstrating the close interrelationship between the immune system and bone metabolism. This is pertinent to immune-mediated diseases, such as rheumatoid arthritis and periodontal disease, where there are chronic inflammation and local bone erosion. Periprosthetic osteolysis is another example of chronic inflammation with associated osteolysis. This may also involve immune mediation when occurring in a patient with rheumatoid arthritis (RA). Similarities in the regulation and mechanisms of bone loss are likely to be related to the inflammatory cytokines expressed in these diseases. This review highlights the role of immune-related factors influencing bone loss particularly in diseases of chronic inflammation where there is associated localized bone loss. The importance of the balance of the RANKL-RANK-OPG axis is discussed as well as the more recently appreciated role that receptors and adaptor proteins involved in the immunoreceptor tyrosine-based activation motif (ITAM) signaling pathway play. Although animal models are briefly discussed, the focus of this review is on the expression of ITAM associated molecules in relation to inflammation induced localized bone loss in RA, chronic periodontitis, and periprosthetic osteolysis, with an emphasis on the soluble and membrane bound factor osteoclast-associated receptor (OSCAR).


Bone Diseases, Metabolic/metabolism , Bone and Bones/metabolism , Inflammation/metabolism , Osteolysis/metabolism , Signal Transduction/physiology , Animals , Arthritis, Rheumatoid/metabolism , Humans , Periodontitis/metabolism
6.
J Biomed Mater Res A ; 103(11): 3572-9, 2015 Nov.
Article En | MEDLINE | ID: mdl-25903444

The study aimed to determine the effects of parthenolide (PAR) on bone volume (BV) and bone surface resorption as assessed by live-animal microcomputed tomography (µCT) and possible osteocyte death as indicated by empty lacunae histologically in polyethylene (PE) particle-induced calvarial osteolysis in mice. Baseline µCT scans were conducted 7 days preimplantation of 2 × 10(8) PE particles/mL over the calvariae (day 0). PAR at 1 mg/kg/day was subcutaneously injected on days 0, 4, 7, and 10. At day 14, BV and surface resorption was analyzed with µCT. Calvarial tissue was processed for histomorphometric osteocyte evaluation. Serum was analyzed for type-1 carboxy-terminal collagen crosslinks (CTX-1) and osteoclast associated receptor (OSCAR) levels by ELISA. PE significantly decreased BV (p = 0.0368), increased surface bone resorption area (p = 0.0022), and increased the percentage of empty lacunae (p = 0.0043). Interestingly, PAR significantly reduced the resorption surface area (p = 0.0022) and the percentage of empty osteocyte lacunae (p = 0.0087) in the PE-calvariae, but it did not affect BV, serum CTX-1 or OSCAR levels. The ability of PAR to inhibit PE-induced surface bone erosion may better reflect the in vivo situation, where bone resorption occurs on the surface at the bone-implant interface and may also be related to the role of osteocytes in this pathology.


Bone Resorption/chemically induced , Bone Resorption/pathology , Osteoclasts/pathology , Osteolysis/chemically induced , Polyethylene/adverse effects , Prostheses and Implants/adverse effects , Sesquiterpenes/pharmacology , Skull/pathology , Animals , Bone Resorption/blood , Bone Resorption/diagnostic imaging , Collagen Type I/blood , Humans , Mice , Models, Animal , Organ Size/drug effects , Osteoarthritis/blood , Osteoarthritis/pathology , Osteoclasts/drug effects , Osteolysis/diagnostic imaging , Osteolysis/pathology , Peptides/blood , Receptors, Cell Surface/blood , Skull/diagnostic imaging , Solubility , X-Ray Microtomography
7.
Sci Rep ; 4: 7595, 2014 Dec 23.
Article En | MEDLINE | ID: mdl-25534583

As the only cells capable of efficiently resorbing bone, osteoclasts are central mediators of both normal bone remodeling and pathologies associates with excessive bone resorption. However, despite the clear evidence of interplay between osteoclasts and the bone surface in vivo, the role of the bone substrate in regulating osteoclast differentiation and activation at a molecular level has not been fully defined. Here, we present the first comprehensive expression profiles of osteoclasts differentiated on authentic resorbable bone substrates. This analysis has identified numerous critical pathways coordinately regulated by osteoclastogenic cytokines and bone substrate, including the transition from proliferation to differentiation, and sphingosine-1-phosphate signaling. Whilst, as expected, much of this program is dependent upon integrin beta 3, the pre-eminent mediator of osteoclast-bone interaction, a surprisingly significant portion of the bone substrate regulated expression signature is independent of this receptor. Together, these findings identify an important hitherto underappreciated role for bone substrate in osteoclastogenesis.


