Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Biomed Res Int ; 2014: 942148, 2014.
Article in English | MEDLINE | ID: mdl-24605340

ABSTRACT

Rheumatoid arthritis (RA) is a systemic autoimmune disease and its targeting of the joints indicates the presence of a candidate autoantigen(s) in synovial joints. Patients with RA show immune responses in their peripheral blood to proteoglycan (PG) aggrecan. One of the most relevant animal models of RA appears to be proteoglycan-induced arthritis (PGIA), and CD4(+) T cells seem to play a crucial role in the initiation of the disease. In this review, the role of various T cell epitopes of aggrecan in the induction of autoreactive T cell activation and arthritis is discussed. We pay special attention to two critically important arthritogenic epitopes, 5/4E8 and P135H, found in the G1 and G3 domains of PG aggrecan, respectively, in the induction of autoimmune arthritis. Finally, results obtained with the recently developed PG-specific TCR transgenic mice system showed that altered T cell apoptosis, the balance of activation, and apoptosis of autoreactive T cells are critical factors in the development of autoimmunity.


Subject(s)
Aggrecans/administration & dosage , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Lymphocyte Activation/immunology , Animals , Apoptosis/drug effects , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Humans , Lymphocyte Activation/drug effects , Mice , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
2.
Genes Immun ; 13(4): 336-45, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22402741

ABSTRACT

Proteoglycan (PG)-induced arthritis (PGIA) is a murine model of rheumatoid arthritis. Arthritis-prone BALB/c mice are 100% susceptible, whereas the major histocompatibility complex-matched DBA/2 strain is completely resistant to PGIA. To reduce the size of the disease-suppressive loci for sequencing and to find causative genes of arthritis, we created a set of BALB/c.DBA/2-congenic/subcongenic strains carrying DBA/2 genomic intervals overlapping the entire Pgia26 locus on chromosome 3 (chr3) and Pgia23/Pgia12 loci on chr19 in the arthritis-susceptible BALB/c background. Upon immunization of these subcongenic strains and their wild-type (BALB/c) littermates, we identified a major Pgia26a sublocus on chr3 that suppressed disease onset, incidence and severity via controlling the complex trait of T-cell responses. The region was reduced to 3 Mbp (11.8 Mbp with flanking regions) in size and contained gene(s) influencing the production of a number of proinflammatory cytokines. Additionally, two independent loci (Pgia26b and Pgia26c) suppressed the clinical scores of arthritis. The Pgia23 locus (∼3 Mbp in size) on chr19 reduced arthritis susceptibility and onset, and the Pgia12 locus (6 Mbp) associated with low arthritis severity. Thus, we have reached the critical sizes of arthritis-associated genomic loci on mouse chr3 and chr19, which are ready for high-throughput sequencing of genomic DNA.


Subject(s)
Arthritis, Experimental/chemically induced , Autoimmune Diseases/genetics , Chromosomes, Mammalian/genetics , Genetic Loci , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Autoantibodies/blood , Autoimmune Diseases/immunology , Cartilage/immunology , Chromosome Mapping , Chromosomes, Mammalian/immunology , Cytokines/immunology , Disease Susceptibility/immunology , Female , Genetic Markers , Humans , Immunity, Cellular , Male , Mice , Mice, Congenic , Mice, Inbred BALB C , Phenotype , Proteoglycans/adverse effects , Proteoglycans/immunology , Quantitative Trait Loci
3.
Clin Exp Immunol ; 167(2): 346-55, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22236012

ABSTRACT

T cell receptor transgenic (TCR-Tg) mice specific for the arthritogenic 5/4E8 epitope in the G1 domain of cartilage proteoglycan were generated and back-crossed into arthritis-prone BALB/c background. Although more than 90% of CD4(+) T cells of all TCR-Tg lines were 5/4E8-specific, one (TCR-TgA) was highly sensitive to G1-induced or spontaneous arthritis, while another (TCR-TgB) was less susceptible. Here we studied whether fine differences in TCR signalling controlled the onset and severity of arthritis. Mice from the two TCR-Tg lines were immunized side by side with purified recombinant human G1 (rhG1) domain for G1 domain of cartilage proteoglycan (PG)-induced arthritis (GIA). TCR-TgA mice developed severe and early-onset arthritis, whereas TCR-TgB mice developed weaker arthritis with delayed onset, although TCR-TgB CD4(+) T cells expressed approximately twice more TCR-Vß4 chain protein. The more severe arthritis in TCR-TgA mice was associated with higher amounts of anti-G1 domain-specific antibodies, larger numbers of B cells and activated T helper cells. Importantly, TCR-TgB CD4(+) T cells were more sensitive to in vitro activation-induced apoptosis, correlating with their higher TCR and CD3 expression and with the increased TCR signal strength. These findings indicate that TCR signal strength determines the clinical outcome of arthritis induction: 'optimal' TCR signal strength leads to strong T cell activation and severe arthritis in TCR-TgA mice, whereas 'supra-optimal' TCR signal leads to enhanced elimination of self-reactive T cells, resulting in attenuated disease.


