Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Osteoarthritis Cartilage ; 18(2): 249-56, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19800448

ABSTRACT

OBJECTIVE: Our goal was to test the hypothesis that specific integrin receptors regulate chondrocyte biosynthetic response to dynamic compression at early times in 3D gel culture, during initial evolution of the pericellular matrix, but prior to significant accumulation of further-removed matrix. The study was motivated by increased use of dynamic loading, in vitro, for early stimulation of tissue engineered cartilage, and the need to understand the effects of loading, in vivo, at early times after implantation of constructs. METHODS: Bovine articular chondrocytes were seeded in 2% agarose gels (15x10(6)cells/mL) and incubated for 18 h with and without the presence of specific integrin blockers (small-molecule peptidomimetics, function-blocking antibodies, and RGD-containing disintegrins). Samples were then subjected to a 24-h dynamic compression regime found previously to stimulate chondrocyte biosynthesis in 3D gel as well as cartilage explant culture (1 Hz, 2.5% dynamic strain amplitude, 7% static offset strain). At the end of loading, proteoglycan (PG) synthesis ((35)S-sulfate incorporation), protein synthesis ((3)H-proline incorporation), DNA content (Hoechst dye 33258) and total glycosaminoglycan (GAG) content (dimethyl methylene blue (DMMB) dye binding) were assessed. RESULTS: Consistent with previous studies, dynamic compression increased PG synthesis and total GAG accumulation compared to free-swelling controls. Blocking alphavbeta3 abolished this response, independent of effects on controls, while blocking beta1 abolished the relative changes in synthesis when changes in free-swelling synthesis rates were observed. CONCLUSIONS: This study suggests that both alphavbeta3 and beta1 play a role in pathways that regulate stimulation of PG synthesis and accumulation by dynamic compression, but through distinct complementary mechanisms.


Subject(s)
Cartilage, Articular/physiology , Glycosaminoglycans/biosynthesis , Integrins/antagonists & inhibitors , Proteoglycans/biosynthesis , Animals , Cartilage, Articular/cytology , Cattle , Cells, Cultured , Chondrocytes , Compressive Strength/physiology , Culture Techniques/methods , Sepharose/chemistry , Stress, Mechanical
2.
Osteoarthritis Cartilage ; 17(5): 627-35, 2009 May.
Article in English | MEDLINE | ID: mdl-19026578

ABSTRACT

OBJECTIVE: To develop an in vivo model for rapid assessment of cartilage aggrecan degradation and its pharmacological modulation. DESIGN: Tumor necrosis factor-alpha (TNFalpha) was injected intra-articularly (IA) in rat knees and aggrecan degradation was monitored at various times following challenge. Articular cartilage was assessed for aggrecan content by Safranin O staining and by immunohistochemistry for the NITEGE epitope. Synovial fluids (SFs) were analyzed for sulfated glycosaminoglycans (GAGs) using the dimethylmethylene blue dye assay and for aggrecan fragments generated by specific cleavage at aggrecanase-sensitive sites by Western blot analysis with neoepitope antibodies. Indomethacin, dexamethasone, and an aggrecanase inhibitor were evaluated for their ability to modulate TNFalpha-induced proteoglycan degradation in vivo. RESULTS: (1) IA injection of TNFalpha in the knee joint of rats resulted in transient aggrecan degradation and release of aggrecanase-generated aggrecan fragments from the articular cartilage into the SF; (2) a correlation was observed between histologically assessed depletion of aggrecan from the articular cartilage and the appearance of specific neoepitopes in the SF; (3) aggrecan degradation was inhibited by an aggrecanase inhibitor as well as by dexamethasone, but not by the non-steroidal anti-inflammatory drug (NSAID), indomethacin. CONCLUSION: TNFalpha injection in the knee joints of rats results in rapid transient cartilage proteoglycan degradation, mediated by cleavage at the aggrecanase sites. Biomarker read-out of specific neoepitopes in the SF enables the use of this mechanism-based model for rapid evaluation of aggrecanase-mediated aggrecan degradation in vivo.