Bone Resorption/metabolism , Cell Differentiation/physiology , Cell Proliferation/physiology , Gene Expression Regulation/physiology , Osteoclasts/metabolism , Animals , Gene Expression Profiling , Mice , Osteoclasts/cytology
8.
Biochim Biophys Acta ; 1833(12): 3306-3313, 2013 Dec.
Article En | MEDLINE | ID: mdl-24100160

The purpose of this study was to determine how dexamethasone (DEX) regulates the expression and activity of αvß3 integrin. FACS analysis showed that DEX treatment induced expression of an activated αvß3 integrin. Its expression remained high as long as DEX was present and continued following DEX removal. FACS analysis showed that the upregulation of αvß3 integrin was the result of an increase in the expression of the ß3 integrin subunit. By real time qPCR, DEX treatment induced a 6.2-fold increase (p<0.04) in ß3 integrin mRNA by day 2 compared to control and remained elevated for 6days of treatment and then an additional 10days once the DEX was removed. The increase in ß3 integrin mRNA levels required only 1day of DEX treatment to increase levels for 4days in the absence of DEX. In contrast, DEX did not alter ß1 integrin mRNA or protein levels. The DEX-induced upregulation of ß3 integrin mRNA was partly due to an increase in its half-life to 60.7h from 22.5h in control cultures (p<0.05) and could be inhibited by RU486 and cycloheximide, suggesting that DEX-induced de novo protein synthesis of an activation factor was needed. The calcineurin inhibitors cyclosporin A (CsA) and FK506 inhibited the DEX induced increase in ß3 integrin mRNA. In summary, the DEX-induced increase in ß3 integrin is a secondary glucocorticoid response that results in prolonged expression of αvß3 integrin and the upregulation of the ß3 integrin subunit through the calcineurin/NFAT pathway.


Calcineurin/metabolism , Dexamethasone/pharmacology , Integrin alphaVbeta3/metabolism , NFATC Transcription Factors/metabolism , Signal Transduction/drug effects , Cell Line , Cycloheximide/pharmacology , Cyclosporine/pharmacology , Ethanol/pharmacology , Half-Life , Humans , Integrin alphaVbeta3/genetics , Mifepristone/pharmacology , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Glucocorticoid/metabolism , Tacrolimus/pharmacology , Trabecular Meshwork/cytology
9.
Ann Rheum Dis ; 72 Suppl 2: ii52-5, 2013 Apr.
Article En | MEDLINE | ID: mdl-23253928

The inflammatory arthropathies that include rheumatoid arthritis, the seronegative spondyloarthropathies and systemic lupus erythematosus are characterised by marked alterations in the architecture and structural integrity of peri-articular bone; however, the pattern and natural history of the skeletal changes differs in these conditions. In part, this can be attributed to differences in the primary anatomical site of the inflammation, but also there is evidence that there are differences in the biological properties and products produced by inflammatory tissues. This review will focus on recent advances in the understanding of the cellular and molecular mechanisms that contribute to the differential pattern of articular bone remodelling in these prototypical inflammatory forms of arthritis.