Subject(s)
Arthritis, Experimental/immunology , Lymphocyte Activation , Proteoglycans/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Aggrecans/immunology , Amino Acid Sequence , Animals , Apoptosis , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cartilage, Articular/immunology , Disease Models, Animal , Disease Susceptibility/immunology , Epitopes, T-Lymphocyte/immunology , Gene Dosage , Humans , Immunization , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , Phosphorylation , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Recombinant Proteins/immunology
4.
Osteoarthritis Cartilage ; 19(4): 458-65, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21262368

ABSTRACT

OBJECTIVE: To study temporomandibular joint (TMJ) involvement in an autoimmune murine model of rheumatoid arthritis (RA), a disease characterized by inflammatory destruction of the synovial joints. Although TMJ dysfunction is frequently found in RA, TMJ involvement in RA remains unclear, and TMJ pathology has not been studied in systemic autoimmune animal models of RA. METHODS: Proteoglycan (PG) aggrecan-induced arthritis (PGIA) was generated in genetically susceptible BALB/c mice. TMJs and joint tissues/cartilage were harvested for histological and immunohistochemical analyses and RNA isolation for quantitative polymerase chain-reaction. Serum cytokine levels were measured in mice with acute or chronic arthritis, and in non-arthritic control animals. RESULTS: Despite the development of destructive synovitis in the limbs, little or no synovial inflammation was found in the TMJs of mice with PGIA. However, the TMJs of arthritic mice showed evidence of aggrecanase- and matrix metalloproteinase-mediated loss of glycosaminoglycan-containing aggrecan, and in the most severe cases, structural damage of cartilage. Serum levels of pro-inflammatory cytokines, including interleukin (IL)-1ß, were elevated in arthritic animals. Expression of the IL-1ß gene was also high in the inflamed limbs, but essentially normal in the TMJs. Local expression of genes encoding matrix-degrading enzymes (aggrecanases and stromelysin) was upregulated to a similar degree in both the limbs and the TMJs. CONCLUSION: We propose that constantly elevated levels of catabolic cytokines, such as IL-1ß, in the circulation (released from inflamed joints) create a pro-inflammatory milieu within the TMJ, causing local upregulation of proteolytic enzymes and subsequent loss of aggrecan from cartilage.


Subject(s)
Cytokines/blood , Temporomandibular Joint/metabolism , Animals , Arthritis, Rheumatoid , Cartilage, Articular , Chronic Disease , Disease Models, Animal , Endopeptidases/metabolism , Glycosaminoglycans/metabolism , Immunohistochemistry , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Inbred BALB C , Osteoarthritis , Synovial Membrane/pathology , Temporomandibular Joint/pathology , Up-Regulation
5.
Osteoarthritis Cartilage ; 17(8): 1014-21, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19364543

ABSTRACT

OBJECTIVES: To assess whether glucosamine (GlcN), an oral supplement commonly taken to relieve the symptoms of osteoarthritis, modulates the immune and inflammatory responses to joint injury in organs proximal to GlcN absorption; namely, the liver and the gut-draining lymph nodes. METHOD: Using a papain-injected knee mouse model, standard histological methods were used to validate our model and document the impact of GlcN (100mg/kg/day) on groups of C57BL/6 mice (n=5). Circulating inflammatory cytokines were assessed by Luminex-based immunoassays and the relevance of this cytokine profile on proteoglycan biosynthesis evaluated using a patellar-cartilage assay. Real-time PCR was used to document the role of the liver in cytokine production. Finally, we appraised the activation of mesenteric lymph nodes (MLNs) lymphocytes by flow cytometry. RESULTS: Papain significantly degraded the proteoglycans in the injected knees by 2 days. Cartilage proteoglycan content was significantly higher in GlcN-treated, papain-injected knees at Day 14. The peak concentration of serum pro-inflammatory cytokines occurred earlier and decreased sooner in the injected, GlcN-supplemented mice; this trend was in agreement with the expression of these factors by the liver. GlcN did not alter the percentage of MLN populations but accelerated their activation. CONCLUSIONS: Oral GlcN alters the physiology of the liver and MLNs, which in turn, could indirectly alter the biology of the injured joint.


Subject(s)
Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Glucosamine/metabolism , Liver/pathology , Osteoarthritis/pathology , Animals , Arthritis, Experimental/immunology , Cartilage, Articular/immunology , Female , Knee Joint/drug effects , Knee Joint/pathology , Liver/drug effects , Lymphocytes/drug effects , Mice , Mice, Inbred C57BL , Osteoarthritis/immunology
6.
Genes Immun ; 9(7): 591-601, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18650834

