Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 20(3)2019 Jan 29.
Article in English | MEDLINE | ID: mdl-30699963

ABSTRACT

A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4 and ADAMTS-5 are the principal aggrecanases in mice and humans; however, mice lacking the catalytic domain of both enzymes (TS-4/5∆cat) have no skeletal phenotype, suggesting there is an alternative aggrecanase for modulating normal growth and development in these mice. We previously identified aggrecanase activity that (a) cleaved at E↓G rather than E↓A bonds in the aggrecan core protein, and (b) was upregulated by retinoic acid but not IL-1α. The present study aimed to identify the alternative aggrecanase. Femoral head cartilage explants from TS-4/5∆cat mice were stimulated with IL-1α or retinoic acid and total RNA was analysed by microarray. In addition to ADAMTS-5 and matrix metalloproteinase (MMP)-13, which are not candidates for the novel aggrecanase, the microarray analyses identified MMP-11, calpain-5 and ADAMTS-9 as candidate aggrecanases upregulated by retinoic acid. When calpain-5 and MMP-11 failed to meet subsequent criteria, ADAMTS-9 emerged as the most likely candidate for the novel aggrecanase. Immunohistochemistry revealed ADAMTS-9 expression throughout the mouse growth plate and strong expression, particularly in the proliferative zone of the TS-4/5-∆cat mice. In conclusion, ADAMTS-9 has a novel specificity for aggrecan, cleaving primarily at E↓G rather than E↓A bonds in mouse cartilage. ADAMTS-9 might have more important roles in normal skeletal development compared with ADAMTS-4 and ADAMTS-5, which have key roles in joint pathology.


Subject(s)
ADAMTS4 Protein/metabolism , ADAMTS5 Protein/metabolism , ADAMTS9 Protein/metabolism , Cartilage/metabolism , Endopeptidases/metabolism , ADAMTS9 Protein/genetics , Aggrecans/metabolism , Animals , Arthritis/genetics , Arthritis/metabolism , Cells, Cultured , Immunohistochemistry , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Mice , RNA, Messenger/metabolism
2.
Respir Res ; 19(1): 155, 2018 Aug 20.
Article in English | MEDLINE | ID: mdl-30126423

ABSTRACT

BACKGROUND: Prenatal glucocorticoid treatment decreases alveolar tissue volumes and facilitates fetal lung maturation, however the mechanisms responsible are largely unknown. This study examines whether changes in versican levels or sulphation patterns of chondroitin sulphate (CS) side chains, are associated with glucocorticoid-induced reductions in peri-alveolar tissue volumes. METHODS: Lung tissue was collected from 1) fetal sheep at 131 ± 0.1 days gestational age (GA) infused with cortisol (122-131d GA) to prematurely induce a pre-parturient-like rise in circulating cortisol, 2) fetal sheep at 143d GA bilaterally adrenalectomised (ADX) at 112d GA to remove endogenous cortisol and 3) fetal sheep at 124d GA in which bolus doses (2 × 11.4 mg) of betamethasone were administered to the pregnant ewe. The level and distribution of versican and CS glycosaminoglycans (GAG) were determined using immunohistochemistry (IHC). Fluorophore assisted carbohydrate electrophoresis (FACE) was used to determine changes in CS sulphation patterns. RESULTS: Cortisol infusion significantly decreased chondrotin-6-sulphate levels (C-6-S) to 16.4 ± 0.7 AU, compared with saline-infused fetuses (18.9 ± 0.7 AU: p = 0.04) but did not significantly alter the level of versican or chondroitin-4-sulphate (C-4-S). ADX significantly increased the level of C-4-S (28.2 ± 2.2 AU), compared with sham-operated fetuses (17.8 ± 2.0 AU; p = 0.006) without altering versican or C-6-S levels. Betamethasone significantly decreased versican, C-4-S and C-6-S in the fetal sheep lung (19.2 ± 0.9 AU, 24.9 ± 1.4 AU and 23.2 ± 1.0 AU, respectively), compared with saline-exposed fetuses (24.3 ± 0.4 AU, p = 0.0004; 33.3±0.6 AU, p = 0.0003; 29.8±1.3 AU, 0.03, respectively). CONCLUSIONS: These results indicate that glucocorticoids alter versican levels and CS side chain microstructure in alveolar lung tissue. Betamethasone appears to have a greater impact on versican and CS side chains than cortisol.