Arthritis, Rheumatoid/physiopathology , Bone Remodeling/physiology , Lupus Erythematosus, Systemic/physiopathology , Spondylarthropathies/physiopathology , Humans , Osteoclasts/physiology
10.
Arthritis Res Ther ; 14(6): R245, 2012 Nov 12.
Article En | MEDLINE | ID: mdl-23146195

INTRODUCTION: The immunoreceptor tyrosine-based activation motif (ITAM) pathway provides osteoclast co-stimulatory signals and regulates proliferation, survival and differentiation of effector immune cells. In the osteoclast, the receptors Triggering Receptor Expressed on Myeloid cells 2 (TREM2) and Osteoclast Associated Receptor (OSCAR) and their respective adaptor proteins, DAP12 and FcRγ mediate ITAM signals and induce calcium signaling and the crucial transcription factor, NFATc1. In rheumatoid arthritis (RA), OSCAR expression by monocytes is inversely correlated with disease activity. Additionally, serum levels of OSCAR are reduced in RA patients versus healthy controls suggesting that expression and secretion or cleavage of soluble (s) OSCAR is immune modulated. Recent data suggest that endothelial cells may also be a source of OSCAR. METHODS: ITAM receptors, their adaptor proteins, and NFATc1 and cathepsin K were detected in human synovial tissues by immunohistochemistry. Synovial tissues from patients with active RA were compared with tissue from patients in remission, osteoarthritis (OA) patients and healthy individuals. OSCAR was measured by immunoassay in synovial fluids recovered from active RA and OA patients. Endothelial cells were cultured with or without 5 ng/mL TNF-α or IL-1ß over 72 hours. Temporal expression of OSCAR mRNA was assessed by qRT PCR and OSCAR protein in the supernatant was measured by ELISA. RESULTS: Significantly higher (P < 0.05) NFATc1-positive inflammatory cell aggregates were found in active RA tissues than in healthy synovial tissue. Similarly, the percentage of OSCAR, FcRγ, DAP12 and TREM2 positive cells was significantly higher in active RA tissues compared to the healthy synovial tissue. Notably, OSCAR was strongly expressed in the microvasculature of the active RA tissues (9/9), inactive RA (8/9) weakly in OA (4/9) but only in the lumen of healthy synovial tissue (0/8). OSCAR levels were detected in synovial fluids from both RA (47 to 152 ng/mL) and OA (112 to 145 ng/mL) patients. Moreover, OSCAR mRNA expression and soluble OSCAR release was stimulated by TNF-α and IL1-ß in cultured endothelial cells. CONCLUSIONS: Increased levels of ITAM related factors were present in synovial tissue from active RA joints compared to OA and healthy joints. OSCAR was strongly expressed by the vasculature of active RA patients and membrane bound and soluble OSCAR was stimulated by inflammatory mediators in endothelial cells in vitro.


Arthritis, Rheumatoid/metabolism , Joints/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Synovial Membrane/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Aged, 80 and over , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Gene Expression/drug effects , Humans , Immunohistochemistry , Interleukin-1beta/pharmacology , Joints/blood supply , Male , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microscopy, Confocal , Middle Aged , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Receptors, Cell Surface/genetics , Receptors, Fc/genetics , Receptors, Fc/metabolism , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Synovial Membrane/blood supply , Tumor Necrosis Factor-alpha/pharmacology
11.
Arthritis Res Ther ; 13(2): R51, 2011 Mar 24.
Article En | MEDLINE | ID: mdl-21435232

INTRODUCTION: TNF-like weak inducer of apoptosis (TWEAK) has been proposed as a mediator of inflammation and bone erosion in rheumatoid arthritis (RA). This study aimed to investigate TWEAK and TWEAK receptor (Fn14) expression in synovial tissue from patients with active and inactive rheumatoid arthritis (RA), osteoarthritis (OA) and normal controls and assess soluble (s)TWEAK levels in the synovial fluids from patients with active RA and OA. Effects of sTWEAK on osteoclasts and osteoblasts were investigated in vitro. METHODS: TWEAK and Fn14 expression were detected in synovial tissues by immunohistochemistry (IHC). Selected tissues were dual labelled with antibodies specific for TWEAK and lineage-selective cell surface markers CD68, Tryptase G, CD22 and CD38. TWEAK mRNA expression was examined in human peripheral blood mononuclear cells (PBMC) sorted on the basis of their expression of CD22. sTWEAK was detected in synovial fluid from OA and RA patients by ELISA. The effect of sTWEAK on PBMC and RAW 264.7 osteoclastogenesis was examined. The effect of sTWEAK on cell surface receptor activator of NF Kappa B Ligand (RANKL) expression by human osteoblasts was determined by flow cytometry. RESULTS: TWEAK and Fn14 expression were significantly higher in synovial tissue from all patient groups compared to the synovial tissue from control subjects (P < 0.05). TWEAK was significantly higher in active compared with inactive RA tissues (P < 0.05). TWEAK expression co-localised with a subset of CD38+ plasma cells and with CD22+ B-lymphocytes in RA tissues. Abundant TWEAK mRNA expression was detected in normal human CD22+ B cells. Higher levels of sTWEAK were observed in synovial fluids isolated from active RA compared with OA patients. sTWEAK did not stimulate osteoclast formation directly from PBMC, however, sTWEAK induced the surface expression of RANKL by human immature, STRO-1+ osteoblasts. CONCLUSIONS: The expression of TWEAK by CD22+ B cells and CD38+ plasma cells in RA synovium represents a novel potential pathogenic pathway. High levels of sTWEAK in active RA synovial fluid and of TWEAK and Fn14 in active RA tissue, together with the effect of TWEAK to induce osteoblastic RANKL expression, is consistent with TWEAK/Fn14 signalling being important in the pathogenesis of inflammation and bone erosion in RA.


Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Receptors, Tumor Necrosis Factor/biosynthesis , Tumor Necrosis Factors/biosynthesis , Aged , B-Lymphocytes/metabolism , Cell Separation , Cytokine TWEAK , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Male , Middle Aged , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Osteoblasts/metabolism , Plasma Cells/metabolism , Receptors, Tumor Necrosis Factor/analysis , Synovial Fluid/chemistry , Synovial Fluid/metabolism , TWEAK Receptor , Tumor Necrosis Factors/analysis
12.
J Cell Physiol ; 226(12): 3413-21, 2011 Dec.
Article En | MEDLINE | ID: mdl-21344395

While attachment to bone is required for optimal osteoclast function, the molecular events that underlie this fact are unclear, other than that the cell requires adhesion to mineralized matrix to assume a fully differentiated phenotype. To address this issue, we cultured murine bone marrow-derived osteoclasts on either cell culture plastic or devitalized mouse calvariae to identify the distinct genetic profile induced by interaction with bone. Among a number of genes previously unknown to be expressed in osteoclasts we found that Annexin A8 (AnxA8) mRNA was markedly up-regulated by bone. AnxA8 protein was present at high levels in osteoclasts present in human tissues recovered from sites of pathological bone loss. The presence of bone mineral was required for up-regulation of AnxA8 mRNA since osteoclasts plated on decalcified bone express AnxA8 at low levels as did osteoclasts plated on native or denatured type I collagen. Finally, AnxA8-regulated cytoskeletal reorganization in osteoclasts generated on a mineralized matrix. Thus, we used a novel approach to define a distinct bone-dependent genetic program associated with terminal osteoclast differentiation and identified Anxa8 as a gene strongly induced late in osteoclast differentiation and a protein that regulates formation of the cell's characteristic actin ring.


Annexins/metabolism , Bone Matrix/metabolism , Cell Differentiation , Osteoclasts/metabolism , Actins/metabolism , Animals , Annexins/genetics , Cell Shape , Cells, Cultured , Cytoskeleton/metabolism , Gene Expression Profiling/methods , Humans , Immunohistochemistry , Mice , Oligonucleotide Array Sequence Analysis , RNA Interference , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic , Up-Regulation
13.
J Biol Chem ; 283(35): 23514-23, 2008 Aug 29.
Article En | MEDLINE | ID: mdl-18593711

