Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Int J Mol Sci ; 20(3)2019 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-30700002

RESUMEN

BACKGROUND: Cartilage regeneration requires a balance of anabolic and catabolic processes. AIM: To examine the susceptibility of fibromodulin (FMOD) and lumican (LUM) to degradation by MMP-13, ADAMTS-4 and ADAMTS-5, the three major degradative proteinases in articular cartilage, in cartilage development and in osteoarthritis (OA). METHODS: Immunolocalization of FMOD and LUM in fetal foot and adult knee cartilages using an FMOD matrix metalloprotease (MMP)-13 neoepitope antibody (TsYG11) and C-terminal anti-FMOD (PR184) and anti-LUM (PR353) antibodies. The in vitro digestion of knee cartilage with MMP-13, A Disintegrin and Metalloprotease with Thrompospondin motifs (ADAMTS)-4 and ADAMTS-5, to assess whether FMOD and LUM fragments observed in Western blots of total knee replacement specimens could be generated. Normal ovine articular cartilage explants were cultured with interleukin (IL)-1 and Oncostatin-M (OSM) ± PGE3162689, a broad spectrum MMP inhibitor, to assess FMOD, LUM and collagen degradation. RESULTS AND DISCUSSION: FMOD and LUM were immunolocalized in metatarsal and phalangeal fetal rudiment cartilages and growth plates. Antibody TsYG11 localized MMP-13-cleaved FMOD in the hypertrophic chondrocytes of the metatarsal growth plates. FMOD was more prominently localized in the superficial cartilage of normal and fibrillated zones in OA cartilage. TsYG11-positive FMOD was located deep in the cartilage samples. Ab TsYG11 identified FMOD fragmentation in Western blots of normal and fibrillated cartilage extracts and total knee replacement cartilage. The C-terminal anti-FMOD, Ab PR-184, failed to identify FMOD fragmentation due to C-terminal processing. The C-terminal LUM, Ab PR-353, identified three LUM fragments in OA cartilages. In vitro digestion of human knee cartilage with MMP-13, ADAMTS-4 and ADAMTS-5 generated FMOD fragments of 54, 45 and 32 kDa similar to in blots of OA cartilage; LUM was less susceptible to fragmentation. Ab PR-353 detected N-terminally processed LUM fragments of 39, 38 and 22 kDa in 65⁻80-year-old OA knee replacement cartilage. FMOD and LUM were differentially processed in MMP-13, ADAMTS-4 and ADAMTS-5 digestions. FMOD was susceptible to degradation by MMP-13, ADAMTS-4 and to a lesser extent by ADAMTS-5; however, LUM was not. MMP-13-cleaved FMOD in metatarsal and phalangeal fetal rudiment and growth plate cartilages suggested roles in skeletogenesis and OA pathogenesis. Explant cultures of ovine cartilage stimulated with IL-1/OSM ± PGE3162689 displayed GAG loss on day 5 due to ADAMTS activity. However, by day 12, the activation of proMMPs occurred as well as the degradation of FMOD and collagen. These changes were inhibited by PGE3162689, partly explaining the FMOD fragments seen in OA and the potential therapeutic utility of PGE3162689.


Asunto(s)
Proteína ADAMTS4/metabolismo , Cartílago Articular/metabolismo , Cartílago Articular/patología , Fibromodulina/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Animales , Humanos , Lumican/metabolismo , Masculino , Persona de Mediana Edad , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/patología , Ovinos
2.
Proc Natl Acad Sci U S A ; 107(8): 3734-9, 2010 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-20133709

RESUMEN

Osteoarthritis (OA), the most common arthritic condition in humans, is characterized by the progressive degeneration of articular cartilage accompanied by chronic joint pain. Inflammatory mediators, such as cytokines and prostaglandin E(2) (PGE(2)) that are elevated in OA joints, play important roles in the progression of cartilage degradation and pain-associated nociceptor sensitivity. We have found that the nuclear receptor family transcription factors Liver X Receptors (LXRalpha and -beta) are expressed in cartilage, with LXRbeta being the predominant isoform. Here we show that genetic disruption of Lxrbeta gene expression in mice results in significantly increased proteoglycan (aggrecan) degradation and PGE(2) production in articular cartilage treated with IL-1beta, indicating a protective role of LXRbeta in cartilage. Using human cartilage explants, we found that activation of LXRs by the synthetic ligand GW3965 significantly reduced cytokine-induced degradation and loss of aggrecan from the tissue. Furthermore, LXR activation dramatically inhibited cytokine-induced PGE(2) production by human osteoarthritic cartilage as well as by a synovial sarcoma cell line. These effects were achieved at least partly by repression of the expression of ADAMTS4, a physiological cartilage aggrecanase, and of cyclooxygenase-2 and microsomal prostaglandin E synthase-1, key enzymes in the PGE(2) synthesis pathway. Consistent with our in vitro observations, oral administration of GW3965 potently alleviated joint pain in a rat meniscal tear model of osteoarthritis.