Subject(s)
Aggrecans/metabolism , Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Knee Joint/pathology , Osteoarthritis/pathology , Proteoglycans/metabolism , Tumor Necrosis Factor-alpha/metabolism , Aggrecans/pharmacology , Animals , Arthritis, Experimental/drug therapy , Blotting, Western , Cartilage, Articular/drug effects , Immunohistochemistry , Injections, Intra-Articular , Knee Joint/drug effects , Male , Osteoarthritis/drug therapy , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/pharmacology
3.
J Med Chem ; 44(21): 3347-50, 2001 Oct 11.
Article in English | MEDLINE | ID: mdl-11585439

ABSTRACT

A pharmacophore model of the P1' site, specific for aggrecanase, was defined using the specificity studies of the matrix metalloproteinases and the similar biological activity of aggrecanase and MMP-8. Incorporation of the side chain of a tyrosine residue into compound 1 as the P1' group provided modest selectivity for aggrecanase over MMP-1, -2, and -9. A cis-(1S)(2R)-amino-2-indanol scaffold was incorporated as a tyrosine mimic (P2') to conformationally constrain 2. Further optimization resulted in compound 11, a potent, selective, and orally bioavailable inhibitor of aggrecanase.


Subject(s)
Asparagine/chemical synthesis , Endopeptidases/metabolism , Hydroxamic Acids/chemical synthesis , Protease Inhibitors/chemical synthesis , Administration, Oral , Animals , Asparagine/analogs & derivatives , Asparagine/chemistry , Asparagine/pharmacokinetics , Asparagine/pharmacology , Biological Availability , Dogs , Drug Design , Endopeptidases/chemistry , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/pharmacology , Matrix Metalloproteinase 1/chemistry , Matrix Metalloproteinase 2/chemistry , Matrix Metalloproteinase 8/chemistry , Matrix Metalloproteinase 9/chemistry , Models, Molecular , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/pharmacology , Protein Binding , Stereoisomerism , Structure-Activity Relationship
4.
Arthritis Rheum ; 44(6): 1401-10, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11407701

ABSTRACT

OBJECTIVE: To assess the role of matrix metalloproteinases (MMPs) in cartilage and bone erosions in Lyme arthritis METHODS: We examined synovial fluid from 10 patients with Lyme arthritis for the presence of MMP-2, MMP-3, MMP-9, and "aggrecanase" activity using gelatinolytic zymography and immunoblot analysis. We developed an in vitro model of Lyme arthritis using cartilage explants and observed changes in cartilage degradation in the presence of Borrelia burgdorferi and/or various protease inhibitors. RESULTS: Synovial fluid from patients with Lyme arthritis was found to contain at least 3 MMPs: gelatinase A (MMP-2), stromelysin (MMP-3), and gelatinase B (MMP-9). In addition, there was evidence in 2 patients of "aggrecanase" activity not accounted for by the above enzymes. Infection of cartilage explants with B. burgdorferi resulted in induction of MMP-3, MMP-9, and "aggrecanase" activity. Increased induction of these enzymes by B. burgdorferi alone was not sufficient to cause cartilage destruction in the explants as measured by glycosaminoglycan (GAG) and hydroxyproline release. However, addition of plasminogen, which can act as an MMP activator, to cultures resulted in significant GAG and hydroxyproline release in the presence of B. burgdorferi. The MMP inhibitor batimastat significantly reduced the GAG release and completely inhibited the collagen degradation. CONCLUSION: MMPs are found in synovial fluids from patients with Lyme arthritis and are induced from cartilage tissue by the presence of B. burgdorferi. Inhibition of MMP activity prevents B. burgdorferi-induced cartilage degradation in vitro.