Snail, a transcriptional repressor of E-cadherin expression, plays a role in the process of epithelial-mesenchymal transition. However, the molecular basis of the role of snail in epithelial-mesenchymal transition has not been fully clarified. Here we show that the expression of snail in epithelial Madin-Darby canine kidney (MDCK) and A431 cells enhances both cell detachment and attachment. Snail did not confer resistance to anoikis induced by loss of contact but instead enhanced cell attachment to extracellular matrices such as fibronectin. This attachment was inhibited by Arg-Gly-Asp (RGD) peptides. Up-regulation of the promoter activity of integrin alphaV was observed in snail-expressing MDCK (MDCK/snail) cells. Snail also enhanced MDCK cell migration toward osteopontin that is a ligand for integrin alphaVbeta3. We confirmed the reduction of basement membrane proteins such as laminin (LN) alpha3, beta3, and gamma2 (laminin-5/LN-5) and of receptors for LN-5 such as integrins alpha3, alpha6, or beta4 in MDCK/snail or in snail-expressing A431 (A431/snail) cells. Nevertheless, suppression of LN-alpha3 chain by transient transfection of small interference RNAs resulted in no enhancement of cell detachment. We also found an induction of matrix metalloproteinase-3 in MDCK/snail and A431/snail cells. However, the inhibition of matrix metalloproteinase-3 showed no significant effect on the detachment of MDCK/snail cells. These results suggest that snail enhances cell detachment by multiple mechanism and leads to cell migration and reattachment at a second site, at least in part, by changing the expression of integrins in the cells.


Basement Membrane/metabolism , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix/metabolism , Integrins/biosynthesis , Transcription Factors/metabolism , Up-Regulation , Animals , Anoikis/drug effects , Anoikis/genetics , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , Dogs , Extracellular Matrix/genetics , Extracellular Matrix Proteins/genetics , Humans , Matrix Metalloproteinase 3/biosynthesis , Matrix Metalloproteinase 3/genetics , Oligopeptides/pharmacology , RNA, Small Interfering/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Up-Regulation/drug effects
14.
J Cell Physiol ; 215(3): 636-44, 2008 Jun.
Article En | MEDLINE | ID: mdl-18288635

Expression of the alpha(v)beta(3) integrin is required for normal osteoclast function. We previously showed that an evolutionary conserved NFATc1 binding site is required for RANKL induction and NFATc1 transactivation of the human beta(3) promoter. The mechanism conferring specificity for RANKL induction and NFATc1 transduction of the beta(3) gene in osteoclast differentiation is unclear since NFATc1 is expressed and activated in numerous cell types that do not express the beta(3) gene. PU.1 is an ETS family transcription factor in myeloid cells associated with expression of various osteoclast genes. The present study investigates the role of NFATc1 in concert with PU.1 in osteoclast-specific transcription of the mouse beta(3) integrin gene. The mouse beta(3) promoter was transactivated by NFATc1 in RAW264.7 cells and deletion or mutation of either of the conserved NFAT and PU.1 binding sites abrogated transactivation. NFATc1 transactivation of the mouse beta(3) promoter was specifically dependent on co-transfected PU.1 in HEK293 cells, to the exclusion of other ETS family members. Direct binding of NFATc1 and PU.1 to their cognate sequences was demonstrated by EMSA and NFATc1 and PU.1 occupy their cognate sites in RANKL-treated mouse marrow precursors in chromatin immuno-precipitation (ChIP) assays. TAT-mediated transduction with dominant-negative NFATc1 dose-dependently blocked endogenous expression of the mouse beta(3) integrin and the formation of TRAP positive multinucleated cells in RANKL-treated mouse macrophages. These data provide evidence that NFATc1, in concert with PU.1, are involved in regulation of beta(3) integrin expression during osteoclast differentiation and suggest that PU.1 confers specificity to the NFATc1 response to macrophage lineage cells.


Gene Expression Regulation , Integrin beta3/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Base Pairing , Base Sequence , Binding Sites , Cell Differentiation , Cell Line , Genes, Dominant , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NFATC Transcription Factors/genetics , Osteoclasts/cytology , Protein Binding , Sequence Deletion , Sequence Homology, Nucleic Acid , Transcriptional Activation/genetics , Transfection
15.
Biochim Biophys Acta ; 1769(11-12): 659-67, 2007.
Article En | MEDLINE | ID: mdl-17949830