Asunto(s)
Cartílago Articular/metabolismo , Dinoprostona/antagonistas & inhibidores , Receptores Nucleares Huérfanos/agonistas , Osteoartritis/complicaciones , Dolor/metabolismo , Proteínas ADAM/antagonistas & inhibidores , Proteína ADAMTS4 , Animales , Benzoatos/farmacología , Bencilaminas/farmacología , Línea Celular Tumoral , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Humanos , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Ligandos , Receptores X del Hígado , Ratones , Ratones Mutantes , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/fisiología , Osteoartritis/metabolismo , Dolor/etiología , Procolágeno N-Endopeptidasa/antagonistas & inhibidores , Prostaglandina-E Sintasas , Ratas
3.
Cartilage ; 14(4): 467-472, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-36912174

RESUMEN

OBJECTIVE: This study was conducted to compare therapeutically relevant properties of platelet-rich plasma (PRP), a commonly used autologous intra-articular treatment for osteoarthritis (OA), with those of a novel placental tissue particulate, PTP-001, which is in development as a regulated biologic treatment for knee OA. DESIGN: Quantitative immunoassays were performed to determine the content of key growth/regulatory biofactors in PTP-001, and in leukocyte-rich (LR)-PRP or leukocyte-poor (LP)-PRP. An anti-inflammatory bioassay was used to evaluate the effects of each treatment on pro-inflammatory cytokine (tumor necrosis factor (TNF)-α) production in a macrophage cell culture system. Gene expression experiments were conducted using a co-culture system of human synoviocytes (pre-stimulated with interleukin (IL)-1ß) and articular chondrocytes, with quantitative polymerase chain reaction analyses of the separate cellular compartments. RESULTS: The concentrations of several biofactors (e.g., basic fibroblast growth factor, tissue inhibitor of metalloproteases-3, interleukin-1 receptor antagonist) representative of diverse disease-relevant mechanisms of action were significantly higher for PTP-001 relative to LR-PRP or LP-PRP. PTP-001 and PRP preparations were able to reduce TNF-α production in macrophage cell cultures; however, greater variability was observed for PRP in comparison with PTP-001. In the chondrocyte/synoviocyte co-culture experiments, PTP-001 and LR-PRP (but not LP-PRP) significantly reduced chondrocyte MMP13 expression in cultures containing IL-1-pretreated synoviocytes. In addition, ADAMTS5 expression was reduced in the chondrocyte compartment following treatment with PTP-001 relative to PRP. CONCLUSION: These findings support evidence of a potent, multifactorial mechanism of action for a consistently manufactured biologic (PTP-001), which may be of greater therapeutic benefit in comparison with more heterogeneous preparations of PRP which may be generated at the time of treatment.


Asunto(s)
Productos Biológicos , Osteoartritis de la Rodilla , Plasma Rico en Plaquetas , Embarazo , Humanos , Femenino , Placenta/metabolismo , Osteoartritis de la Rodilla/metabolismo , Citocinas/metabolismo , Plasma Rico en Plaquetas/metabolismo
4.
Connect Tissue Res ; 53(2): 132-41, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21966936

RESUMEN

Lubricin, also referred to as superficial zone protein, has been reported to be a proteoglycan. However, the structure of its glycosaminoglycan chain has not been well characterized, and this study was undertaken to investigate the structure of the glycosaminoglycan chain that decorated lubricin in human synovial fluid to provide insight into its biological role. Lubricin was detected as a major band at approximately 360 kDa which co-migrated in sodium dodecyl sulfate-polyacrylamide gel electrophoresis with a chondroitin sulfate (CS)-containing proteoglycan that was detected by both monoclonal antibodies (MAb) 2-B-6 and MAb 3-B-3 after chondroitinase ABC treatment and keratan sulfate (KS) that was detected by MAb 5-D-4. Further analysis of lubricin-containing fractions that eluted from an anion exchange column indicated that the major population of lubricin could be separated from the CS and KS stubs which indicated that this fraction of lubricin was not decorated with glycosaminoglycan chain and was the glycoprotein form of lubricin. Lubricin present in fractions that also contained CS was found to be decorated with CS structures which were reactive with MAb 3-B-3 after chondroitinase ABC digestion using a sandwich enzyme-linked immunosorbent assay approach. Aggrecan was not found to form complexes with lubricin in synovial fluid which confirmed that the MAb 3-B-3 CS and MAb 5-D-4 KS structures decorated lubricin. These data demonstrate that lubricin present in human synovial fluid was a heterogeneous population with both glycoprotein and proteoglycan forms.


Asunto(s)
Glicoproteínas/química , Glicosaminoglicanos/química , Líquido Sinovial/química , Fraccionamiento Químico , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Humanos , Sulfato de Queratano/química , Sulfato de Queratano/metabolismo , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/patología , Isoformas de Proteínas , Líquido Sinovial/metabolismo
5.
Nature ; 434(7033): 644-8, 2005 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-15800624

RESUMEN

Human osteoarthritis is a progressive disease of the joints characterized by degradation of articular cartilage. Although disease initiation may be multifactorial, the cartilage destruction appears to be a result of uncontrolled proteolytic extracellular matrix destruction. A major component of the cartilage extracellular matrix is aggrecan, a proteoglycan that imparts compressive resistance to the tissue. Aggrecan is cleaved at a specific 'aggrecanase' site in human osteoarthritic cartilage; this cleavage can be performed by several members of ADAMTS family of metalloproteases. The relative contribution of individual ADAMTS proteases to cartilage destruction during osteoarthritis has not been resolved. Here we describe experiments with a genetically modified mouse in which the catalytic domain of ADAMTS5 (aggrecanase-2) was deleted. After surgically induced joint instability, there was significant reduction in the severity of cartilage destruction in the ADAMTS5 knockout mice compared with wild-type mice. This is the first report of a single gene deletion capable of abrogating the course of cartilage destruction in an animal model of osteoarthritis. These results demonstrate that ADAMTS5 is the primary 'aggrecanase' responsible for aggrecan degradation in a murine model of osteoarthritis, and suggest rational strategies for therapeutic intervention in osteoarthritis.