Subject(s)
Chondroitin Sulfates/biosynthesis , Fetal Development/physiology , Glucocorticoids/pharmacology , Lung/metabolism , Proteoglycans/biosynthesis , Versicans/biosynthesis , Animals , Female , Fetal Development/drug effects , Fetus , Lung/drug effects , Lung/growth & development , Pregnancy , Sheep
3.
JCI Insight ; 3(6)2018 03 22.
Article in English | MEDLINE | ID: mdl-29563338

ABSTRACT

Pain is the predominant symptom of osteoarthritis, but the connection between joint damage and the genesis of pain is not well understood. Loss of articular cartilage is a hallmark of osteoarthritis, and it occurs through enzymatic degradation of aggrecan by cleavage mediated by a disintegrin and metalloproteinase with thrombospondin motif 4 (ADAMTS-4) or ADAMTS-5 in the interglobular domain (E373-374A). Further cleavage by MMPs (N341-342F) releases a 32-amino-acid aggrecan fragment (32-mer). We investigated the role of this 32-mer in driving joint pain. We found that the 32-mer excites dorsal root ganglion nociceptive neurons, both in culture and in intact explants. Treatment of cultured sensory neurons with the 32-mer induced expression of the proalgesic chemokine CCL2. These effects were mediated through TLR2, which we demonstrated was expressed by nociceptive neurons. In addition, intra-articular injection of the 32-mer fragment provoked knee hyperalgesia in WT but not Tlr2-null mice. Blocking the production or action of the 32-mer in transgenic mice prevented the development of knee hyperalgesia in a murine model of osteoarthritis. These findings suggest that the aggrecan 32-mer fragment directly activates TLR2 on joint nociceptors and is an important mediator of the development of osteoarthritis-associated joint pain.


Subject(s)
Aggrecans/metabolism , Arthralgia/metabolism , Osteoarthritis/metabolism , Toll-Like Receptor 2/metabolism , ADAMTS4 Protein/metabolism , ADAMTS5 Protein/metabolism , Animals , Calcium/metabolism , Cartilage, Articular/metabolism , Chemokine CCL2/metabolism , Disease Models, Animal , Ganglion Cysts/metabolism , Matrix Metalloproteinases , Mice , Mice, Knockout , Mice, Transgenic , Osteoarthritis/genetics , Toll-Like Receptor 2/genetics
4.
Arthritis Rheumatol ; 70(3): 383-395, 2018 03.
Article in English | MEDLINE | ID: mdl-29145712

ABSTRACT

OBJECTIVE: To identify candidate microRNAs (miRNAs) that potentially regulate the initiation and progression of osteoarthritis (OA). METHODS: OA was induced in 10-12-week-old male wild-type C57BL/6 mice and in mice resistant to aggrecanase cleavage (Acan p.374ALGS→374NVYS) by destabilization of the medial meniscus (DMM). Pathologic changes of OA were scored histologically. RNA from cartilage and subchondral bone was harvested in parallel by laser microdissection at 1 week and 6 weeks postsurgery. Global miRNA expression profiling was performed using Agilent microarrays and was validated by quantitative polymerase chain reaction analysis. RESULTS: Wild-type DMM mice had characteristic cartilage degeneration, subchondral bone sclerosis, and osteophyte formation. While no miRNA dysregulation was seen in subchondral bone, 139 miRNAs were differentially expressed in cartilage obtained at 1 and/or 6 weeks after OA initiation from wild-type mice that underwent DMM. To prioritize OA candidates, dysregulated miRNAs with human orthologs were filtered, and paired miRNA/messenger RNA (mRNA) expression analysis was conducted to identify those with corresponding changes in mRNA target transcripts in the DMM mouse cartilage. An important cohort also overlapped with miRNAs identified in human end-stage OA. Comparisons of miRNA dysregulation in DMM mouse cartilage where aggrecan cleavage was genetically ablated demonstrated that all candidates were independent of aggrecan breakdown, earmarking these as important to the critical stages of OA initiation. Furthermore, functional enrichment analysis and data annotation revealed the responses to mechanical stimuli, apoptotic processes, and core extracellular matrix structural and regulatory factors to be potentially influenced by OA-dysregulated miRNA/mRNA networks. CONCLUSION: Our comprehensive analyses identified high-priority miRNA candidates that have potential as biomarkers and therapeutic targets in human OA.


Subject(s)
Aggrecans/metabolism , Arthritis, Experimental/metabolism , Cartilage, Articular/metabolism , MicroRNAs/metabolism , Osteoarthritis/metabolism , Animals , Cartilage, Articular/pathology , Disease Models, Animal , Disease Progression , Endopeptidases , Humans , Male , Mice , Mice, Inbred C57BL , Microarray Analysis , Real-Time Polymerase Chain Reaction
5.
J Biomech ; 49(9): 1634-1640, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27086115