The calcitonin receptor (CTR) is expressed in a wide variety of tissues and cell types. In bone, its expression is restricted to osteoclasts, the cells that mediate bone resorption. The human CTR (hCTR) gene has a complex structural organization that exhibits similarity to the porcine (pCTR) and mouse (mCTR) CTR genes. In these species, alternative splicing of a single gene generates multiple CTR isoforms that are distributed in both tissue-specific and species-specific patterns. However, the structural organization of the 5' putative regulatory region and transcriptional mechanisms responsible for tissue-specific expression of the different CTR isoforms are not fully defined. The present studies were undertaken to characterize the structural organization of the 5'-region of the hCTR and identify the regulatory regions involved in osteoclast-specific transcriptional activation. Analysis of mRNA prepared from human osteoclasts using reverse transcription-polymerase chain reaction (RT-PCR) and transient transfection of hCTR promoter-luciferase reporter constructs identified two regions in the 5'-flanking sequence of the hCTR gene that regulated CTR gene expression in osteoclasts. Both of these putative promoters were responsive to the osteoclast-inducing cytokine, receptor activator of NF-kappaB ligand (RANKL) and demonstrated trans-activation by the RANKL-induced transcription factor nuclear factor of activated T cells (NFATc1), consistent with a role in regulating CTR gene expression in osteoclasts.


Gene Expression Regulation , Osteoclasts/metabolism , Promoter Regions, Genetic , Receptors, Calcitonin/genetics , Animals , Cells, Cultured , Humans , Mice , RANK Ligand/metabolism , RNA, Messenger , Tissue Distribution , Transfection
16.
In Vitro Cell Dev Biol Anim ; 42(7): 182-8, 2006.
Article En | MEDLINE | ID: mdl-16948499

Osteoclasts are bone-resorbing cells that differentiate from macrophage precursors in response to receptor activator of NF-kappaB ligand (RANKL). In vitro models of osteoclast differentiation are principally based on primary cell cultures, which are poorly suited to molecular and transgene studies because of the limitations associated with the use of primary macrophage. RAW264.7 is a transfectable macrophage cell line with the capacity to form osteoclast-like cells. In the present study, we have identified osteoclast precursors among clones of RAW264.7 cells. RAW264.7 cell were cloned by limiting dilution and induced to osteoclast differentiation by treatment with recombinant RANKL. Individual RAW264.7 cell clones formed tartrate resistant acid phosphatase (TRAP)-positive multinuclear cells to various degrees with RANKL treatment. All clones tested expressed the RANKL receptor RANK. Each of the clones expressed the osteoclast marker genes TRAP and cathepsin-K mRNA with RANKL treatment. However, we noted that only select clones were able to form large, well-spread, TRAP-positive multinuclear cells. Clones capable of forming large TRAP-positive multinuclear cells also expressed beta3 integrin and calcitonin receptor mRNAs and were capable of resorbing a mineralized matrix. All clones tested activated NF-kappaB with RANKL treatment. cDNA expression profiling of osteoclast precursor RAW264.7 cell clones demonstrates appropriate expression of a large number of genes before and after osteoclastic differentiation. These osteoclast precursor RAW264.7 cell clones provide a valuable model for dissecting the cellular and molecular regulation of osteoclast differentiation and activation.


Osteoclasts/cytology , Stem Cells/cytology , Acid Phosphatase/metabolism , Animals , Bone Resorption/genetics , Bone Resorption/metabolism , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Line , Clone Cells , DNA, Complementary/metabolism , Durapatite/metabolism , Gene Expression Profiling , Genetic Markers , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Isoenzymes/metabolism , Membrane Glycoproteins/metabolism , Mice , RANK Ligand , RNA, Messenger/metabolism , Receptor Activator of Nuclear Factor-kappa B , Receptors, Calcitonin/genetics , Receptors, Calcitonin/metabolism , Recombinant Fusion Proteins/metabolism , Stem Cells/metabolism , Tartrate-Resistant Acid Phosphatase
17.
Arthritis Res Ther ; 8(3): R70, 2006.
Article En | MEDLINE | ID: mdl-16613614