Asunto(s)
Cartílago Articular/metabolismo , Modelos Animales de Enfermedad , Eliminación de Gen , Metaloendopeptidasas/deficiencia , Metaloendopeptidasas/genética , Osteoartritis/metabolismo , Proteínas ADAM , Proteína ADAMTS5 , Animales , Dominio Catalítico , Endopeptidasas/química , Endopeptidasas/deficiencia , Endopeptidasas/genética , Endopeptidasas/metabolismo , Exones/genética , Cabeza Femoral , Placa de Crecimiento/metabolismo , Articulaciones/patología , Articulaciones/fisiopatología , Metaloendopeptidasas/química , Metaloendopeptidasas/metabolismo , Ratones , Ratones Noqueados , Osteoartritis/genética , Osteoartritis/patología , Osteoartritis/fisiopatología , Proteoglicanos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
6.
BMC Musculoskelet Disord ; 12: 164, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21762512

RESUMEN

BACKGROUND: Tenascin-C (TN-C) is an extracellular matrix glycoprotein that is involved in tissue injury and repair processes. We analyzed TN-C expression in normal and osteoarthritic (OA) human cartilage, and evaluated its capacity to induce inflammatory and catabolic mediators in chondrocytes in vitro. The effect of TN-C on proteoglycan loss from articular cartilage in culture was also assessed. METHODS: TN-C in culture media, cartilage extracts, and synovial fluid of human and animal joints was quantified using a sandwich ELISA and/or analyzed by Western immunoblotting. mRNA expression of TN-C and aggrecanases were analyzed by Taqman assays. Human and bovine primary chondrocytes and/or explant culture systems were utilized to study TN-C induced inflammatory or catabolic mediators and proteoglycan loss. Total proteoglycan and aggrecanase -generated ARG-aggrecan fragments were quantified in human and rat synovial fluids by ELISA. RESULTS: TN-C protein and mRNA expression were significantly upregulated in OA cartilage with a concomitant elevation of TN-C levels in the synovial fluid of OA patients. IL-1 enhanced TN-C expression in articular cartilage. Addition of TN-C induced IL-6, PGE2, and nitrate release and upregulated ADAMTS4 mRNA in cultured primary human and bovine chondrocytes. TN-C treatment resulted in an increased loss of proteoglycan from cartilage explants in culture. A correlation was observed between TN-C and aggrecanase generated ARG-aggrecan fragment levels in the synovial fluid of human OA joints and in the lavage of rat joints that underwent surgical induction of OA. CONCLUSIONS: TN-C expression in the knee cartilage and TN-C levels measured in the synovial fluid are significantly enhanced in OA patients. Our findings suggest that the elevated levels of TN-C could induce inflammatory mediators and promote matrix degradation in OA joints.


Asunto(s)
Cartílago Articular/patología , Condrocitos/patología , Matriz Extracelular/metabolismo , Mediadores de Inflamación/metabolismo , Osteoartritis de la Rodilla/patología , Tenascina/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Cartílago Articular/metabolismo , Bovinos , Línea Celular Tumoral , Células Cultivadas , Condrocitos/metabolismo , Matriz Extracelular/patología , Femenino , Humanos , Mediadores de Inflamación/fisiología , Masculino , Persona de Mediana Edad , Osteoartritis de la Rodilla/etiología , Osteoartritis de la Rodilla/metabolismo , Ratas , Ratas Endogámicas Lew , Líquido Sinovial/metabolismo , Tenascina/biosíntesis , Tenascina/genética , Regulación hacia Arriba/fisiología
7.
J Biol Chem ; 284(40): 27352-9, 2009 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-19657146

RESUMEN

Mass spectrometry-based proteomic analyses performed on cartilage tissue extracts identified the serine protease HtrA1/PRSS11 as a major protein component of human articular cartilage, with elevated levels occurring in association with osteoarthritis. Overexpression of a catalytically active form of HtrA1, but not an active site mutant (S328A), caused a marked reduction in proteoglycan content in chondrocyte-seeded alginate cultures. Aggrecan degradation fragments were detected in conditioned media from the alginate cultures overexpressing active HtrA1. Incubation of native or recombinant aggrecan with wild type HtrA1 resulted in distinct cleavage of these substrates. Cleavage of aggrecan by HtrA1 was strongly enhanced by HtrA1 agonists such as CPII, a C-terminal hexapeptide derived from the C-propeptide of procollagen IIalpha1 (i.e. chondrocalcin). A novel HtrA1-susceptible cleavage site within the interglobular domain (IGD) of aggrecan was identified, and an antibody that specifically recognizes the neoepitope sequence (VQTV(356)) generated at the HtrA1 cleavage site was developed. Western blot analysis demonstrated that HtrA1-generated aggrecan fragments containing the VQTV(356) neoepitope were significantly more abundant in osteoarthritic cartilage compared with cartilage from healthy joints, implicating HtrA1 as a critical protease involved in proteoglycan turnover and cartilage degradation during degenerative joint disease.