ABSTRACT

Proteoglycan (PG)-induced arthritis (PGIA) is an autoimmune inflammatory disease controlled by multiple genes in the murine genome. BALB/c x DBA/2 congenic strains carrying four major PGIA chromosome loci were immunized, and positions of loci on chromosomes 3, 7, 8 and 19 (loci Pgia26, Pgia21, Pgia4 and Pgia12, respectively) were confirmed. Each congenic strain exhibited a different pattern of regulation of clinical and immunologic features of PGIA, and these features were significantly influenced by gender. Locus Pgia26 delayed PGIA onset in males and females, and the effect was associated with a lower rate of antigen-induced lymphocyte proliferation and lower production of interferon-gamma (IFN-gamma), tumor necrosis factor-alpha (TNF-alpha) and interleukin-4 (IL-4). Pgia12 similarly delayed onset in males, but the effect was achieved by elevated proliferation of PG-specific lymphocytes and enhanced production of IFN-gamma and IL-4. The effect of the Pgia21 locus was arthritis-suppressive in females but PGIA-permissive in congenic males. These opposite effects are attributed to two-fold higher serum autoantibody and IL-6 levels in males than in females. Our study supports the idea that each congenic strain represents a different immunologic subtype of PGIA, providing an explanation for the complex etiology and various clinical phenotypes of rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Models, Immunological , Phenotype , Animals , Arthritis, Experimental/chemically induced , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Crosses, Genetic , Female , Inflammation Mediators/toxicity , Male , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred DBA , Proteoglycans/toxicity
7.
Osteoarthritis Cartilage ; 15(7): 719-34, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17360199

ABSTRACT

OBJECTIVE: Human osteoarthritis (OA) is characterized by aggrecanase-mediated depletion of cartilage aggrecan. We have examined the abundance, location and some biochemical properties of the six known aggrecanases (A disintegrin and metalloproteinase with thrombospondin-like motifs 1 (ADAMTS1) 4, 5, 8, 9 and 15) in normal and OA human cartilages. METHODS: Formalin-fixed, ethylenediamine tetraacetic acid (EDTA)-decalcified sections of full-depth cartilage from human OA tibial plateaus and normal control samples were studied by confocal imaging. Probes included specific antibodies to aggrecanases and two aggrecan epitopes, as well as biotinylated hyaluronan binding protein (HABP) for hyaluronan (HA) visualization. Cartilage extracts were analyzed by Western blot for the individual proteinases and aggrecan fragments. RESULTS: ADAMTS5 was present in association with cells throughout normal cartilage and was markedly increased in OA, particularly in clonal groups in the superficial and transitional zones, where it was predominantly co-localized with HA. Consistent with the confocal analysis, a high molecular weight complex of ADAMTS5 and HA was isolated from human OA cartilage by isotonic salt extraction and chromatography on Superose 6. The complex eluted with an apparent molecular size of about 2x10(6) and contained major ADAMTS5 forms of 150, 60, 40 and 30kDa. The yield of most forms on sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) was markedly enhanced by prior digestion of the complex with either Streptomyces hyaluronidase or chondroitinase ABC. CONCLUSION: ADAMTS5 abundance and distribution in human OA cartilages is consistent with a central role for this enzyme in destructive aggrecanolysis. HA-dependent sequestration of ADAMTS5 in the pericellular matrix may be a mechanism for regulating the activity of this proteinase in human OA cartilage.


Subject(s)
ADAM Proteins/metabolism , Aggrecans/metabolism , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Endopeptidases/metabolism , Hyaluronic Acid/metabolism , Aged , Female , Humans , Male , Microscopy, Confocal/methods , Middle Aged , Osteoarthritis/metabolism
8.
Ann Rheum Dis ; 64(7): 981-7, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15640265

ABSTRACT

BACKGROUND: Proteoglycan aggrecan (PG)-induced arthritis (PGIA) is the only systemic autoimmune murine model which affects the axial skeleton, but no studies have been performed characterising the progression of spine involvement. OBJECTIVES: To follow pathological events in experimental spondylitis, and underline its clinical, radiographic, and histological similarities to human ankylosing spondylitis (AS); and to determine whether the spondyloarthropathy is a shared phenomenon with PGIA, or an "independent" disease. METHODS: Arthritis/spondylitis susceptible BALB/c and resistant DBA/2 mice, and their F1 and F2 hybrids were immunised with cartilage PG, and radiographic and histological studies were performed before onset and weekly during the progression of spondylitis. RESULTS: About 70% of the PG immunised BALB/c mice develop spondyloarthropathy (proteoglycan-induced spondylitis (PGISp), and the progression of the disease is very similar to human AS. It begins with inflammation in the sacroiliac joints and with enthesitis, and then progresses upwards, affecting multiple intervertebral disks. In F2 hybrids of arthritis/spondylitis susceptible BALB/c and resistant DBA/2 mice the incidence of arthritis was 43.5%, whereas the incidence of spondylitis was >60%. Some arthritic F2 hybrid mice had no spondylitis, whereas others developed spondylitis in the absence of peripheral arthritis. CONCLUSIONS: The PGISp model provides a valuable tool for studying autoimmune reactions in spondylitis, and identifying genetic loci associated with spondyloarthropathy.