Prosthetic wear debris-induced peri-implant osteolysis is a major cause of aseptic loosening after total joint replacement. In this condition, wear particles released from the implant components induce a granulomatous inflammatory reaction at the interface between implant and adjacent bone, leading to progressive bone resorption and loss of fixation. The present study was undertaken to characterize definitively the phenotype of osteoclast-like cells associated with regions of peri-implant focal bone resorption and to compare the phenotypic features of these cells with those of mononucleated and multinucleated cells associated with polyethylene wear particles. Peri-implant tissues were obtained from patients undergoing hip revision surgery for aseptic loosening after total joint replacement. Cells were examined for the expression of several markers associated with the osteoclast phenotype using immunohistochemistry, histochemistry, and/or in situ hybridization. CD68 protein, a marker expressed by multiple macrophage lineage cell types, was detected in mononucleated and multinucleated cells associated with polyethylene particles and the bone surface. Cathepsin K and tartrate-resistant acid phosphatase were expressed highly in both mononucleated and multinucleated cells associated with the bone surface. Levels of expression were much lower in cells associated with polyethylene particles. High levels of beta3 integrin protein were detected in cells in contact with bone. Multinucleated cells associated with polyethylene particles exhibited faint positive staining. Calcitonin receptor mRNA expression was detected solely in multinucleated cells present in resorption lacunae on the bone surface and was absent in cells associated with polyethylene particles. Our findings provide further evidence that cells expressing the full repertoire of osteoclast phenotypic markers are involved in the pathogenesis of peri-implant osteolysis after total joint replacement. They also demonstrate that foreign body giant cells, although believed to be phenotypically and functionally distinct from osteoclasts, express many osteoclast-associated genes and gene products. However, the levels and patterns of expression of these genes in the two cell types differ. We speculate that, in addition to the role of cytokines and growth factors, the substrate with which these cells interact plays a critical role in their differential phenotypic and functional properties.


Arthroplasty, Replacement/adverse effects , Bone Resorption , Osteoclasts/pathology , Osteolysis/etiology , Prosthesis Implantation/adverse effects , Acid Phosphatase/metabolism , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Cathepsin K , Cathepsins/metabolism , Humans , Isoenzymes/metabolism , Macrophages/pathology , Osteolysis/enzymology , Osteolysis/physiopathology , RNA, Messenger/genetics , Receptors, Calcitonin/genetics , Tartrate-Resistant Acid Phosphatase
18.
Gene ; 372: 92-102, 2006 May 10.
Article En | MEDLINE | ID: mdl-16513293

The transcription factor NFATc1 plays an essential role in transducing signals from RANKL in osteoclast differentiation. To date, however, the specific transcriptional targets of NFATc1 are unknown. Expression of the beta3 integrin is required for normal osteoclast function. We therefore examined the role of NFATc1 in human beta3 integrin expression in osteoclast differentiation. Analysis of the mouse and human beta3 gene promoters revealed considerable sequence homology across a 1.3 kb region upstream of the transcription start site (TSS), with conserved NFAT binding elements present. The region -1242 to +29 (relative to the TSS) was cloned as a luciferase reporter construct (pB3-1.3) and a deletion construct removing to -997 (pB3-1) made. The deletion of 245 bp 5' removed three conserved NFAT sites including a consensus NFAT:AP-1 site. The pB3-1.3 reporter construct was induced by treatment with RANKL in the range 2.5-40 ng/ml and dose-dependently induced by co-transfection with human NFATc1 in RAW264.7 cells. The pB3-1 deletion construct was minimally induced with RANKL treatment and unresponsive to co-transfected NFATc1. Direct NFAT binding to two of the consensus NFAT sites within this 245 bp 5' region was demonstrated by EMSA and supershift with anti-NFAT antibodies. Mutation of two of the conserved NFAT sites in the -1242 to -997 fragment was required to prevent binding. The double NFAT mutant, in the context of the full-length promoter was unresponsive to RANKL treatment or co-transfected NFATc1. We generated cell-permeable TAT-dominant-negative (dn)NFATc1 fusion proteins to assess the effect of blockade of NFAT signaling. Transduction with dnNFAT inhibited RANKL induction of the human beta3 integrin promoter. Involvement of the NFATc1-calcineurin pathway in regulating the human beta3 integrin promoter was further confirmed using the calcineurin pathway inhibitory peptide 11R-VIVIT. Together these results establish the beta3 gene as a direct target of NFATc1 in RANKL-dependent osteoclast formation.