ABSTRACT

Aggrecan loss in human and animal cartilage precedes clinical symptoms of osteoarthritis, suggesting that aggrecan loss is an initiating step in cartilage pathology. Characterizing early stages of cartilage degeneration caused by aging and overuse is important in the search for therapeutics. In this study, atomic force microscopy (AFM)-based force-displacement micromechanics, AFM-based wide bandwidth nanomechanics (nanodynamic), and histologic assessments were used to study changes in distal femur cartilage of wildtype mice and mice in which the aggrecan interglobular domain was mutated to make the cartilage aggrecanase-resistant. Half the animals were subjected to voluntary running-wheel exercise of varying durations. Wildtype mice at three selected age groups were compared. While histological assessment was not sensitive enough to capture any statistically significant changes in these relatively young populations of mice, micromechanical assessment captured changes in the quasi-equilibrium structural-elastic behavior of the cartilage matrix. Additionally, nanodynamic assessment captured changes in the fluid-solid poroelastic behavior and the high frequency stiffness of the tissue, which proved to be the most sensitive assessment of changes in cartilage associated with aging and joint-overuse. In wildtype mice, aging caused softening of the cartilage tissue at the microscale and at the nanoscale. Softening with increased animal age was found at high loading rates (frequencies), suggesting an increase in hydraulic permeability, with implications for loss of function pertinent to running and impact-injury. Running caused substantial changes in fluid-solid interactions in aggrecanase-resistant mice, suggestive of tissue degradation. However, higher nanodynamic stiffness magnitude and lower hydraulic permeability was observed in running aggrecanase-resistant mice compared to running wildtype controls at the same age, thereby suggesting protection from joint-overuse.


Subject(s)
Aggrecans/genetics , Cartilage/metabolism , Gene Knock-In Techniques , Mechanical Phenomena , Nanotechnology , Aggrecans/metabolism , Aging/metabolism , Animals , Biomechanical Phenomena , Cattle , Endopeptidases/metabolism , Femur/metabolism , Humans , Mice , Microscopy, Atomic Force , Osteoarthritis/metabolism , Permeability
6.
J Proteome Res ; 15(3): 1033-50, 2016 Mar 04.
Article in English | MEDLINE | ID: mdl-26794603

ABSTRACT

The destruction of articular cartilage in osteoarthritis involves chondrocyte dysfunction and imbalanced extracellular matrix (ECM) homeostasis. Pro-inflammatory cytokines such as interleukin-1α (IL-1α) contribute to osteoarthritis pathophysiology, but the effects of IL-1α on chondrocytes within their tissue microenvironment have not been fully evaluated. To redress this we used label-free quantitative proteomics to analyze the chondrocyte response to IL-1α within a native cartilage ECM. Mouse femoral heads were cultured with and without IL-1α, and both the tissue proteome and proteins released into the media were analyzed. New elements of the chondrocyte response to IL-1α related to cellular stress included markers for protein misfolding (Armet, Creld2, and Hyou1), enzymes involved in glutathione biosynthesis and regeneration (Gstp1, Gsto1, and Gsr), and oxidative stress proteins (Prdx2, Txn, Atox1, Hmox1, and Vnn1). Other proteins previously not associated with the IL-1α response in cartilage included ECM components (Smoc2, Kera, and Crispld1) and cysteine proteases (cathepsin Z and legumain), while chondroadherin and cartilage-derived C-type lectin (Clec3a) were identified as novel products of IL-1α-induced cartilage degradation. This first proteome-level view of the cartilage IL-1α response identified candidate biomarkers of cartilage destruction and novel targets for therapeutic intervention in osteoarthritis.


Subject(s)
Cartilage, Articular/metabolism , Chondrocytes/metabolism , Animals , Cartilage, Articular/pathology , Cells, Cultured , Interleukin-1alpha/physiology , Mice, Inbred C57BL , Proteome/metabolism , Stress, Physiological
7.
Arthritis Rheumatol ; 68(5): 1165-71, 2016 05.
Article in English | MEDLINE | ID: mdl-26663140

ABSTRACT

OBJECTIVE: The pathogenesis of osteoarthritis (OA) is poorly understood. Loss of the proteoglycan aggrecan from cartilage is an early event. Recently, we identified a role for the JNK pathway, particularly JNK-2, in human articular chondrocytes in vitro in regulating aggrecan degradation. The present study was undertaken to investigate whether JNK-2 has a similar function in vivo and to examine its role in gene expression. METHODS: Aggrecan fragments were analyzed by Western blotting. OA was induced by destabilization of the medial meniscus (DMM) and assessed at 4, 8, and 12 weeks after surgery. Knee sections were stained with Safranin O. Medial compartments were scored by histologic grading for aggrecan loss and cartilage damage. RNA was extracted from JNK-2(-/-) and wild-type mouse knees 6 hours after DMM or after interleukin-1 stimulation of the proximal epiphysis, and expression of 33 DMM-regulated genes was analyzed with quantitative polymerase chain reaction-customized array cards. RESULTS: In vitro, basal and interleukin-1- or tumor necrosis factor-stimulated release of aggrecanase-generated aggrecan fragments was greatly reduced in cartilage from JNK-2(-/-) mice. In the OA model, JNK-2(-/-) mice exhibited significant reduction of aggrecanase-generated fragments and cartilage damage. Of 33 genes investigated, 13 were significantly down-regulated in JNK-2(-/-) mice compared with wild-type mice, following DMM. These included Has1, Adamts4, Tnf, Il6, Il18, Il18rap, Il1a, Inhba, Cd68, Ngf, Ccr2, Wnt16, and Tnfaip6, but not Adamts5. CONCLUSION: Our results demonstrate that JNK-2 regulates aggrecan degradation in cultured murine cartilage and surgically induced OA in vivo following mechanical destabilization of the knee joint. This implicates the JNK signaling pathway in OA and suggests potential novel approaches to therapy.