Subject(s)
Arthritis, Infectious/enzymology , Lyme Disease/enzymology , Matrix Metalloproteinases/metabolism , Animals , Arthritis, Infectious/etiology , Blotting, Western , Borrelia burgdorferi Group/genetics , Borrelia burgdorferi Group/isolation & purification , Cartilage/chemistry , Cartilage/cytology , Cattle , Culture Techniques , DNA, Bacterial/analysis , Endopeptidases/metabolism , Enzyme-Linked Immunosorbent Assay , Glycosaminoglycans/analysis , Humans , Knee Joint/enzymology , Knee Joint/microbiology , Lyme Disease/complications , Polymerase Chain Reaction , Synovial Fluid/enzymology , Synovial Fluid/microbiology
5.
J Biol Chem ; 275(50): 39096-102, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-10991945

ABSTRACT

Aggrecan degradation involves proteolytic cleavage of the core protein within the interglobular domain. Because aggrecan is highly glycosylated with chondroitin sulfate (CS) and keratan sulfate (KS), we investigated whether glycosylation affects digestion by aggrecanase at the Glu(373)-Ala(374) bond. Treatment of bovine aggrecan monomers to remove CS and KS resulted in loss of cleavage at this site, suggesting that glycosaminoglycans (GAGs) play a role in cleavage at the Glu(373)-Ala(374) bond. In contrast, MMP-3 cleavage at the Ser(341)-Phe(342) bond was not affected by glycosidase treatment of aggrecan. Removal of KS, but not CS, prevented cleavage at the Glu(373)-Ala(374) bond. Thus, KS residues may be important for recognition of this cleavage site by aggrecanase. KS glycosylation has been observed at sites adjacent to the Glu(373)-Ala(374) bond in steer aggrecan, but not in calf aggrecan (Barry, F. P., Rosenberg, L. C., Gaw, J. U., Gaw, J. U., Koob, T. J., and Neame, P. J. (1995) J. Biol. Chem. 270, 20516-20524). Interestingly, although we found that aggrecanase degraded both calf and steer cartilage aggrecan, the proportion of fragments generated by cleavage at the Glu(373)-Ala(374) bond was higher in steer than in calf, consistent with our observations using aggrecan treated to remove KS. We conclude that the GAG content of aggrecan influences the specificity of aggrecanase for cleavage at the Glu(373)-Ala(374) bond and suggest that age may be a factor in aggrecanase degradation of cartilage.


Subject(s)
Aging , Endopeptidases/metabolism , Extracellular Matrix Proteins , Glycoside Hydrolases , Proteoglycans/metabolism , Age Factors , Aggrecans , Alanine/chemistry , Animals , Blotting, Western , Cattle , Chondroitin Sulfates/metabolism , Chondroitinases and Chondroitin Lyases/pharmacology , Freezing , Glutamine/chemistry , Glycosaminoglycans/metabolism , Glycosylation , Keratan Sulfate/metabolism , Lectins, C-Type , Matrix Metalloproteinase 3/metabolism , Phenylalanine/chemistry , Serine/chemistry , Time Factors , beta-Galactosidase/pharmacology
6.
J Biol Chem ; 275(30): 22695-703, 2000 Jul 28.
Article in English | MEDLINE | ID: mdl-10801887

ABSTRACT

Brain-enriched hyaluronan binding (BEHAB)/brevican is a brain-specific extracellular matrix protein containing a cleavage site between Glu(395)-Ser(396), which bears remarkable homology to the "aggrecanase" site in the cartilage proteoglycan aggrecan. Expression of BEHAB/brevican is dramatically increased in human gliomas, notoriously invasive tumors. Recently, we showed that the rat 9L gliosarcoma cell line, which does not express BEHAB/brevican and forms non-invasive tumors when grown as intracranial grafts, can form invasive tumors when transfected with a 5' cDNA fragment of BEHAB/brevican, but not when transfected with the full-length cDNA. In marked contrast, the highly invasive CNS-1 glioma cell line expresses and cleaves BEHAB/brevican protein when grown as an intracranial graft. These results suggest that both synthesis and cleavage of BEHAB/brevican protein may play a role in the invasiveness of gliomas. We report here, using an antibody developed to the neoepitope created by BEHAB/brevican cleavage at the Glu(395)-Ser(396) site, that the CNS-1 cells are able to cleave the protein in vitro. We characterized the CNS-1-derived cleavage activity by assaying its ability to cleave BEHAB/brevican proteoglycan, and determined that the enzyme is a constitutively expressed, secreted activity. Using a variety of protease inhibitors, reverse transcriptase-polymerase chain reaction, and specific antibodies, we determined that this activity is likely to be a member of the ADAMTS family of metalloproteinases, specifically ADAMTS4. These results suggest a novel function for ADAMTS family members in BEHAB/brevican cleavage and glioma and indicate that inhibition of ADAMTS in glioma may provide a novel therapeutic strategy.