Asunto(s)
Agrecanos/química , Agrecanos/metabolismo , Serina Endopeptidasas/metabolismo , Factores de Edad , Anciano , Anciano de 80 o más Años , Agrecanos/análisis , Agrecanos/inmunología , Alginatos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cartílago/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Condrocitos/metabolismo , Condrocitos/patología , Progresión de la Enfermedad , Epítopos/química , Epítopos/inmunología , Femenino , Regulación de la Expresión Génica , Ácido Glucurónico , Ácidos Hexurónicos , Serina Peptidasa A1 que Requiere Temperaturas Altas , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación , Osteoartritis/metabolismo , Osteoartritis/patología , Serina Endopeptidasas/genética
8.
Bioorg Med Chem Lett ; 18(24): 6454-7, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18974001

RESUMEN

N-((8-Hydroxy-5-substituted-quinolin-7-yl)(phenyl)methyl)-2-phenyloxy/amino-acetamide inhibitors of ADAMTS-5 (Aggrecanase-2) have been prepared. Selected compounds 10, 14, 25, and 53 show sub-microM ADAMTS-5 potency and good selectivity over the related metalloproteases ADAMTS-4 (Aggrecanase-1), MMP-13, and MMP-12. Compound 53 shows a good balance of potent ADAMTS-5 inhibition, moderate CYP3A4 inhibition and good rat liver microsome stability. This series of compounds represents progress towards selective ADAMTS-5 inhibitors as disease modifying osteoarthritis agents.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/química , Acetamidas/síntesis química , Acetamidas/farmacología , Proteínas ADAM/metabolismo , Proteína ADAMTS4 , Proteína ADAMTS5 , Animales , Química Farmacéutica/métodos , Citocromo P-450 CYP3A , Inhibidores del Citocromo P-450 CYP3A , Inhibidores Enzimáticos del Citocromo P-450 , Diseño de Fármacos , Humanos , Concentración 50 Inhibidora , Microsomas Hepáticos/efectos de los fármacos , Modelos Químicos , Osteoartritis/tratamiento farmacológico , Procolágeno N-Endopeptidasa/metabolismo , Ratas
9.
Open Access Rheumatol ; 10: 43-54, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29849470

RESUMEN

BACKGROUND: Pain and limitations in joint mobility associated with knee osteoarthritis (OA) are clinically challenging to manage, and advanced progression of disease can often lead to total knee arthroplasty. Intra-articular injection of hyaluronic acid (HA), also referred to as viscosupplementation, is a non-surgical treatment approach for OA, the effectiveness of which may depend on the HA composition, and the length of time over which it resides in the joint. One of the available options for such therapies includes NASHA (Durolane HA), a non-animal, biofermentation-derived product, which is manufactured using a process that stabilizes the HA molecules to slow down their rate of degradation and produce a unique formulation with a terminal half-life of ~1 month. The objectives of the current review were to assess, in patients with OA of the knee, the efficacy and safety of intra-articular treatment with NASHA relative to control (saline) injections, other HA products, and other injectables (corticosteroids, platelet-rich plasma, mesenchymal stem cells). METHODS: This systematic evidence review examines patient outcomes following NASHA treatment as described in published data from studies conducted in subjects with knee OA. A Preferred Reporting Items for Systematic Reviews and Meta-analyses-compliant literature search strategy yielded 11 eligible clinical studies with a variety of comparator arms. Outcomes assessed at various time points following intra-articular treatment included measures of pain, function, quality of life, and incidence of treatment-related adverse events (AEs). RESULTS: The available evidence reported for the clinical studies assessed demonstrates sustained and effective relief of knee OA symptoms following a single injection of NASHA. In addition, an excellent biocompatibility profile is observed for NASHA as an intra-articular therapy for OA, as reflected by the low rate of AEs associated with treatment. CONCLUSION: Treatment with NASHA is an effective and safe single-injection procedure, which can be beneficial in the clinical management of knee OA.

10.
Biochim Biophys Acta ; 1760(3): 517-24, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16507336

RESUMEN

Aggrecanases are ADAMTS (a disintegrin and metalloproteinase with thrombospondin type I motifs) proteases capable of primary (patho)physiological cleavage at specific Glu-Xaa bonds within the core protein of the hyaluronan-binding proteoglycan aggrecan. Accumulating evidence suggests that regulation of the activity of one such aggrecanase, ADAMTS-4 (or Aggrecanase-1), involves post-translational C-terminal processing (truncation) which modulates both glycosaminoglycan (GAG)-binding affinity and enzymatic activity. In the present study, we compared the effects of C-terminal truncation on the GAG-binding properties and aggrecanase activity of ADAMTS-5 (Aggrecanase-2) relative to three other ADAMTS family members, ADAMTS-9, ADAMTS-16 and ADAMTS-18. Full-length recombinant human ADAMTS-5 (M(r) approximately 85 kDa; ADAMTS-5p85) underwent autolytic cleavage during expression by CHO/A2 cells, and co-purified with C-terminally truncated (tr) isoforms of M(r) approximately 60 kDa (ADAMTS-5p60 and M(r) approximately 45 kDa (ADAMTS-5p45). All three ADAMTS-5 isoforms bound to sulfated GAGs (heparin and chondroitin sulfate (CS)). An ADAMTS-5p45 structural mimetic, terminating at Phe628 and comprising the catalytic domain, disintegrin-like domain and thrombospondin type I repeat (TSR)-1 domain (designated trADAMTS-5F628), also bound to heparin, and exhibited potent aggrecanase activity toward cleavage sites both in the aggrecan CS-2-attachment region (at Glu1771-Ala1772) and in the interglobular domain (at Glu373-Ala374). Further truncation (deletion of the TSR-1 domain) of ADAMTS-5 significantly reduced aggrecanase activity, although appreciable GAG (heparin)-binding affinity was maintained. Other TSR-1 domain-bearing truncated ADAMTS constructs demonstrating either positive GAG-binding ability (trADAMTS-9F649) or negligible GAG-affinity (trADAMTS-16F647 and trADAMTS-18F650) displayed comparably low aggrecanase activities. Thus, the presence of TSR-1 on truncated ADAMTSs appears to be necessary, but not sufficient, for effective aggrecanase-mediated catalysis of target Glu-Xaa bonds. Similarly, GAG-binding ability, irrespective of the presence of a TSR-1 domain, does not necessarily empower truncated ADAMTSs with proficient aggrecanase activity.