Subject(s)
Disease Models, Animal , Intervertebral Disc , Sacroiliac Joint , Spondylitis, Ankylosing/immunology , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Disease Progression , Female , Immunization , Intervertebral Disc/immunology , Intervertebral Disc/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Models, Animal , Proteoglycans , Sacroiliac Joint/immunology , Sacroiliac Joint/pathology , Spondylitis, Ankylosing/chemically induced , Spondylitis, Ankylosing/pathology
9.
Am J Med Sci ; 327(4): 188-95, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15084914

ABSTRACT

Two autoimmune murine models--proteoglycan (aggrecan)-induced arthritis (PGIA) and collagen-induced arthritis (CIA)--were developed in parent strains, F1 and F2 hybrids of major histocompatibility complex (MHC)-matched (H-2) BALB/c x DBA/2 and MHC-unmatched (H-2/H-2) BALB/c x DBA/1 intercrosses. The major goal of this comparative study was to identify disease (model)-specific (PGIA or CIA) and shared clinical and immunologic loci in 2 types of genetic intercrosses. Qualitative (binary/susceptibility) and quantitative (severity and onset) clinical trait loci were separated and analyzed independently or together with various pathophysiologic/immunologic traits, such as antigen-specific T- and B-cell responses and cytokine production. The major quantitative trait locus (QTL) was the MHC on chromosome 17, which was especially dominant in CIA. In addition, chromosomes 3, 5, 10, and X contained shared clinical loci in both models, and a total of 8 QTLs (clinical traits together with immunologic traits) were colocalized in PGIA and CIA.


Subject(s)
Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Proteoglycans/toxicity , Quantitative Trait Loci , Animals , Antibodies/immunology , Antibodies/metabolism , Arthritis, Rheumatoid/chemically induced , Cytokines/immunology , Cytokines/metabolism , Disease Susceptibility , Genetic Linkage , Humans , Major Histocompatibility Complex , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Proteoglycans/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
10.
Clin Exp Immunol ; 129(2): 224-31, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12165077

ABSTRACT

Mucosal tolerance is a natural mechanism that prevents immunological reactions to antigens by altering the activity of immune cells of pathogenic clones without modulating the entire immune system. This 'natural immune suppression' can be exploited when antigen(s) of the target organ in an autoimmune disease is used for mucosal treatment. Being inspired by the experimental results in animal models, clinical trials using type II collagen for mucosal treatment have been conducted in rheumatoid arthritis. High-density proteoglycan (aggrecan) is another major macromolecular component in articular cartilage, and may be a candidate autoantigen for provoking immune reactions in patients with rheumatoid arthritis. Indeed, like type II collagen, systemic immunization of genetically susceptible mice with proteoglycan (PG) aggrecan induces progressive autoimmune polyarthritis. Here, we investigated whether intranasally applied PG can be effective in suppressing PG-induced arthritis (PGIA) in BALB/c mice. We found that nasal administration of 100 microg PG exerted a strong suppressive effect on both the incidence and severity of the disease, most probably by reducing responsiveness towards the immunizing PG antigen. When we transferred PGIA into genetically matched but immunodeficient SCID mice, we were able to establish a tolerized state, but only if the recipient SCID mice received lymphocytes from tolerized animals and intranasal treatment with PG was continued. Without nasally administered antigen, the transferred anergic cells recovered and arthritis rapidly developed in a severe form. Intranasal PG treatment of recipient SCID mice was ineffective when cells from non-tolerized arthritic donors were transferred, in which case the regular weekly 'tolerizing' dose of PG made the disease worse. Our results suggest that mucosal treatment in an already existing disease may result in paradoxical outcomes.


Subject(s)
Antigens/administration & dosage , Arthritis, Experimental/immunology , Arthritis, Experimental/therapy , Extracellular Matrix Proteins , Administration, Intranasal , Adoptive Transfer , Aggrecans , Animals , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/therapy , Autoimmunity , Collagen Type II/immunology , Cytokines/metabolism , Female , Humans , Immune Tolerance , Immunity, Mucosal , Immunosuppression Therapy , Interleukin-2/biosynthesis , Lectins, C-Type , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, SCID , Proteoglycans/administration & dosage , Proteoglycans/immunology , Self Tolerance , T-Lymphocytes/immunology
11.
Am J Pathol ; 159(5): 1711-21, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11696432

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha)-stimulated gene-6 (TSG-6) is up-regulated by various cytokines and growth factors. TSG-6 binds to hyaluronan in inflamed synovial tissue and forms a complex with a serine protease inter-alpha-trypsin inhibitor (IalphaI), increasing the protease inhibitory effect of IalphaI >100-fold. The TSG-6/IalphaI complex then blocks serine proteases, including the plasminogen-plasmin activation, probably the most important component in the activation processes of matrix metalloproteinases. To gain insight into the mechanisms of TSG-6 action in arthritis, we have used an autoimmune murine model (proteoglycan-induced arthritis) for systemic, and a monoarticular form of arthritis (antigen-induced arthritis) for local treatment of arthritis with recombinant mouse TSG-6 (rmTSG-6). Intravenous injection of rmTSG-6 induced a dramatic reduction of edema in acutely inflamed joints by immobilizing CD44-bound hyaluronan and, in long-term treatment, protected cartilage from degradation and blocked subchondral and periosteal bone erosion in inflamed joints. The intra-articular injection of a single dose (100 microg) of rmTSG-6 exhibited a strong chondroprotective effect for up to 5 to 7 days, preventing cartilage proteoglycan from metalloproteinase-induced degradation. In contrast, rmTSG-6 did not postpone the onset, nor reduce the incidence of arthritis. We were unable to detect any significant differences between control and rmTSG-6-treated animals when various serum markers (including pro- and anti-inflammatory cytokines, auto- and heteroantibody productions) or antigen-specific T-cell responses were compared, nor when the expressions of numerous cell surface receptors or adhesion molecules were measured. TSG-6 seems to play a critical negative regulatory feed-back function in inflammation, especially in arthritic processes.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis/drug therapy , Arthritis/pathology , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cell Adhesion Molecules/therapeutic use , Animals , Antigens/immunology , Arthritis/complications , Arthritis/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/physiopathology , Binding Sites , Binding, Competitive , Biomarkers , Cell Adhesion Molecules/metabolism , Edema/etiology , Edema/pathology , Hyaluronic Acid/metabolism , Knee Joint/drug effects , Knee Joint/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Preventive Medicine/methods , Proteoglycans/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Time Factors
12.
Mol Cell Biochem ; 222(1-2): 127-36, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11678594