Cell Differentiation , Gene Expression Regulation , Integrin beta3/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/cytology , Promoter Regions, Genetic/genetics , Animals , Binding Sites/genetics , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Membrane Permeability , Conserved Sequence/genetics , Dose-Response Relationship, Drug , Gene Products, tat , Humans , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , Mutation/genetics , Oligopeptides/pharmacology , Protein Binding , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Response Elements/genetics , Transcriptional Activation/drug effects
19.
Immunol Rev ; 208: 228-51, 2005 Dec.
Article En | MEDLINE | ID: mdl-16313352

Rheumatoid arthritis, juvenile idiopathic arthritis, the seronegative spondyloarthropathies including psoriatic arthritis, and systemic lupus erythematosus are all examples of rheumatic diseases in which inflammation is associated with skeletal pathology. Although some of the mechanisms of skeletal remodeling are shared among these diseases, each disease has a unique impact on articular bone or on the axial or appendicular skeleton. Studies in human disease and in animal models of arthritis have identified the osteoclast as the predominant cell type mediating bone loss in arthritis. Many of the cytokines and growth factors implicated in the inflammatory processes in rheumatic diseases have also been demonstrated to impact osteoclast differentiation and function either directly, by acting on cells of the osteoclast-lineage, or indirectly, by acting on other cell types to modulate expression of the key osteoclastogenic factor receptor activator of nuclear factor (NF) kappaB ligand (RANKL) and/or its inhibitor osteoprotegerin (OPG). Further elucidation of the mechanisms responsible for inflammation-induced bone loss will potentially lead to the identification of novel therapeutic strategies for the prevention of bone loss in these diseases. In this review, we provide an overview of the cell types, inflammatory mediators, and mechanisms that are implicated in bone loss and new bone formation in inflammatory joint diseases.


Arthritis, Rheumatoid/etiology , Inflammation/complications , Lupus Erythematosus, Systemic/etiology , Spondylarthropathies/etiology , Animals , Arthritis, Juvenile/etiology , Arthritis, Rheumatoid/pathology , Bone Remodeling , Bone Resorption/etiology , Bone and Bones/pathology , Carrier Proteins/physiology , Cytokines/physiology , Glycoproteins/physiology , Humans , Membrane Glycoproteins/physiology , Osteoclasts/physiology , Osteogenesis , Osteoprotegerin , Parathyroid Hormone-Related Protein/physiology , Prostaglandins/physiology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction
20.
J Rheumatol ; 30(11): 2319-24, 2003 Nov.
Article En | MEDLINE | ID: mdl-14677171

OBJECTIVE: To assess the interpatient, interbiopsy, and intrabiopsy variability of receptor activator of nuclear factor kB ligand (RANKL) and osteoprotegerin (OPG) immunostaining within synovial tissue from rheumatoid knee joints with active synovitis, using digital image analysis. METHODS: Synovial biopsy specimens were obtained from patients with rheumatoid arthritis (RA) and active synovitis. Immunohistologic analysis was performed on frozen synovial tissue biopsy specimens from 6 patients using a monoclonal antibody (Mab) to detect RANKL (626) or OPG (805 or 8051). Patients with a minimum of 4 synovial biopsies were included in the study. Sections were evaluated by computer assisted image analysis to assess between-patient, between-biopsy, and intra-biopsy variability of OPG and RANKL protein expression. The study was designed to deliberately maximize the variability. RESULTS: Computerized image analysis of staining with Mab to RANKL and OPG revealed variance for each antibody across the 3 components of the total variability. CONCLUSION: Our study shows that variability in synovial immunostaining of RANKL and OPG protein is a significant and complex problem. We discuss methods to reduce this variability and suggest that the auspices of OMERACT may be employed to advance the study of synovium in collaborative international studies.


Arthritis, Rheumatoid/metabolism , Carrier Proteins/metabolism , Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Synovial Membrane/metabolism , Aged , Antibodies, Monoclonal , Color , Female , Humans , Image Processing, Computer-Assisted , Immunohistochemistry/methods , Male , Middle Aged , Osteoprotegerin , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptors, Tumor Necrosis Factor , Staining and Labeling
...