Asunto(s)
Proteínas ADAM/metabolismo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Endopeptidasas/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Glicosaminoglicanos/metabolismo , Lectinas Tipo C/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Proteínas ADAMTS , Proteína ADAMTS5 , Proteína ADAMTS9 , Agrecanos , Animales , Células CHO , Bovinos , Cricetinae , Cricetulus , Humanos , Isoformas de Proteínas/metabolismo
11.
J Orthop Res ; 25(3): 283-92, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17106881

RESUMEN

Lubricin is a secreted, cytoprotective glycoprotein that contributes to the essential boundary lubrication mechanisms necessary for maintaining low friction levels at articular cartilage surfaces. Diminishment of lubricin function is thereby implicated as an adverse contributing factor in degenerative joint diseases such as osteoarthritis. Lubricin occurs as a soluble component of synovial fluid, and is synthesized and localized in the superficial layer of articular cartilage (and thus has also been described as "superficial zone protein", or SZP); however, defined interactions responsible for lubricin retention at this site are not well characterized. In the current studies, we identified molecular determinants that enable lubricin to effectively bind to articular cartilage surfaces. Efficient and specific binding to the superficial zone was observed for synovial lubricin, as well as for recombinant full-length lubricin and a protein construct comprising the lubricin C-terminal (hemopexin-like) domain (LUB-C, encoded by exons 7-12). A construct representing the N-terminal region of lubricin (LUB-N, encoded by exons 2-5) exhibited no appreciable cartilage-binding ability, but displayed the capacity to dimerize, and thus potentially influence lubricin aggregation. Disulfide bond disruption significantly attenuated recombinant lubricin and LUB-C binding to cartilage surfaces, demonstrating a requirement for protein secondary structure in facilitating the appropriate localization of lubricin at relevant tissue interfaces. These findings help identify additional key attributes contributing to lubricin functionality, which would be expected to be instrumental in maintaining joint homeostasis.


Asunto(s)
Cartílago Articular/metabolismo , Glicoproteínas/metabolismo , Animales , Bovinos , Glicoproteínas/química , Glicoproteínas/aislamiento & purificación , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
12.
Front Biosci ; 11: 544-69, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16146752

RESUMEN

Members of the MMP (matrix metalloproteinase) and ADAMTS (a disintegrin and metalloproteinase with thrombospondin type I motifs) families of enzymes are capable of cleaving a diverse array of cellular, extracellular and extracellular matrix substrates, including collagens and procollagens, proteoglycans, cytokines and cytokine ligands, chemokines, elastin and von Willebrand factor, thereby modulating tissue structure and function during both health and disease. Physiologically relevant roles attributable to various members of these metalloproteinase families have been discerned from functional studies correlating in vitro substrate processing events with catabolic cleavages occurring in vivo/in situ, and the consequences thereof. Mechanisms regulating the post-translational activities of MMPs and ADAMTSs can clearly also have an influential impact on cell metabolism and tissue structure/function, and a number of functional studies have addressed the contributions of ancillary (non-catalytic) domains and endogenous inhibitors in this regard. Further revelations and affirmations of proteinase function, in an in vivo context, have emanated with the characterization of genetically manipulated animals misexpressing specific MMPs or ADAMTSs (or their substrates). An increased understanding thereby attained for the physiological functions of MMPs and ADAMTSs, and the means by which their activities are controlled, may lead to the realization of rational therapeutic strategies to counteract pathologies associated with aberrant proteolysis of homeostatic tissue macromolecules.


Asunto(s)
Proteínas ADAM/fisiología , Metaloproteinasas de la Matriz/fisiología , Proteínas ADAM/metabolismo , Secuencias de Aminoácidos , Animales , Catálisis , Dominio Catalítico , Proteínas de la Matriz Extracelular , Humanos , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Invasividad Neoplásica , Neoplasias/patología , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Especificidad por Sustrato
13.
Matrix Biol ; 21(7): 593-602, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12475643

RESUMEN

Cartilage superficial zone protein/proteoglycan (SZP) or proteoglycan 4 (PRG4), has been demonstrated to have the potential for several distinct biological functions including cytoprotection, lubrication and matrix binding. In the present study, we have examined both the immunolocalisation and the mRNA expression pattern of PRG4 in tissue harvested from the compressed and tensional regions of young and mature bovine tendons. Immunohistochemical analyses, utilizing monoclonal antibody 3-A-4 which recognizes a conformational-dependent epitope on native PRG4, demonstrated that PRG4 is present predominantly at the surface of fibrocartilaginous regions of tendon, with the intensity of immunoreactivity in this region increasing with age. RT-PCR analyses revealed that the expression of PRG4 mRNA can be modulated by exposure to cytokines and growth factors. In addition, analyses of human pathological tendon revealed that PRG4 may also be expressed as an alternatively spliced form lacking exons which encode part of the N-terminal matrix-binding and cell-proliferative domain; however, it remains to be determined whether such splice variants are a feature of human tendon, regardless of disease state. Taken together, these data indicate that PRG4 may play an important cytoprotective role by preventing cellular adhesion to the tendon surface as well as providing lubrication during normal tendon function, in a manner complimentary to cartilage PRG4. Structural modifications to SZP, together with a reduction in synthesis during tendon inflammation with injury and disease may account for the formation of tendon adhesions and contribute to the overall dysfunction of the tissue.