ABSTRACT

Metal toxicity from sources such as orthopaedic implants was investigated in terms of immune system hyper-reactivity to metal implant alloy degradation products. Lymphocyte response to serum protein complexed with metal from implant alloy degradation was investigated in this in vitro study using primary human lymphocytes from healthy volunteers (n = 10). Cobalt chromium molybdenum alloy (Co-Cr-Mo, ASTM F-75) and titanium alloy (Ti-6Al-4V, ASTM F-136) beads (70 microm) were incubated in agitated human serum at 37 degrees Celsius to simulate naturally occurring metal implant alloy degradation processes. Particulate free serum samples, which were incubated with metal, were then separated into molecular weight based fractions. The amounts of soluble Cr and Ti within each serum fraction were measured and correlated with lymphocyte proliferation response to the individual serum fractions. Lymphocytes from each subject were cultured with 11 autologous molecular weight based serum fractions either with or without added metal. Two molecular weight ranges of human serum proteins were associated with the binding of Cr and Ti from Co-Cr-Mo and Ti implant alloy degradation (at < 30 and 180-330 kDa). High molecular weight serum proteins (approximately 180 kDa) demonstrated greater lymphocyte reactivity when complexed with metal released from Co-Cr-Mo alloy and Ti alloy than with low (5-30 kDa) and midrange (30-77 kDa) serum proteins. When the amount of lymphocyte stimulation was normalized to both the moles of metal and the moles of protein within each fraction (Metal-Protein Complex Reactivity Index, MPCRI), Cr from Co-Cr-Mo alloy degradation demonstrated approximately 10 fold greater reactivity than Ti in the higher molecular weight serum proteins (approximately 180-250 kDa). This in vitro study demonstrated a lymphocyte proliferative response to both Co-Cr-Mo and Ti alloy metalloprotein degradation products. This response was greatest when the metals were complexed with high molecular weight proteins, and with metal-protein complexes formed from Co-Cr-Mo alloy degradation.


Subject(s)
Chromium/blood , Lymphocyte Activation/drug effects , Prostheses and Implants/adverse effects , Titanium/blood , Titanium/toxicity , Vitallium/toxicity , Adult , Alloys , Biocompatible Materials/toxicity , Blood Proteins/metabolism , Female , Humans , In Vitro Techniques , Male , Middle Aged , Orthopedics , Tissue Distribution , Titanium/pharmacokinetics , Vitallium/pharmacokinetics
13.
J Immunol ; 167(1): 123-31, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11418640

ABSTRACT

CD44 is a widely expressed integral membrane glycoprotein that serves as a specific adhesion receptor for the extracellular matrix glycosaminoglycan hyaluronan. CD44 participates in a variety of physiological and pathological processes through its role in cell adhesion. Under appropriate conditions, the ectodomain of CD44 is proteolytically removed from the cell surface. In this study we show that excessive CD44 shedding can be induced in mouse fibroblasts and monocytes upon exposure of these cells to a CD44-specific Ab immobilized on plastic, whereas treatment with phorbol ester induces significantly enhanced CD44 release from the monocytes only. CD44 shedding proceeds normally in fibroblasts and monocytes deficient in TNF-alpha converting enzyme (TACE), a sheddase involved in the processing of several substrates. Conversely, activation of the CD44 protease has no effect on the release of TNF-alpha from TACE-expressing cells, although the same metalloprotease inhibitor effectively blocks both TACE and the CD44 sheddase. Concomitant with anti-CD44 Ab- or phorbol ester-induced CD44 shedding, dramatic changes are observed in cell morphology and the structure of the actin cytoskeleton. Disruption of actin assembly with cytochalasin reduces CD44 shedding, but not the release of TNF-alpha. Moreover, pharmacological activation of Rho family GTPases Rac1 and Cdc42, which regulate actin filament assembly into distinct cytoskeletal structures, has a profound effect on CD44 release. We conclude that the CD44 sheddase and TACE are distinct enzymes, and that Ab- and phorbol ester-enhanced cleavage of CD44 is controlled in a cell type-dependent fashion by Rho GTPases through the cytoskeleton.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cytoskeleton/immunology , Cytoskeleton/metabolism , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , ADAM Proteins , ADAM17 Protein , Animals , Antibodies, Monoclonal/metabolism , Bone Marrow Cells/immunology , Cell Adhesion/immunology , Cell Line, Transformed , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Culture Media, Conditioned , Fibroblasts/enzymology , Fibroblasts/immunology , Fibroblasts/metabolism , Genes, myc/immunology , Genes, ras/immunology , Hyaluronan Receptors/biosynthesis , Hyaluronan Receptors/genetics , Hydrolysis , Kinetics , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/deficiency , Mice , Mice, Inbred BALB C , Monocytes/enzymology , Monocytes/immunology , Monocytes/metabolism , RNA, Messenger/biosynthesis , Synovial Membrane/cytology , Synovial Membrane/immunology , Synovial Membrane/metabolism , Tetradecanoylphorbol Acetate/pharmacology
14.
J Biomed Mater Res ; 56(3): 427-36, 2001 Sep 05.
Article in English | MEDLINE | ID: mdl-11372061