Subject(s)
ADAM Proteins/metabolism , Brain/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Disintegrins/metabolism , Hyaluronic Acid/metabolism , Metalloendopeptidases/metabolism , Nerve Tissue Proteins/metabolism , Thrombospondins/chemistry , ADAM Proteins/antagonists & inhibitors , ADAMTS4 Protein , Animals , Base Sequence , Brevican , DNA Primers , Glioma/metabolism , Glioma/pathology , Humans , Hydrolysis , Lectins, C-Type , Metalloendopeptidases/antagonists & inhibitors , Precipitin Tests , Protease Inhibitors/pharmacology , Rats , Transfection , Tumor Cells, Cultured
7.
Arch Biochem Biophys ; 383(2): 256-64, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11185561

ABSTRACT

The goal of this study was to examine the effects of mechanical compression on chondrocyte biosynthesis of extracellular matrix (ECM) components during culture in a new alginate disk culture system. Specifically, we have examined chondrocyte biosynthesis rates, and the structure of aggrecan core protein species present in the cell-associated matrix (CM), in the further removed matrix (FRM) and in the surrounding culture medium. In this alginate disk culture system, chondrocytes can be subjected to mechanical deformations similar to those experienced in vivo. Our results show that over an 8-week culture period, chondrocytes synthesize a functional ECM and can respond to mechanical forces similarly to chondrocytes maintained in native cartilage. In the alginate disk system, static compression was shown to decrease and dynamic compression to increase synthesis of aggrecan of bovine chondrocytes. Western blot analysis of the core proteins of aggrecan molecules identified a number of different species that were present in different relative amounts in the CM, FRM, and medium. Over 21 days of culture, the predominant form of aggrecan found in the ECM was a full-length link-stabilized species. In addition, our data show that the application of 40 h of static compression caused an increase in the proportion of newly synthesized aggrecan molecules released into the medium. However, this was not accompanied by a significant change in the size and composition of aggrecan and aggrecan fragments in the different compartments, suggesting that mechanical compression did not alter the catabolic pathways. Together, these data show that chondrocyte function is maintained in an alginate disk culture system and that this culture system is a useful model to examine chondrocyte ECM assembly and some aspects of catabolism normally found in vivo.


Subject(s)
Alginates/chemistry , Alginates/metabolism , Cell Culture Techniques/methods , Chondrocytes/metabolism , Extracellular Matrix Proteins , Extracellular Matrix/metabolism , Aggrecans , Animals , Blotting, Western , Cartilage/chemistry , Cartilage/metabolism , Cattle , Chondrocytes/chemistry , Epitopes , Extracellular Matrix/chemistry , Lectins, C-Type , Proteoglycans/chemistry , Proteoglycans/metabolism , Time Factors
8.
J Biol Chem ; 274(45): 32387-95, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10542281

ABSTRACT

A recombinant human aggrecan G1-G2 fragment comprising amino acids Val(1)-Arg(656) has been expressed in Sf21 cells using a baculovirus expression system. The recombinant G1-G2 (rG1-G2) was purified to homogeneity by hyaluronan-Sepharose affinity chromatography followed by high performance liquid chromatography gel filtration, and gave a single band of M(r) 90,000-95,000 by silver stain or immunoblotting with monoclonal antibody 1-C-6. The expressed G1-G2 bound to both hyaluronan and link protein indicating that the immunoglobulin-fold motif and proteoglycan tandem repeat loops of the G1 domain were correctly folded. Further analysis of secondary structure by rotary shadowing electron microscopy confirmed a double globe appearance, but revealed that the rG1-G2 was more compact than its native counterpart. The size of rG1-G2 by SDS-polyacrylamide gel electorphoresis was unchanged following digestion with keratanase and keratanase II and reduced by only 2-5 kDa following digestion with either O-glycosidase or N-glycosidase F. Recombinant G1-G2 was digested with purified matrix metalloproteinases (MMP), isolated aggrecanase, purified atrolysin C, or proteinases present in conditioned medium from cartilage explant cultures, and the products analyzed on SDS gels by silver stain and immunoblotting. Neoepitope antibodies recognizing the N-terminal F(342)FGVG or C-terminal DIPEN(341) sequences were used to confirm MMP cleavage at the Asn(341) downward arrow Phe bond, while neoepitope antibodies recognizing the N-terminal A(374)RGSV or C-terminal ITEGE(373) sequences were used to confirm aggrecanase cleavage at the Glu(373) downward arrow Ala bond. Cleavage at the authentic MMP and aggrecanase sites revealed that these proteinases have the same specificity for rG1-G2 as for native aggrecan. Incubation of rG1-G2 with conditioned medium from porcine cartilage cultures revealed that active soluble aggrecanase but no active MMPs, was released following stimulation with interleukin-1alpha or retinoic acid. Atrolysin C, which cleaves native bovine aggrecan at both the aggrecanase and MMP sites, efficiently cleaved rG1-G2 at the aggrecanase site but failed to cleave at the MMP site. In contrast, native glycosylated G1-G2 with or without keratanase treatment was cleaved by atrolysin C at both the aggrecanase and MMP sites. The results suggest that the presence or absence per se of keratan sulfate on native G1-G2 does not affect the activity of atrolysin C toward the two sites.


Subject(s)
Endopeptidases/metabolism , Extracellular Matrix Proteins , Matrix Metalloproteinases/metabolism , Proteoglycans/metabolism , Aggrecans , Alanine/metabolism , Animals , Asparagine/metabolism , Cattle , Cloning, Molecular , Glutamine/metabolism , Humans , Lectins, C-Type , Metalloendopeptidases/metabolism , Phenylalanine/metabolism , Protein Conformation , Proteoglycans/genetics , Recombinant Proteins/metabolism , Substrate Specificity
9.
Ann N Y Acad Sci ; 878: 92-107, 1999 Jun 30.
Article in English | MEDLINE | ID: mdl-10415722

ABSTRACT

In arthritic diseases there is a gradual erosion of cartilage that leads to a loss of joint function. Aggrecan, which provides cartilage with its properties of compressibility and elasticity, is the first matrix component to undergo measurable loss in arthritis. This loss of aggrecan appears to be due to an increased rate of degradation, that can be attributed to proteolytic cleavage of the core protein within the interglobular domain (IGD). Two major sites of cleavage have been identified within the IGD. One, between the amino acids Asn341-Phe342, where the matrix metalloproteinases (MMPs) have been shown to clip; and the other, between Glu373-Ala374, which is attributed to a novel protease, "aggrecanase." We have generated aggrecanase in conditioned media from IL-1-stimulated bovine nasal cartilage and have used an enzymatic assay to evaluate this proteinase activity. In these studies we follow the generation of aggrecanase and MMPs in response to IL-1 in this system and examine the contribution of these enzymes in aggrecan degredation. Our data suggest that aggrecanase is a key enzyme in cartilage aggrecan degradation that represents a novel target for cartilage protection therapy in arthritis.


Subject(s)
Cartilage/enzymology , Endopeptidases/metabolism , Matrix Metalloproteinase 3/metabolism , Animals , Cartilage/drug effects , Cattle , Endopeptidases/genetics , Interleukin-1/pharmacology , Kinetics , Matrix Metalloproteinase 3/genetics , Metalloendopeptidases/metabolism , Nasal Septum , Organ Culture Techniques , Time Factors
10.
Science ; 284(5420): 1664-6, 1999 Jun 04.
Article in English | MEDLINE | ID: mdl-10356395

ABSTRACT

We purified, cloned, and expressed aggrecanase, a protease that is thought to be responsible for the degradation of cartilage aggrecan in arthritic diseases. Aggrecanase-1 [a disintegrin and metalloproteinase with thrombospondin motifs-4 (ADAMTS-4)] is a member of the ADAMTS protein family that cleaves aggrecan at the glutamic acid-373-alanine-374 bond. The identification of this protease provides a specific target for the development of therapeutics to prevent cartilage degradation in arthritis.


Subject(s)
Extracellular Matrix Proteins , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , ADAM Proteins , ADAMTS1 Protein , ADAMTS4 Protein , Aggrecans , Amino Acid Sequence , Arthritis/drug therapy , Cartilage/metabolism , Catalytic Domain , Cloning, Molecular , Disintegrins/chemistry , Disintegrins/metabolism , Humans , Hydroxamic Acids/pharmacology , Interleukin-1/pharmacology , Lectins, C-Type , Metalloendopeptidases/isolation & purification , Metalloendopeptidases/metabolism , Molecular Sequence Data , Procollagen N-Endopeptidase , Protease Inhibitors/pharmacology , Protein Sorting Signals , Proteoglycans/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Analysis
11.
Bioorg Med Chem Lett ; 9(10): 1453-8, 1999 May 17.
Article in English | MEDLINE | ID: mdl-10360755

ABSTRACT

A novel series of 13- and 14-membered macrocyclic amines was developed by linking the P1 and P2' groups. The synthesis entails stereoselective Frater alkylation to install the anti-succinate configuration and macrocyclic amination via nucleophilic displacement. This strategy resulted in a new class of conformationally constrained inhibitors that are potent and selective for MMP-8 and 9 over MMP-1 and 3.


Subject(s)
Amines/pharmacology , Extracellular Matrix/enzymology , Metalloendopeptidases/antagonists & inhibitors , Protease Inhibitors/pharmacology , Amines/chemistry , Computer Simulation , Models, Molecular , Protease Inhibitors/chemistry
12.
Bioorg Med Chem Lett ; 9(9): 1279-84, 1999 May 03.
Article in English | MEDLINE | ID: mdl-10340614

ABSTRACT

Several macrocyclic, hydroxamate derivatives were synthesized and evaluated as inhibitors of matrix metalloproteinases (MMPs) and tumour necrosis factor-alpha (TNF-alpha) production. These macrocycles are anti-succinate based inhibitors linked from P1 to P2'. A variety of functionality was installed at the P1-P2' linkage, which gave inhibitors that displayed excellent MMP inhibition and good TNF-alpha suppression.


Subject(s)
Hydroxamic Acids/chemistry , Metalloendopeptidases/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Crystallography, X-Ray , Humans , Inhibitory Concentration 50 , Kinetics , Lipopolysaccharides/metabolism , Matrix Metalloproteinase 1 , Matrix Metalloproteinase 9 , Matrix Metalloproteinase Inhibitors , Metalloendopeptidases/classification , Models, Chemical , Models, Molecular
13.
J Biol Chem ; 274(10): 6594-601, 1999 Mar 05.
Article in English | MEDLINE | ID: mdl-10037754

ABSTRACT

A method was developed for generating soluble, active "aggrecanase" in conditioned media from interleukin-1-stimulated bovine nasal cartilage cultures. Using bovine nasal cartilage conditioned media as a source of the aggrecanase enzyme, an enzymatic assay was established employing purified aggrecan monomers as a substrate and monitoring specific aggrecanase-mediated cleavage products by Western analysis using the monoclonal antibody, BC-3 (which recognizes the new N terminus, ARGS, on fragments produced by cleavage between amino acid residues Glu373 and Ala374). Using this assay we have characterized cartilage aggrecanase with respect to assay kinetics, pH and salt optima, heat sensitivity, and stability upon storage. Aggrecanase activity was inhibited by the metalloprotease inhibitor, EDTA, while a panel of inhibitors of serine, cysteine, and aspartic proteinases had no effect, suggesting that aggrecanase is a metalloproteinase. Sensitivity to known matrix metalloproteinase inhibitors as well as to the endogenous tissue inhibitor of metalloproteinases, TIMP-1, further support the notion that aggrecanase is a metalloproteinase potentially related to the ADAM family or MMP family of proteases previously implicated in the catabolism of the extracellular matrix.


Subject(s)
Cartilage/enzymology , Endopeptidases/isolation & purification , Animals , Cattle , Cell Culture Techniques/methods , Culture Media, Conditioned , Endopeptidases/metabolism , Enzyme Activation , Interleukin-1/pharmacology , Substrate Specificity
14.
Osteoarthritis Cartilage ; 6(3): 214-28, 1998 May.
Article in English | MEDLINE | ID: mdl-9682788

ABSTRACT

OBJECTIVE: To evaluate the relationship between specific cleavage of aggrecan at the Glu373-Ala374 'aggrecanase' site and degradation and release of proteoglycan catabolites from cartilage in explant cultures. DESIGN: The monoclonal antibody, BC-3, which specifically recognizes the new N-terminus, ARGSVIL, generated by cleavage of aggrecan at the Glu373-Ala374 'aggrecanase' site, was used to follow the generation of fragments produced by cleavage at this site as compared to degradation of proteoglycan as assessed by glycosaminoglycan (GAG) release from cartilage in response to cytokines and the ability of inhibitors to block this cleavage. RESULTS: (1) There was a strong correlation between specific cleavage at the Glu373-Ala374 bond and the release of aggrecan catabolites in response to interleukin-1 (IL-1) or tumour necrosis factor (TNF) stimulation. (2) This cleavage in the interglobular domain of aggrecan was inhibited by the inclusion of cycloheximide, thus indicating a requirement for de novo protein synthesis in the induction of 'aggrecanase' activity. (3) The inhibitors, indomethacin, naproxen, tenidap, dexamethasone and doxycycline were ineffective in blocking either specific cleavage at the 'aggrecanase' site or aggrecan degradation as measured by GAG release from cartilage. (4) In contrast, compounds which act through two different mechanisms to inhibit MMPs were effective in blocking both specific cleavage at the 'aggrecanase' site and proteoglycan degradation. CONCLUSIONS: Our data suggest that 'aggrecanase' is primarily responsible for proteoglycan cleavage in these experimental systems and that this protease has properties in common with metalloproteases including members of the MMP and ADAM family. Inhibition of 'aggrecanase' may have utility in preventing cartilage loss in arthritis.


Subject(s)
Cartilage/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Extracellular Matrix Proteins , Interleukin-1/pharmacology , Proteoglycans/drug effects , Proteoglycans/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Aggrecans , Animals , Blotting, Northern , Cartilage/drug effects , Cattle , Cycloheximide/pharmacology , Glycosaminoglycans/metabolism , Glycosylation/drug effects , Lectins, C-Type , Organ Culture Techniques , Time Factors
16.
J Biol Chem ; 273(10): 5846-50, 1998 Mar 06.
Article in English | MEDLINE | ID: mdl-9488721

ABSTRACT

Two primary cleavage sites have been identified within the interglobular domain of the cartilage aggrecan core protein: one is between amino acid residues Asn 341 and Phe342, where many matrix metalloproteinases (MMP) have been shown to cleave; and the other is between amino acid residues Glu373 and Ala374. Although cleavage at the Glu373-Ala374 site is believed to play a critical role in cartilage aggrecan degradation in arthritic diseases, the enzyme responsible for cleavage at this site, "aggrecanase," has not been identified. Members of the ADAM (a disintegrin and metalloproteinase) family of proteins, which shows structural homology to the snake venom hemorrhagic metalloproteinases (reprolysins), have recently been demonstrated to be expressed in articular chondrocytes. Because many ADAM family members have a putative proteinase function, this raises the possibility that aggrecanase may be a member of this family of proteases. To examine whether reprolysins have the ability to cleave aggrecan at either the aggrecanase site or the MMP site, the snake venom hemorrhagic toxin metalloproteinase HT-d (atrolysin C) was tested for its ability to cleave bovine aggrecan monomer. Cleavage was monitored using the BC-3 antibody, which recognizes aggrecan fragments with the new NH2 terminus ARGSV generated by cleavage at the aggrecanase site, and with the AF-28 antibody, which recognizes aggrecan fragments with the new NH2 terminus FFGVG generated by cleavage at the MMP site. Cleavage at both the aggrecanase and MMP sites occurred in a concentration-dependent manner with 100 nM atrolysin C or greater. AF-28-reactive fragments were generated by 30 min of incubation, and levels were maximal by 8 h; BC-3-reactive fragments were detected at 2 h and continued to increase through 48 h, thus suggesting that atrolysin C can cleave at the MMP and aggrecanase sites. NH2-terminal aggrecan fragments generated by cleavage at the aggrecanase site were also detected using antisera recognizing the new COOH terminus, NITEGE, formed by cleavage at the Glu373-Ala374 bond, indicating that cleavage at this site does not require prior cleavage at the MMP site. These data provide the first demonstration that a reprolysin can cleave the core protein of aggrecan and the first example of a specific protease that can cleave at the aggrecanase site independent of cleavage at the MMP cleavage site.


Subject(s)
Cartilage/chemistry , Endopeptidases/metabolism , Extracellular Matrix Proteins , Metalloendopeptidases/metabolism , Proteoglycans/metabolism , Aggrecans , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Cattle , Lectins, C-Type , Peptide Fragments/immunology , Peptide Fragments/metabolism , Substrate Specificity
17.
Inflamm Res ; 47(3): 115-21, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9562336

ABSTRACT

OBJECTIVE: To evaluate the effect of the antioxidant-like anti-inflammatory agent, ebselen, on cartilage proteoglycan degradation and to determine whether its cartilage protectant activity is related to its antioxidant activity. MATERIALS AND METHODS: Cartilage in organ culture was stimulated with interleukin-1 (IL-1), and proteoglycan degradation was assessed by measuring the amount of sulfated glycosaminoglycan released into the media, proteoglycan synthesis evaluated by [35S]-sulfate incorporation, and prostaglandin E2 (PGE2) release determined by radioimmunoassay (RIA). Glutathione peroxidase (GSH-Px) activity was evaluated in a coupled test system using NADPH/GSSG reductase as an indicator and cyclooxygenase activity was evaluated using sheep seminal vesicle prostaglandin synthase. RESULTS: Ebselen caused a concentration-dependent inhibition of IL-1-stimulated proteoglycan degradation with an IC50 of 4.7 microM. Cartilage PGE2 release was also reduced in the presence of ebselen (IC50 = 6.2 microM). However, at concentrations up to 100 microM, ebselen had no effect on the inhibition of proteoglycan synthesis by IL-1. Induction of proteoglycan breakdown was also inhibited by a sulfur analog of ebselen. This analog was devoid of GSH-Px activity and was 50-fold less potent in cyclooxygenase inhibitory activity, but was equipotent to ebselen in inhibiting cartilage degradation. CONCLUSIONS: Ebselen, unlike other NSAIDs, blocks cartilage proteoglycan breakdown without inhibiting proteoglycan synthesis. This effect is independent of its GSH-Px activity and its ability to inhibit cyclooxygenase and PGE2 production. Therefore, this compound may provide a new mechanism for protecting cartilage matrix from degradative factors in arthritic joints.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Azoles/pharmacology , Cartilage/drug effects , Cyclooxygenase Inhibitors/pharmacology , Inflammation/drug therapy , Interleukin-1/pharmacology , Organoselenium Compounds/pharmacology , Proteoglycans/metabolism , Animals , Arthritis/drug therapy , Cartilage/metabolism , Cattle , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Glutathione Disulfide/analysis , Glutathione Peroxidase/metabolism , Glycosaminoglycans/metabolism , Inflammation/chemically induced , Isoindoles , NADP/analysis , Organ Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...