Subject(s)
Aggrecans/metabolism , Arthritis, Experimental/genetics , Cartilage, Articular/metabolism , Gene Expression Regulation/genetics , Mitogen-Activated Protein Kinase 9/genetics , Osteoarthritis, Knee/genetics , RNA, Messenger/metabolism , Aggrecans/drug effects , Animals , Blotting, Western , Cartilage, Articular/drug effects , Disease Models, Animal , Endopeptidases/drug effects , Endopeptidases/metabolism , Epiphyses , Femur , Gene Expression Regulation/drug effects , Hip Joint , Interleukin-1/pharmacology , Knee Joint , Male , Menisci, Tibial/surgery , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology
8.
J Biol Chem ; 291(7): 3197-208, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26668318

ABSTRACT

The metalloproteinase ADAMTS-5 (A disintegrin and metalloproteinase with thrombospondin motifs) degrades aggrecan, a proteoglycan essential for cartilage structure and function. ADAMTS-5 is the major aggrecanase in mouse cartilage, and is also likely to be the major aggrecanase in humans. ADAMTS-5 is a multidomain enzyme, but the function of the C-terminal ancillary domains is poorly understood. We show that mutant ADAMTS-5 lacking the catalytic domain, but with a full suite of ancillary domains inhibits wild type ADAMTS activity, in vitro and in vivo, in a dominant-negative manner. The data suggest that mutant ADAMTS-5 binds to wild type ADAMTS-5; thus we tested the hypothesis that ADAMTS-5 associates to form oligomers. Co-elution, competition, and in situ PLA experiments using full-length and truncated recombinant ADAMTS-5 confirmed that ADAMTS-5 molecules interact, and showed that the catalytic and disintegrin-like domains support these intermolecular interactions. Cross-linking experiments revealed that recombinant ADAMTS-5 formed large, reduction-sensitive oligomers with a nominal molecular mass of ∼ 400 kDa. The oligomers were unimolecular and proteolytically active. ADAMTS-5 truncates comprising the disintegrin and/or catalytic domains were able to competitively block full-length ADAMTS-5-mediated aggrecan cleavage, measured by production of the G1-EGE(373) neoepitope. These results show that ADAMTS-5 oligomerization is required for full aggrecanase activity, and they provide evidence that blocking oligomerization inhibits ADAMTS-5 activity. The data identify the surface provided by the catalytic and disintegrin-like domains of ADAMTS-5 as a legitimate target for the design of aggrecanase inhibitors.


Subject(s)
ADAM Proteins/metabolism , Aggrecans/metabolism , Arthritis, Experimental/enzymology , Knee Joint/enzymology , ADAM Proteins/chemistry , ADAM Proteins/genetics , ADAM Proteins/isolation & purification , ADAMTS5 Protein , Aggrecans/isolation & purification , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Catalytic Domain , Cross-Linking Reagents/chemistry , Crosses, Genetic , Dimerization , Enzyme Activation , Gene Deletion , HEK293 Cells , Humans , Knee Joint/immunology , Knee Joint/pathology , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Weight , Mutant Proteins , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Proteolysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
10.
Arthritis Rheumatol ; 67(5): 1240-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25707860

ABSTRACT

OBJECTIVE: To determine whether an aggrecan 32-mer fragment derived from dual ADAMTS and matrix metalloproteinase (MMP) cleavage in the aggrecan interglobular domain was bioactive and, if so, to elucidate its mechanism of action. METHODS: Mouse primary chondrocytes, synovial fibroblasts, or peritoneal macrophages, human primary chondrocytes, and cells or cell lines from myeloid differentiation factor 88 (MyD88)-deficient and Toll-like receptor 2 (TLR-2)-deficient mice were stimulated with synthetic mouse 32-mer peptide, human 32-mer peptide, a 32-mer scrambled peptide, or native, glycosylated 32-mer peptide. Cells stimulated with 32-mer peptide were analyzed for changes in messenger RNA (mRNA) expression by quantitative polymerase chain reaction. Conditioned medium was analyzed for levels of interleukin-6 protein by an AlphaLISA or for levels of MMP-3 and MMP-13 protein by Western blotting. NF-κB activation was measured in a luciferase reporter assay. RESULTS: Treatment of mouse cells or cartilage explants with 32-mer peptide or scrambled peptide revealed that the 32-mer peptide, but not the scrambled peptide, had antianabolic, procatabolic, and proinflammatory bioactivity in vitro. Chondrocytes, synovial fibroblasts, and macrophages from MyD88-deficient mice failed to respond to 32-mer peptide stimulation. A macrophage cell line derived from TLR-2-deficient mice also failed to respond to 32-mer peptide stimulation. Stimulation of human chondrocytes with human 32-mer peptide increased the expression of catabolic markers at the mRNA and protein levels. Mouse and human 32-mer peptide stimulated NF-κB activation in a TLR-2-dependent reporter assay, and the response of chondrocytes from both species to native, glycosylated 32-mer peptide was similar to the response to synthetic peptides. CONCLUSION: The aggrecan 32-mer fragment is a novel endogenous ligand of TLR-2 with the potential to accelerate cartilage destruction in vivo.