ABSTRACT

The lymphocyte response to serum protein complexed with metal from implant alloy degradation was investigated in this in vitro study using primary human lymphocytes from healthy volunteers (n = 10). Cobalt chromium molybdenum alloy (Co-Cr-Mo, ASTM F-75) and titanium alloy (Ti-6Al-4V, ASTM F-136) beads (70 microm) were incubated in agitated human serum at 37 degrees C to simulate naturally occurring metal implant alloy degradation processes. Particulate free serum samples that had been incubated with metal were then separated into molecular weight based fractions. The amounts of soluble Cr and Ti within each serum fraction were measured and correlated with lymphocyte proliferation response to the individual serum fractions. Lymphocytes from each subject were cultured with 11 autologous molecular weight based serum fractions either with or without added metal. Two molecular weight ranges of human serum proteins were associated with the binding of Cr and Ti from Co-Cr-Mo and Ti implant alloy degradation (at <30 and 180-250 kDa). High molecular weight serum proteins ( approximately 180 kDa) demonstrated greater lymphocyte reactivity when complexed with Cr alloy and Ti alloy than low (5-30 kDa) and midrange (30-77 kDa) serum proteins. When the amount of lymphocyte stimulation was normalized to both the moles of metal and the moles of protein within each fraction (metal-protein complex reactivity index), Cr from Co-Cr-Mo alloy degradation demonstrated approximately 10-fold greater reactivity than Ti in the higher molecular weight serum proteins ( approximately 180 kDa). This in vitro study demonstrated a lymphocyte proliferative response to both Co-Cr-Mo and Ti alloy metalloprotein degradation products. This response was greatest when the metals were complexed with high molecular weight proteins, and with metal-protein complexes formed from Co-Cr-Mo alloy degradation.


Subject(s)
Absorbable Implants , Biocompatible Materials , Cobalt , Titanium , Adult , Alloys , Blood Proteins , Female , Humans , Lymphocyte Activation , Lymphocytes , Male , Middle Aged , Protein Binding
15.
J Immunol ; 166(8): 4981-6, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290777

ABSTRACT

Proteoglycan (PG)-induced arthritis (PGIA) is a novel autoimmune murine model for rheumatoid arthritis induced by immunization with cartilage PG in susceptible BALB/c mice. In this model, hyperproliferation of peripheral CD4(+) T cells has been observed in vitro with Ag stimulation, suggesting the breakdown of peripheral tolerance. Activation-induced cell death (AICD) is a major mechanism for peripheral T cell tolerance. A defect in AICD may result in autoimmunity. We report in this study that although CD4(+) T cells from both BALB/c and B6 mice, identically immunized with human cartilage PG or OVA, express equally high levels of Fas at the cell surface, CD4(+) T cells from human cartilage PG-immunized BALB/c mice, which develop arthritis, fail to undergo AICD. This defect in AICD in PGIA may lead to the accumulation of autoreactive Th1 cells in the periphery. The impaired AICD in PGIA might be ascribed to an aberrant expression of Fas-like IL-1beta-converting enzyme-inhibitory protein, which precludes caspase-8 activation at the death-inducing signaling complex, and subsequently suppresses the caspase cascade initiated by Fas-Fas ligand interaction. Moreover, this aberrant expression of Fas-like IL-1beta-converting enzyme-inhibitory protein may also mediate TCR-induced hyperproliferation of CD4(+) T cells from arthritic BALB/c mice. Our data provide the first insight into the molecular mechanism(s) of defective AICD in autoimmune arthritis.