Asunto(s)
Proteoglicanos/metabolismo , Tendones/metabolismo , Animales , Bovinos , Humanos , Inmunohistoquímica , Proteoglicanos/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Matrix Biol ; 21(3): 271-88, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12009333

RESUMEN

Monoclonal antibody (MAb) technology was used to examine aggrecan metabolites and the role of aggrecanases and matrix metalloproteinases (MMPs) in proteolysis of the interglobular domain (IGD) and C-terminus of aggrecan. An in vitro model of progressive cartilage degradation characterized by early proteoglycan loss and late stage collagen catabolism was evaluated in conjunction with a broad-spectrum inhibitor of MMPs. We have for the first time demonstrated that IGD cleavage by MMPs occurs during this late stage cartilage degeneration, both as a primary event in association with glycosaminoglycan (GAG) release from the tissue and secondarily in trimming of aggrecanase-generated G1 metabolites. Additionally, we have shown that MMPs were responsible for C-terminal catabolism of aggrecan and generation of chondroitin sulfate (CS) deficient aggrecan monomers and that this aggrecan truncation occurred prior to detectable IGD cleavage by MMPs. The onset of this later stage MMP activity was also evident by the generation of MMP-specific link protein catabolites in this model culture system. Recombinant MMP-1, -3 and -13 were all capable of C-terminally truncating aggrecan with at least two cleavage sites N-terminal to the CS attachment domains of aggrecan. Through analysis of aggrecan metabolites in pathological synovial fluids from human, canine and equine sources, we have demonstrated the presence of aggrecan catabolites that appear to have resulted from similar C-terminal processing of aggrecan as that induced in our in vitro culture systems. Finally, by developing a new MAb recognizing a linear epitope in the IGD of aggrecan, we have identified two novel aggrecan metabolites generated by an as yet unidentified proteolytic event. Collectively, these results suggest that C-terminal processing of aggrecan by MMPs may contribute to the depletion of cartilage GAG that leads to loss of tissue function in aging and disease. Furthermore, analysis of aggrecan metabolites resulting from both C-terminal and IGD cleavage by MMPs may prove useful in monitoring different stages in the progression of cartilage degeneration.


Asunto(s)
Cartílago Articular/metabolismo , Colagenasas/metabolismo , Proteínas de la Matriz Extracelular , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Proteoglicanos/metabolismo , Agrecanos , Animales , Sitios de Unión , Cartílago Articular/patología , Bovinos , Colágeno/metabolismo , Lectinas Tipo C , Metaloproteinasa 13 de la Matriz , Factores de Tiempo
15.
Matrix Biol ; 23(4): 219-30, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15296936

RESUMEN

Members of the ADAMTS (a disintegrin and metalloprotease with thrombospondin motifs) family share common structural features including a disintegrin domain, a zinc metalloprotease domain, and at least one thrombospondin motif. Aberrant expression of several of these proteins has led to an understanding of their role in human disease; however, a link to function for many has not yet been made. One such uncharacterized family member, ADAMTS-8, shares significant protein sequence homology with a subgroup of ADAMTSs that includes ADAMTS-1, ADAMTS-4, ADAMTS-5, and ADAMTS-15. Each of these proteases has been shown to cleave 'aggrecanase-susceptible' site(s) within the extracellular matrix (ECM) proteoglycan aggrecan, and ADAMTS-4 and ADAMTS-5 have been postulated to play a role in the depletion of articular cartilage in osteoarthritic disease. Based on sequence relationships, in the present study we examined the ability of ADAMTS-8 to exhibit 'aggrecanase' activity. A neoepitope monoclonal antibody (MAb; AGG-C1; anti-NITEGE373) was developed and used to demonstrate the ability of ADAMTS-8 to cleave aggrecan at the aggrecanase-susceptible Glu373-Ala374 peptide bond. In addition, expression analyses demonstrated the presence of ADAMTS-8 mRNA transcripts in normal and osteoarthritic human cartilage.


Asunto(s)
Cartílago Articular/enzimología , Endopeptidasas/metabolismo , Metaloendopeptidasas/metabolismo , Proteínas ADAM , Proteína ADAMTS9 , Agrecanos , Animales , Anticuerpos Monoclonales/inmunología , Western Blotting , Células CHO , Cricetinae , Cricetulus , Ensayo de Inmunoadsorción Enzimática/métodos , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Lectinas Tipo C , Metaloendopeptidasas/genética , Metaloendopeptidasas/inmunología , Metaloendopeptidasas/aislamiento & purificación , Osteoartritis/metabolismo , Reacción en Cadena de la Polimerasa , Proteoglicanos/metabolismo , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Distribución Tisular
16.
Biochem Soc Symp ; (70): 107-14, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14587286