Subject(s)
Aggrecans/pharmacology , Chondrocytes/drug effects , Fibroblasts/drug effects , Macrophages, Peritoneal/drug effects , Myeloid Differentiation Factor 88/drug effects , Peptide Fragments/pharmacology , RNA, Messenger/metabolism , Toll-Like Receptor 2/drug effects , Adolescent , Aggrecans/metabolism , Animals , Blotting, Western , Cell Line , Chondrocytes/metabolism , Fibroblasts/metabolism , Humans , Interleukin-6/metabolism , Macrophages, Peritoneal/metabolism , Male , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , Mice , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Peptide Fragments/metabolism , Real-Time Polymerase Chain Reaction , Synovial Membrane/cytology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism
11.
J Biomech ; 48(1): 162-5, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25435386

ABSTRACT

Murine models of osteoarthritis (OA) and post-traumatic OA have been widely used to study the development and progression of these diseases using genetically engineered mouse strains along with surgical or biochemical interventions. However, due to the small size and thickness of murine cartilage, the relationship between mechanical properties, molecular structure and cartilage composition has not been well studied. We adapted a recently developed AFM-based nano-rheology system to probe the dynamic nanomechanical properties of murine cartilage over a wide frequency range of 1 Hz to 10 kHz, and studied the role of glycosaminoglycan (GAG) on the dynamic modulus and poroelastic properties of murine femoral cartilage. We showed that poroelastic properties, highlighting fluid-solid interactions, are more sensitive indicators of loss of mechanical function compared to equilibrium properties in which fluid flow is negligible. These fluid-flow-dependent properties include the hydraulic permeability (an indicator of the resistance of matrix to fluid flow) and the high frequency modulus, obtained at high rates of loading relevant to jumping and impact injury in vivo. Utilizing a fibril-reinforced finite element model, we estimated the poroelastic properties of mouse cartilage over a wide range of loading rates for the first time, and show that the hydraulic permeability increased by a factor ~16 from knormal=7.80×10(-16)±1.3×10(-16) m(4)/N s to kGAG-depleted=1.26×10(-14)±6.73×10(-15) m(4)/N s after GAG depletion. The high-frequency modulus, which is related to fluid pressurization and the fibrillar network, decreased significantly after GAG depletion. In contrast, the equilibrium modulus, which is fluid-flow independent, did not show a statistically significant alteration following GAG depletion.


Subject(s)
Cartilage/physiology , Glycosaminoglycans/physiology , Microscopy, Atomic Force , Osteoarthritis , Rheology/methods , Aggrecans/metabolism , Animals , Biomechanical Phenomena , Disease Models, Animal , Extracellular Matrix/metabolism , Femur , Mice , Mice, Inbred C3H , Permeability
12.
J Immunol ; 191(3): 1404-12, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23797671

ABSTRACT

Mouse mast cell protease (mMCP)-6-null C57BL/6 mice lost less aggrecan proteoglycan from the extracellular matrix of their articular cartilage during inflammatory arthritis than wild-type (WT) C57BL/6 mice, suggesting that this mast cell (MC)-specific mouse tryptase plays prominent roles in articular cartilage catabolism. We used ex vivo mouse femoral head explants to determine how mMCP-6 and its human ortholog hTryptase-ß mediate aggrecanolysis. Exposure of the explants to recombinant hTryptase-ß, recombinant mMCP-6, or lysates harvested from WT mouse peritoneal MCs (PMCs) significantly increased the levels of enzymatically active matrix metalloproteinases (MMP) in cartilage and significantly induced aggrecan loss into the conditioned media, relative to replicate explants exposed to medium alone or lysates collected from mMCP-6-null PMCs. Treatment of cartilage explants with tetramer-forming tryptases generated aggrecan fragments that contained C-terminal DIPEN and N-terminal FFGVG neoepitopes, consistent with MMP-dependent aggrecanolysis. In support of these data, hTryptase-ß was unable to induce aggrecan release from the femoral head explants obtained from Chloe mice that resist MMP cleavage at the DIPEN↓FFGVG site in the interglobular domain of aggrecan. In addition, the abilities of mMCP-6-containing lysates from WT PMCs to induce aggrecanolysis were prevented by inhibitors of MMP-3 and MMP-13. Finally, recombinant hTryptase-ß was able to activate latent pro-MMP-3 and pro-MMP-13 in vitro. The accumulated data suggest that human and mouse tetramer-forming tryptases are MMP convertases that mediate cartilage damage and the proteolytic loss of aggrecan proteoglycans in arthritis, in part, by activating the zymogen forms of MMP-3 and MMP-13, which are constitutively present in articular cartilage.