Subject(s)
Apoptosis/immunology , Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Intracellular Signaling Peptides and Proteins , Proteoglycans/immunology , Signal Transduction/immunology , fas Receptor/physiology , Animals , Arthritis, Rheumatoid/pathology , CASP8 and FADD-Like Apoptosis Regulating Protein , CD4-Positive T-Lymphocytes/metabolism , Carrier Proteins/biosynthesis , Carrier Proteins/physiology , Cartilage, Articular/immunology , Cells, Cultured , Disease Models, Animal , Fas Ligand Protein , Female , Humans , Immunologic Memory , Immunophenotyping , Injections, Intraperitoneal , Ligands , Lymphocyte Activation/immunology , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proteoglycans/administration & dosage , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , fas Receptor/biosynthesis , fas Receptor/metabolism
16.
Arthritis Rheum ; 44(3): 682-92, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11263784

ABSTRACT

OBJECTIVE: To identify and screen the level of arthritis susceptibility in C3H murine strains known to be resistant to proteoglycan (aggrecan)-induced arthritis, and to measure and correlate various immunologic and inflammatory parameters with susceptibility to either arthritis or spondylitis in various C3H substrains. METHODS: Mice of 10 C3H substrains (subcolonies) were immunized with cartilage proteoglycan (aggrecan) for induction of arthritis. Animals were assessed for clinical symptoms, and the peripheral joints and spine were studied by histologic methods. Proteoglycan-specific T cell responses (T cell proliferation and production of interleukin-2 [IL-2], interferon-y, and IL-4) and the B cell response to lipopolysaccharide (LPS) were measured in spleen cell cultures. Serum levels of heteroantibodies and autoantibodies as well as various cytokines (IL-6, IL-10, IL-12, and tumor necrosis factor alpha) and soluble CD44 were determined by enzyme-linked immunosorbent assay. RESULTS: Immunization with cartilage proteoglycan induced severe arthritis in the C3H/HeJCr substrain (95-100% incidence), whereas the original parent mice of the C3H/HeJ colony were resistant to proteoglycan (aggrecan)-induced arthritis. Furthermore, the progressive polyarthritis that is characteristic in susceptible C3H/HeJCr mice was accompanied by progressive inflammation around the spine. In subsequent experiments, 10 different C3H colonies with largely identical genetic backgrounds (all originating from the National Institutes of Health or Jackson Laboratory) exhibited extreme differences in susceptibility. Although none of the laboratory findings, including LPS hyporesponsiveness, immunologic parameters, and inflammatory markers, showed a correlation with susceptibility or resistance in the C3H/HeJCr and C3H/HeJ substrains, respectively, significant differences were found when all arthritic C3H mice were compared with all nonarthritic animals, regardless of their substrain origin. CONCLUSION: Because many of the C3H substrains lost arthritis susceptibility or acquired resistance, our results suggest that a preferred site for a mutation(s) in a gene(s) in a relatively upstream position of the inflammatory cascade is present. This is the first autoimmune model that exhibits extreme differences in arthritis susceptibility in the same murine strain, and is therefore a valuable tool for identification of arthritis-susceptible (or arthritis-suppressive) genes.


Subject(s)
Arthritis/chemically induced , Proteoglycans/genetics , Spondylitis, Ankylosing/chemically induced , Animals , Arthritis/genetics , Female , Genetic Predisposition to Disease , Intervertebral Disc/pathology , Joints/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Spine/pathology , Spondylitis, Ankylosing/genetics
17.
Arthritis Rheum ; 44(12): 2922-31, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11762953

ABSTRACT

OBJECTIVE: To study the role of CD44, the principal hyaluronan (HA) receptor, in experimental arthritis. METHODS: We generated CD44 gene deficiency in arthritis-susceptible DBA/1LacJ mice to study the role of CD44 directly in collagen-induced arthritis (CIA). Wild-type and CD44-deficient mice were immunized with chicken type II collagen, and the onset and severity of CIA were monitored up to day 64. The immune status of immunized mice was determined at the end of the experiments. Cell transfer experiments were performed to monitor lymphocyte traffic to the inflamed joints. RESULTS: Mice homozygous for the CD44 mutation developed normally and showed no phenotypic defects. Although they showed a normal response to immunization with type II collagen and had Th1/Th2 ratios comparable with those in wild-type animals, CD44-deficient mice exhibited significant reductions in both the incidence and severity of CIA. This was accompanied by altered serum levels of HA, reduced expression of L-selectin, and a delayed entry of intravenously injected CD44-deficient donor lymphocytes into the arthritic joints of recipient mice. CONCLUSION: While CD44 is not essential for morphogenesis and autoimmunity, this cell surface receptor seems to play an important role in the development of arthritis, most likely by directing leukocyte traffic to the site of inflammation.


Subject(s)
Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Hyaluronan Receptors/genetics , Animals , Arthritis, Experimental/epidemiology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Adhesion Molecules/genetics , Cell Communication/immunology , Chemotaxis, Leukocyte/immunology , Collagen Type II , Hyaluronic Acid/blood , Immunity, Innate , Immunization , Incidence , Joints/immunology , Male , Mice , Mice, Inbred DBA , Mice, Knockout , Rats , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology
18.
J Immunol ; 165(9): 5278-86, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11046062