RESUMEN

Degradation of the cartilage proteoglycan, aggrecan, is an essential aspect of normal growth and development, and of joint pathology. The roles of different proteolytic enzymes in this process can be determined from the sites of cleavage in the aggrecan core protein, which generates novel termini (neoepitopes). Antibodies specific for the different neoepitopes generated by such cleavage events provide powerful tools with which to analyse these processes. The same approach can be used to differentiate the processed, active forms of proteases from their inactive pro-forms. Since the proteolytic processing of these enzymes requires the removal of the inhibitory pro-region, it also results in the generation of N-terminal neoepitopes. Using the newborn rat long bone as a model system, it was shown that the active form of ADAMTS-4 [ADAM (a disintegrin and metalloproteinase) with thrombospondin motifs-4], but not ADAMTS-5, co-localizes with the aggrecan cleavage neoepitopes known to be produced by this metalloproteinase. Thus, in long bone growth, aggrecan turnover seems to be dependent on ADAMTS-4 activity. To demonstrate the molecular basis of the specificity of anti-neoepitope antibodies, the Fv region of a monoclonal antibody specific for a neoepitope generated by the ADAMTS-4-mediated cleavage of aggrecan has been modelled and the binding of the peptide epitope simulated. In the docked structure, the N-terminus of the peptide antigen is clearly buried in the binding-site cavity. The absence of an open cleft makes it impossible for the intact substrate to pass through the binding site, providing a rationale for the specificity of this class of antibodies.


Asunto(s)
Anticuerpos/inmunología , Especificidad de Anticuerpos , Cartílago Articular/metabolismo , Epítopos/inmunología , Secuencia de Aminoácidos , Cartílago Articular/inmunología , Epítopos/química , Humanos , Hidrólisis , Datos de Secuencia Molecular
17.
PLoS One ; 8(9): e74094, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24040176

RESUMEN

HTRA1 is a member of the High Temperature Requirement (HTRA1) family of serine proteases, which play a role in several biological and pathological processes. In part, HTRA1 regulation occurs by inhibiting the TGF-ß signaling pathway, however the mechanism of inhibition has not been fully defined. Previous studies have shown that HTRA1 is expressed in a variety of tissues, including sites of skeletal development. HTRA1 has also been implicated in the process of bone formation, although the precise manner of regulation is still unknown. This study investigated how HTRA1 regulates TGF-ß signaling and examined the in vivo effects of the loss of HTRA1. We demonstrated that recombinant HTRA1 was capable of cleaving both type II and type III TGF-ß receptors (TßRII and TßRIII) in vitro in a dose-dependent manner, but it did not affect the integrity of TßRI or TGF-ß. Overexpression of HTRA1 led to decreased levels of both TßRII and III on the cell surface but had no effect on TßRI. Silencing HTRA1 expression significantly increased TGF-ß binding to the cell surface and TGF-ß responsiveness within the cell. To examine the role of HTRA1 in vivo, we generated mice with a targeted gene deletion of HTRA1. Embryonic fibroblasts isolated from these mice displayed an increase in TGF-ß-induced expression of several genes known to promote bone formation. Importantly, the loss of HTRA1 in the knockout mice resulted in a marked increase in trabecular bone mass. This study has identified a novel regulatory mechanism by which HTRA1 antagonizes TGF-ß signaling, and has shown that HTRA1 plays a key role in the regulation of bone formation.


Asunto(s)
Osteogénesis/fisiología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Serina Endopeptidasas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular , Fibroblastos/metabolismo , Orden Génico , Silenciador del Gen , Serina Peptidasa A1 que Requiere Temperaturas Altas , Humanos , Masculino , Ratones , Ratones Noqueados , Unión Proteica , Proteolisis , Serina Endopeptidasas/genética , Transcripción Genética
18.
AAPS J ; 14(1): 97-104, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22228117

RESUMEN

We have recently demonstrated that intra-articular (IA) administration of human recombinant lubricin, LUB:1, significantly inhibited cartilage degeneration and pain in the rat meniscal tear model of post-traumatic arthritis. In this report, we show that after a single IA injection to naïve rats and rats that underwent unilateral meniscal tear, [(125)I]LUB:1 had a tri-phasic disposition profile, with the alpha, beta, and gamma half-life estimates of 4.5 h, 1.5 days, and 2.1 weeks, respectively. We hypothesize that the terminal phase kinetics was related to [(125)I]LUB:1 binding to its ligands. [(125)I]LUB:1 was detected on articular cartilage surfaces as long as 28 days after single IA injection. Micro-autoradiography analysis suggested that [(125)I]LUB:1 tended to localize to damaged joint surfaces in rats with meniscal tear. After a single intravenous (IV) dose to rats, [(125)I]LUB:1 was eliminated rapidly from the systemic circulation, with a mean total body clearance of 154 mL/h/kg and a mean elimination half-life (t (1/2)) of 6.7 h. Overall, LUB:1 has met a desired disposition profile of a potential therapeutic intended for an IA administration: target tissue (knee) retention and fast elimination from the systemic circulation after a single IA or IV dose.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Glicoproteínas/farmacocinética , Articulación de la Rodilla/efectos de los fármacos , Animales , Artritis Experimental/patología , Autorradiografía/métodos , Células CHO , Cricetinae , Cricetulus , Femenino , Glicoproteínas/administración & dosificación , Glicoproteínas/farmacología , Semivida , Humanos , Inyecciones Intraarticulares , Inyecciones Intravenosas , Articulación de la Rodilla/patología , Masculino , Ratas , Ratas Endogámicas Lew , Proteínas Recombinantes , Factores de Tiempo , Distribución Tisular
19.
Curr Drug Targets ; 11(5): 614-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20199389