Subject(s)
Aggrecans/metabolism , Cartilage, Articular/metabolism , Mast Cells/immunology , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , Animals , Arthritis/metabolism , Cells, Cultured , Enzyme Precursors/metabolism , Extracellular Matrix/metabolism , Inflammation , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Tryptases/deficiency , Tryptases/genetics , Tryptases/metabolism
13.
Arthritis Rheum ; 65(6): 1547-60, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23436205

ABSTRACT

OBJECTIVE: To identify changes in gene expression in mice with osteoarthritis (OA) in order to explore the mechanisms of the disease. METHODS: Gene expression profiling was performed in cartilage from mice with surgically induced OA. We used wild-type (WT) mice and Adamts5Δcat mice, in which ADAMTS-5 activity is lacking and aggrecan loss and cartilage erosion are inhibited, to distinguish gene expression changes that are independent of ADAMTS-5 activity and cartilage breakdown. Mechanical instability was introduced into the knee joints of 10-week-old male mice via surgical destabilization of the medial meniscus (DMM). Cartilage from the developing lesion in the destabilized medial meniscus and corresponding regions in sham-operated joints was harvested by microdissection at 1, 2, and 6 weeks postsurgery, and RNA was extracted, amplified, and hybridized to whole-genome microarrays. RESULTS: Several previously identified OA-related genes, including Ptgs2, Crlf1, and Inhba, and novel genes, such as Phdla2 and Il11, were up-regulated in both WT mice and Adamts5Δcat mice, indicating that they are independent of ADAMTS-5 activity. The altered expression of other genes, including Col10a1, the sentinel marker of cartilage hypertrophy, and Wnt/ß-catenin pathway genes, required ADAMTS-5 activity. Cell death pathway genes were dysregulated, and Tp53, Foxo4, and Xbp1 endoplasmic reticulum-stress transcriptional networks were activated. Analysis of degradome genes identified up-regulation of many proteases, including Mmp3, Capn2, and the novel cartilage proteases Prss46 and Klk8. Comparison with other studies identified 16 genes also dysregulated in rat and human OA as priorities for study. CONCLUSION: We have identified, for the first time, several genes that have an ADAMTS-5-independent role in OA, identifying them as possible OA initiation candidates. This work provides new insights into the sequence of gene dysregulation and the molecular basis of cartilage destruction in OA.


Subject(s)
ADAM Proteins/deficiency , Cartilage, Articular/pathology , Osteoarthritis/genetics , Osteoarthritis/pathology , Transcriptome , ADAM Proteins/genetics , ADAMTS5 Protein , Animals , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL
14.
Biol Open ; 1(4): 318-28, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-23213422

ABSTRACT

The abundant proteoglycan, aggrecan, is resorbed from growth plate cartilage during endochondral bone ossification, yet mice with genetically-ablated aggrecan-degrading activity have no defects in bone formation. To account for this apparent anomaly, we propose that lysosomal hydrolases degrade extracellular, hyaluronan-bound aggrecan aggregates in growth plate cartilage, and that lysosomal hydrolases are released from hypertrophic chondrocytes into growth plate cartilage via Ca(2+)-dependent lysosomal exocytosis. In this study we confirm that hypertrophic chondrocytes release hydrolases via lysosomal exocytosis in vitro and we show in vivo evidence for lysosomal exocytosis in hypertrophic chondrocytes during skeletal development. We show that lysosome-associated membrane protein 1 (LAMP1) is detected at the cell surface following in vitro treatment of epiphyseal chondrocytes with the calcium ionophore, ionomycin. Furthermore, we show that in addition to the lysosomal exocytosis markers, cathepsin D and ß-hexosaminidase, ionomycin induces release of aggrecan- and hyaluronan-degrading activity from cultured epiphyseal chondrocytes. We identify VAMP-8 and VAMP7 as v-SNARE proteins with potential roles in lysosomal exocytosis in hypertrophic chondrocytes, based on their colocalisation with LAMP1 at the cell surface in secondary ossification centers in mouse tibiae. We propose that resorbing growth plate cartilage involves release of destructive hydrolases from hypertrophic chondrocytes, via lysosomal exocytosis.

15.
Arthritis Rheum ; 64(12): 4151-61; author reply 4162-3, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22886575

ABSTRACT

OBJECTIVE: To investigate aggrecan degradation in juvenile idiopathic arthritis (JIA). METHODS: The pattern and abundance of aggrecan fragments in synovial fluid (SF) aspirates from JIA patients were analyzed and compared with aggrecan fragments in SF from patients with other arthritides, children with knee injury, and a knee-healthy reference group. Concentrations of sulfated glycosaminoglycan (sGAG) in SF were measured by Alcian blue precipitation assay. Aggrecan fragments were purified by dissociative CsCl density-gradient centrifugation, deglycosylated, and analyzed by Western blot using antibodies specific for either aggrecanase-derived ARGS, SELE, and KEEE neoepitopes or the aggrecan G3 domain. RESULTS: The concentration of sGAG in SF from patients with JIA was significantly lower compared with that in SF from patients with osteoarthritis (OA) (P < 0.001), patients with juvenile knee injury (P = 0.006), and knee-healthy controls (P = 0.022). Western blot analysis revealed KEEE, SELE, and G3 fragments generated by aggrecanase cleavage in the chondroitin sulfate-rich region of aggrecan in patients with JIA. The pattern of aggrecan fragments in JIA patients was not identical to that in pooled OA SF, although there were notable similarities. Surprisingly, aggrecanase-derived ARGS fragments were barely detectable in JIA SF, in marked contrast to levels in OA SF. CONCLUSION: Aggrecanases appear to cleave minimally in the interglobular domain of aggrecan in JIA patients despite robust levels of cleavage in the chondroitin sulfate-rich region. These results suggest that in JIA, unlike other arthritides, aggrecanase cleavage in the aggrecan interglobular domain might not be a major pathogenic event.


Subject(s)
Aggrecans/metabolism , Arthritis, Juvenile/metabolism , Endopeptidases/metabolism , Peptide Fragments/metabolism , Synovial Fluid/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Glycosaminoglycans/metabolism , Humans , Infant , Knee Injuries/metabolism , Male , Middle Aged , Osteoarthritis, Knee/metabolism , Protein Structure, Tertiary , Sulfates/metabolism , Young Adult
16.
Biochim Biophys Acta ; 1812(12): 1616-29, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21914474

ABSTRACT

Proteoglycans are key components of extracellular matrices, providing structural support as well as influencing cellular behaviour in physiological and pathological processes. The diversity of proteoglycan function reported in the literature is equally matched by diversity in proteoglycan structure. Members of the ADAMTS (A Disintegrin And Metalloproteinase with ThromboSpondin motifs) family of enzymes degrade proteoglycans and thereby have the potential to alter tissue architecture and regulate cellular function. In this review, we focus on ADAMTS enzymes that degrade the lectican and small leucine-rich repeat families of proteoglycans. We discuss the known ADAMTS cleavage sites and the consequences of cleavage at these sites. We illustrate our discussion with examples from the literature in which ADAMTS proteolysis of proteoglycans makes profound changes to tissue function.


Subject(s)
ADAM Proteins/metabolism , Proteoglycans/metabolism , ADAM Proteins/chemistry , Animals , Brevican/metabolism , Glioma/blood supply , Glioma/metabolism , Humans , Morphogenesis , Neovascularization, Pathologic/metabolism , Organ Specificity , Ovulation/metabolism , Protein Structure, Tertiary , Proteolysis , Vascular Diseases/metabolism , Versicans/metabolism
17.
Nat Protoc ; 6(3): 388-404, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21372818

ABSTRACT

Proteolysis of the cartilage proteoglycan aggrecan is a feature of arthritis. We present a method for analyzing aggrecanolysis in in vitro cultures of 3-week-old mouse femoral head cartilage based on traditional methods developed for large animal species. Investigators can choose either a simple analysis that detects several aggrecan fragments released into culture medium only or a more comprehensive study that detects all fragments present in both the medium and the cartilage matrix. The protocol comprises (i) cartilage culture and optional cartilage extraction, (ii) a quick and simple colorimetric assay for quantitating aggrecan and (iii) neoepitope western blotting to identify specific aggrecan fragments partitioning to the medium or cartilage compartments. The crucial difference between the methods for mice and larger animals is that the proportion of aggrecan in a given sample is normalized to total aggrecan rather than to tissue wet weight. This necessary break from tradition arises because tiny volumes of liquid clinging to mouse cartilage can increase the apparent tissue wet weight, causing unacceptable errors. The protocol has broad application for the in vitro analysis of transgenic mice, particularly those with mutations that affect cartilage remodeling, arthritic disease and skeletal development. The protocol is robust, reliable and takes 7-11 d to complete.


Subject(s)
ADAM Proteins/metabolism , Aggrecans/analysis , Cartilage/enzymology , Aggrecans/metabolism , Animals , Arthritis/pathology , Blotting, Western , Cartilage/chemistry , Methylene Blue/analogs & derivatives , Mice , Mice, Inbred C57BL , Mice, Transgenic
18.
Best Pract Res Clin Rheumatol ; 25(6): 751-66, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22265258

ABSTRACT

Articular cartilage is a uniquely ordered tissue that is designed to resist compression and redistribute load, but is poorly equipped for self-repair. The chondrocyte is the only resident cell type, responsible for maintaining a specialised and extensive matrix that is avascular and lacks innervation. These attributes, as well as the slow turnover rate of aggrecan and type II collagen in mature articular cartilage, present a considerable challenge to the tissue engineer. Similarly, those attempting to halt the progression of cartilage erosion must contend with these unusual characteristics. This review explores the gaps in our knowledge of cartilage biology and pathology, including what is known about the relative contribution of collagenases and aggrecanases to cartilage degradation, the need to regulate the chondrocytic phenotype and the putative role of chondrocyte hypertrophy in the pathogenesis of degenerative and rheumatic joint disease. Recent advances in cartilage tissue engineering are also reviewed.


Subject(s)
Cartilage, Articular , Chondrocytes , Cartilage, Articular/pathology , Cartilage, Articular/physiology , Chondrocytes/pathology , Chondrocytes/physiology , Collagenases/metabolism , Endopeptidases/metabolism , Humans , Osteoarthritis/pathology , Osteoarthritis/therapy , Rheumatic Diseases/pathology , Rheumatic Diseases/therapy , Tissue Engineering
19.
J Biol Chem ; 285(46): 36216-24, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20843796

ABSTRACT

Degradation of the cartilage proteoglycan aggrecan is one of the earliest events that occurs in association with osteoarthritis. Little is known concerning the fate of the residual N-terminal G1 domains of cleaved aggrecan; domains that remain bound to hyaluronan. In this study, 68-72-kDa bands representative of aggrecan G1 domains containing ITEGE(373) neoepitope were detected within a hyaluronidase-sensitive pool at the cell surface of bovine articular chondrocytes and within a hyaluronidase-insensitive, intracellular pool. To determine the mechanisms that contribute to this distribution, CD44 expression was knocked down by siRNA or function by CD44-DN. Both approaches prevented the retention and internalization of G1-ITEGE. Inhibition of CD44 transit into lipid rafts blocked the endocytosis of G1-ITEGE but not the retention at the cell surface. Chondrocytes derived from CD44 null mice also exhibited limited potential for retention and internalization of G1-VTEGE. The consequence of a lack of chondrocyte-mediated endocytosis of these domains in cartilage of the CD44 null mice was the accumulation of the degradation fragments within the tissue. Additionally, chondrocytes or fibroblasts derived from CD44 null mice exhibited little capacity for retention and internalization of exogenous G1-ITEGE derived from bovine cartilage explants. Bovine or wild type mouse fibroblasts were able to bind and internalize bovine-derived G1-ITEGE. Although several pathways are available for the clearance of these domains, CD44-mediated cellular internalization is the most prominent.


Subject(s)
Aggrecans/metabolism , Chondrocytes/metabolism , Epitopes/metabolism , Hyaluronan Receptors/metabolism , Aggrecans/genetics , Amino Acid Sequence , Animals , Binding Sites/genetics , Blotting, Western , Cartilage, Articular/cytology , Cattle , Cells, Cultured , Cholesterol/metabolism , Chondrocytes/cytology , Endocytosis/drug effects , Epitopes/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Hyaluronan Receptors/genetics , Interleukin-1beta/pharmacology , Lipoylation , Male , Membrane Microdomains/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Microscopy, Fluorescence , RNA Interference , Synovial Membrane/cytology
20.
Arthritis Rheum ; 62(11): 3365-73, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20662062

ABSTRACT

OBJECTIVE: To compare the regulation of aggrecanase messenger RNA (mRNA) and enzyme activity by proinflammatory cytokines in primary mouse chondrocytes. METHODS: Primary chondrocytes were isolated from knee epiphyses of 6-8-day-old mice and cultured as monolayers. The cells were incubated with tumor necrosis factor α (TNFα), oncostatin M (OSM), or interleukin-6 (IL-6)/soluble IL-6 receptor, and mRNA levels were measured by quantitative polymerase chain reaction at various time points. To measure aggrecanase activity, the cells were incubated with cytokine in the presence of exogenous aggrecan, and substrate cleavage was measured using antibodies to neoepitopes. RESULTS: Expression of both ADAMTS-4 and ADAMTS-5 mRNA was up-regulated by TNFα and OSM. ADAMTS-5 mRNA expression was also up-regulated by IL-6. Treatment of wild-type mouse chondrocytes with each of the 3 cytokines increased cleavage of aggrecan at Glu(373)↓(374) Ala and Glu(1670)↓(1671) Gly; in chondrocytes lacking ADAMTS-5 activity, there was negligible cleavage at either site despite increased expression of ADAMTS-4 mRNA in the presence of TNFα or OSM. None of the cytokines substantially altered mRNA expression of ADAMTS-1 or ADAMTS-9. CONCLUSION: Despite substantial increases in the expression of ADAMTS-4 mRNA induced by TNFα and OSM, these cytokines induced little if any increase in aggrecanolysis in ADAMTS-5-deficient mouse chondrocytes. Our data show a poor correlation between the level of cytokine-induced ADAMTS-4 mRNA expression and the level of aggrecan-degrading activity in cultured chondrocytes.


Subject(s)
ADAM Proteins/metabolism , Chondrocytes/enzymology , Cytokines/pharmacology , Endopeptidases/metabolism , ADAM Proteins/genetics , Animals , Chondrocytes/drug effects , Endopeptidases/genetics , Mice , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...