ABSTRACT

Proteoglycan-induced arthritis (PGIA) is a murine model for rheumatoid arthritis (RA) both in terms of its pathology and its genetics. PGIA can only be induced in susceptible mouse strains and their F(2) progeny. Using the F(2) hybrids resulting from an F(1) intercross of a newly identified susceptible (C3H/HeJCr) and an established resistant (C57BL/6) strain of mouse, our goals were to: 1) identify the strain-specific loci that confer PGIA susceptibility, 2) determine whether any pathophysiological parameters could be used as markers that distinguish between nonarthritic and arthritic mice, and 3) analyze the effect of the MHC haplotype on quantitative trait loci (QTL) detection. To identify QTLs, we performed a genome scan on the F(2) hybrids. For pathophysiological analyses, we measured pro- and antiinflammatory cytokines such as IL-1, IL-6, IFN-gamma, IL-4, IL-10, IL-12, Ag-specific T cell proliferation and IL-2 production, serum IgG1 and IgG2 levels of both auto- and heteroantibodies, and soluble CD44. We have identified four new PGIA-linked QTLs (Pgia13 through Pgia16) and confirmed two (Pgia5, Pgia10) from our previous study. All new MHC-independent QTLs were associated with either disease onset or severity. Comprehensive statistical analysis demonstrated that while soluble CD44, IL-6, and IgG1 vs. IgG2 heteroantibody levels differed significantly between the arthritic and nonarthritic groups, only Ab-related parameters colocalized with the QTLs. Importantly, the mixed haplotype (H-2(b) and H-2(k)) of the C3H x C57BL/6 F(2) intercross reduced the detection of several previously identified QTLs to suggestive levels, indicating a masking effect of unmatched MHCs.


Subject(s)
Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Crosses, Genetic , Genetic Predisposition to Disease/genetics , Quantitative Trait, Heritable , Animals , Arthritis, Rheumatoid/physiopathology , Disease Models, Animal , Female , Genetic Markers/genetics , Genetic Markers/immunology , Genome , Injections, Intraperitoneal , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Polymorphism, Genetic/immunology , Proteoglycans/administration & dosage , Proteoglycans/immunology , Species Specificity , Statistics, Nonparametric
19.
J Biomed Mater Res ; 53(5): 480-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10984695

ABSTRACT

The determination of biocompatibility has been dominated historically by the characterization of candidate materials based upon the observation of adverse host responses. However, some adverse responses are subtle in clinical settings and continue to foster debate and investigation. One of these responses is "metal allergy" or hypersensitivity to metallic biomaterials. Current methods used to diagnose hypersensitivity reactions, such as dermal patch testing and migration inhibition assays, are not well accepted in orthopedic practice as a means for the characterization of hypersensitivity to metallic joint-replacement components. An increasing need to resolve whether metal sensitivity may be a significant and/or predisposing factor for eliciting an over-aggressive immune response in patients with metallic implant components requires improved and standardized widespread study. Here we present three in vitro methodologies: (1) a proliferation assay, (2) cytokine analysis using ELISA, and (3) a migration inhibition assay. When in conjunction with one another, these assays may be used to more comprehensively quantify metal-induced hypersensitivity responses. Therefore, these methodologies are detailed with the intent of facilitating multi-center large-scale studies. In the following cases, a multi-assay approach for measuring the prevalence of delayed-type hypersensitivity in orthopedic patients shows the propensity to yield a more comprehensive and, therefore, more conclusive determination than currently employed patch testing or single assay techniques.


Subject(s)
Alloys/toxicity , Arthroplasty, Replacement, Hip/adverse effects , Hip Prosthesis/adverse effects , Hypersensitivity, Delayed/diagnosis , Lymphocytes/immunology , Metals/adverse effects , Monocytes/physiology , Adult , Aged , Chemotaxis, Leukocyte/drug effects , Cytokines/blood , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hypersensitivity, Delayed/etiology , Hypersensitivity, Delayed/prevention & control , In Vitro Techniques , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Male , Middle Aged , Monocytes/drug effects
20.
J Immunol ; 165(6): 2970-4, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10975804

ABSTRACT

Most thymocytes that have not successfully rearranged their TCR genes or that express a receptor with subthreshold avidity for self-Ag/MHC enter a default apoptosis pathway, death by neglect. Spontaneous thymocyte apoptosis (STA), at least in part, may mimic this process in vitro. However, the molecular mechanism(s) by which thymocytes undergo this spontaneous apoptosis remains unknown. Here, we report that caspsase-1 and caspase-3 are activated during STA, but these caspases are dispensable for this apoptotic process. The inhibition of STA by a pan-caspase inhibitor, zVAD, suggests that multiple caspase pathways exist. Importantly, the early release of cytochrome c from mitochondria closely correlates with the degradation of Bcl-2 and Bcl-xL and a decrease in the ratios of Bcl-2 and Bcl-xL to Bax during STA. These findings suggest that the degradation of Bcl-2 and Bcl-xL may favor Bax to induce cytochrome c release from mitochondria, which subsequently activates downstream caspases in STA. Our data provide the first biochemical insight into the molecular mechanism of STA.


Subject(s)
Apoptosis/immunology , Mitochondria/immunology , Signal Transduction/immunology , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Caspase 1/physiology , Caspase 3 , Caspases/physiology , Cytochrome c Group/metabolism , Enzyme Activation/immunology , Kinetics , Membrane Potentials/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondria/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , T-Lymphocytes/enzymology , Thymus Gland/enzymology , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...