RESUMEN

Therapeutic alleviation of the pathophysiology of osteoarthritis (OA) is a great and unmet medical challenge. At the basic science level, significant progress has facilitated the identification of distinct pathways and targets which appear to be central to the OA-associated deterioration of articular cartilage. For example, the dysregulated activities of aggrecanases such as ADAMTS-4 and ADAMTS-5, and collagenases such as MMP-13, point to strategies for the development of selective protease inhibitors to curtail OA disease progression. Likewise, blockade of disease-associated "pro-catabolic" cytokines may offer promising opportunities in this regard. Other novel biotherapeutic approaches are also emerging, including the use of recombinant lubricin molecules for intraarticular supplementation. Expression profiling of cartilage (and other joint tissues) to identify OA-associated genes continues to yield new potential therapeutic options, including the 'upstream' targeting of key intracellular regulators. Moving forward into the clinic, the critical evaluation and optimization of modalities for therapeutic delivery, as well as the availability and utility of appropriate disease biomarkers and ability to determine relevant patient populations, will be other important considerations in directing the advancement of novel OA therapies.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Glicoproteínas/uso terapéutico , Osteoartritis/tratamiento farmacológico , Inhibidores de Proteasas/uso terapéutico , Agrecanos/metabolismo , Animales , Cartílago/metabolismo , Citocinas/antagonistas & inhibidores , Vías de Administración de Medicamentos , Glicoproteínas/metabolismo , Humanos , Osteoartritis/metabolismo , Inhibidores de Proteasas/farmacología
20.
Arthritis Res Ther ; 12(6): R229, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21194441

RESUMEN

INTRODUCTION: Acute trauma involving the anterior cruciate ligament is believed to be a major risk factor for the development of post-traumatic osteoarthritis 10 to 20 years post-injury. In this study, to better understand the early biological changes which occur after acute injury, we investigated synovial fluid and serum biomarkers. METHODS: We collected serum from 11 patients without pre-existing osteoarthritis from a pilot intervention trial (5 placebo and 6 drug treated) using an intra-articular interleukin-1 receptor antagonist (IL-1Ra) therapy, 9 of which also supplied matched synovial fluid samples at presentation to the clinic after acute knee injury (mean 15.2 ± 7.2 days) and at the follow-up visit for reconstructive surgery (mean 47.6 ± 12.4 days). To exclude patients with pre-existing osteoarthritis (OA), the study was limited to individuals younger than 40 years of age (mean 23 ± 3.5) with no prior history of joint symptoms or trauma. We profiled a total of 21 biomarkers; 20 biomarkers in synovial fluid and 13 in serum with 12 biomarkers measured in both fluids. Biomarkers analyzed in this study were found to be independent of treatment (P > 0.05) as measured by Mann-Whitney and two-way ANOVA. RESULTS: We observed significant decreases in synovial fluid (sf) biomarker concentrations from baseline to follow-up for (sf)C-Reactive protein (CRP) (P = 0.039), (sf)lubricin (P = 0.008) and the proteoglycan biomarkers: (sf)Glycosaminoglycan (GAG) (P = 0.019), and (sf)Alanine-Arginine-Glycine-Serine (ARGS) aggrecan (P = 0.004). In contrast, we observed significant increases in the collagen biomarkers: (sf)C-terminal crosslinked telopeptide type II collagen (CTxII) (P = 0.012), (sf)C1,2C (P = 0.039), (sf)C-terminal crosslinked telopeptide type I collagen (CTxI) (P = 0.004), and (sf)N-terminal telopeptides of type I collagen (NTx) (P = 0.008). The concentrations of seven biomarkers were significantly higher in synovial fluid than serum suggesting release from the signal knee: IL-1ß (P < 0.0001), fetal aggrecan FA846 (P = 0.0001), CTxI (P = 0.0002), NTx (P = 0.012), osteocalcin (P = 0.012), Cartilage oligomeric matrix protein (COMP) (P = 0.0001) and matrix metalloproteinase (MMP)-3 (P = 0.0001). For these seven biomarkers we found significant correlations between the serum and synovial fluid concentrations for only CTxI (P = 0.0002), NTx (P < 0.0001), osteocalcin (P = 0.0002) and MMP-3 (P = 0.038). CONCLUSIONS: These data strongly suggest that the biology after acute injury reflects that seen in cartilage explant models stimulated with pro-inflammatory cytokines, which are characterized by an initial wave of proteoglycan loss followed by subsequent collagen loss. As the rise of collagen biomarkers in synovial fluid occurs within the first month after injury, and as collagen loss is thought to be irreversible, very early treatment with agents to either reduce inflammation and/or reduce collagen loss may have the potential to reduce the onset of future post-traumatic osteoarthritis. TRIAL REGISTRATION: The samples used in this study were derived from a clinical trial NCT00332254 registered with ClinicalTrial.gov.


Asunto(s)
Biomarcadores/análisis , Colágeno/análisis , Traumatismos de la Rodilla/metabolismo , Proteoglicanos/análisis , Antiinflamatorios/administración & dosificación , Colágeno/metabolismo , Método Doble Ciego , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inflamación/metabolismo , Inyecciones Intraarticulares , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Traumatismos de la Rodilla/tratamiento farmacológico , Masculino , Proyectos Piloto , Proteoglicanos/metabolismo , Líquido Sinovial/química , Adulto Joven
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda