Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
1.
Eur Cell Mater ; 37: 347-359, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31044415

ABSTRACT

Osteoarthritis (OA) is a degenerative joint disease characterised by a progressive degradation of articular cartilage and underlaying bone and is associated with pain and disability. Currently, there is no medical treatment to reverse or even retard OA. Based on our previous reports, where we establish the repair potential of short Link N (sLN) in the intervertebral disc, a cartilage-like tissue, we hypothesise that sLN may hold similar promises in the repair of articular cartilage. This study aimed to determine if sLN, could prevent OA disease progression. Skeletally mature New Zealand white rabbits underwent unilateral anterior cruciate ligament transection (ACLT) of their left femorotibial joints to induce joint degeneration typical of OA. Beginning 3 weeks post-operatively, and every three weeks thereafter for 12 weeks, either saline (1 mL) or sLN (100 µg in 1 mL saline) was injected intraarticularly into the operated knee. Six additional rabbits underwent sham surgery but without ACLT or post-operative injections. The effects on gross joint morphology and cartilage histologic changes were evaluated. In the Saline group, prominent erosion of articular cartilage occurred in both femoral condyle compartments and the lateral compartment of the tibial plateau while, sLN treatment reduced the severity of the cartilage damage in these compartments of the knee showing erosion. Furthermore, statistically significant differences were detected between the joint OA score of the saline and sLN treated groups (p = 0.0118). Therefore, periodic intraarticular injection of sLN is a promising nonsurgical treatment for preventing or retarding OA progression, by reducing cartilage degradation.


Subject(s)
Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/pharmacology , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Proteoglycans/metabolism , Proteoglycans/pharmacology , Animals , Anterior Cruciate Ligament/drug effects , Anterior Cruciate Ligament/metabolism , Anterior Cruciate Ligament Injuries/drug therapy , Anterior Cruciate Ligament Injuries/metabolism , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Disease Models, Animal , Disease Progression , Femur/drug effects , Femur/metabolism , Injections, Intra-Articular/methods , Knee Joint/drug effects , Knee Joint/metabolism , Rabbits , Tibia/drug effects , Tibia/metabolism
2.
Eur Cell Mater ; 31: 26-39, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26728497

ABSTRACT

Low back pain originating from intervertebral disc (IVD) degeneration affects the quality of life for millions of people, and it is a major contributor to global healthcare costs. Long-term culture of intact IVDs is necessary to develop ex vivo models of human IVD degeneration and repair, where the relationship between mechanobiology, disc matrix composition and metabolism can be better understood. A bioreactor was developed that facilitates culture of intact human IVDs in a controlled, dynamically loaded environment. Tissue integrity and cell viability was evaluated under 3 different loading conditions: low 0.1-0.3, medium 0.1-0.3 and high 0.1-1.2 MPa. Cell viability was maintained > 80 % throughout the disc at low and medium loads, whereas it dropped to approximately 70 % (NP) and 50 % (AF) under high loads. Although cell viability was affected at high loads, there was no evidence of sGAG loss, changes in newly synthesised collagen type II or chondroadherin fragmentation. Sulphated GAG content remained at a stable level of approximately 50 µg sGAG/mg tissue in all loading protocols. To evaluate the feasibility of tissue repair strategies with cell supplementation, human NP cells were transplanted into discs within a thermoreversible hyaluronan hydrogel. The discs were loaded under medium loads, and the injected cells remained largely localised to the NP region. This study demonstrates the feasibility of culturing human IVDs for 14 days under cyclic dynamic loading conditions. The system allows the determination a safe range-of-loading and presents a platform to evaluate cell therapies and help to elucidate the effect of load following cell-based therapies.


Subject(s)
Bioreactors , Cell- and Tissue-Based Therapy/methods , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Degeneration/therapy , Intervertebral Disc/cytology , Adolescent , Adult , Aged , Cell Survival , Child , Female , Guided Tissue Regeneration , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate , Low Back Pain/etiology , Low Back Pain/therapy , Male , Middle Aged , Models, Biological , Organ Culture Techniques , Stress, Physiological/physiology , Young Adult
3.
Eur Cell Mater ; 28: 98-110; discussion 110-1, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25214017

ABSTRACT

Excessive mechanical loading or acute trauma to intervertebral discs (IVDs) is thought to contribute to degeneration and pain. However, the exact mechanisms by which mechanical injury initiates and promotes degeneration remain unclear. This study investigates biochemical changes and extracellular matrix disruption in whole-organ human IVD cultures following acute mechanical injury. Isolated healthy human IVDs were rapidly compressed by 5% (non-injured) or 30% (injured) of disc height. 30% strain consistently cracked cartilage endplates, confirming disc trauma. Three days post-loading, conditioned media were assessed for proteoglycan content and released cytokines. Tissue extracts were assessed for proteoglycan content and for aggrecan integrity. Conditioned media were applied to PC12 cells to evaluate if factors inducing neurite growth were released. Compared to controls, IVD injury caused significant cell death. Injury also caused significantly reduced tissue proteoglycan content with a reciprocal increase of proteoglycan content in culture media. Increased aggrecan fragmentation was observed in injured tissue due to increased matrix metalloproteinase and aggrecanase activity. Injured-IVD conditioned media contained significantly elevated interleukin (IL)-5, IL-6, IL-7, IL-8, MCP-2, GROα, and MIG, and ELISA analysis showed significantly increased nerve growth factor levels compared to non-injured media. Injured-disc media caused significant neurite sprouting in PC12 cells compared to non-injured media. Acute mechanical injury of human IVDs ex vivo initiates release of factors and enzyme activity associated with degeneration and back pain. This work provides direct evidence linking acute trauma, inflammatory factors, neo-innervation and potential degeneration and discogenic pain in vivo.


Subject(s)
Extracellular Matrix Proteins/metabolism , Intervertebral Disc Degeneration/etiology , Intervertebral Disc/metabolism , Stress, Mechanical , Adult , Cell Death , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Fractures, Cartilage/complications , Fractures, Cartilage/metabolism , Humans , Intervertebral Disc/injuries , Intervertebral Disc Degeneration/metabolism , Middle Aged , Neurites/drug effects , Pain/etiology , Pain/metabolism
4.
Eur Cell Mater ; 28: 129-36; discussion 136, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25214019

ABSTRACT

Analysis of both the aggregated and non-aggregated fractions of aggrecan isolated from adult human intervertebral disc using immunoblotting with antibodies specific for the different domains constituting the aggrecan core protein or atomic force microscopy revealed that many components contained the G1 domain. However, little of the disc aggrecan was able to reform aggregates with hyaluronan, as determined by gel filtration chromatography, suggesting that the G1 domains had been rendered non-functional. Since previous studies have shown that disc aggrecan undergoes non-enzymatic glycation with age, the functional effect of such modification was investigated in vitro using bovine aggrecan isolated from young animals. Incubation of monomeric aggrecan with ribose to induce glycation rendered it unable to form complexes with hyaluronan stable to agarose gel electrophoresis or gel filtration chromatography. Similarly, extended treatment of intact proteoglycan aggregate with ribose resulted in destabilisation of the complex with separation of the aggrecan from the hyaluronan. Although it is clear that proteolysis occurs in the intervertebral disc and gives rise to some non-aggregating molecules, a different mechanism is required to explain the presence of many non-aggregating molecules bearing the G1 domain. The products of non-enzymatic glycation of the globular domains of aggrecan would account for this phenomenon and explain why some of the non-aggregating molecules are still large proteoglycans. While such molecules may be retained in the nucleus pulposus, they may be able to diffuse within it, reducing the ability of the tissue to resist compression under asymmetric loading such as bending and ultimately contributing to disc degeneration.


Subject(s)
Aggrecans/metabolism , Intervertebral Disc/metabolism , Proteolysis , Adult , Animals , Cattle , Humans , Protein Aggregates
5.
Biochem Biophys Res Commun ; 433(1): 151-6, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23438440

ABSTRACT

This study examines the effect of TNFα on whole bovine intervertebral discs in organ culture and its association with changes characteristic of intervertebral disc degeneration (IDD) in order to inform future treatments to mitigate the chronic inflammatory state commonly found with painful IDD. Pro-inflammatory cytokines such as TNFα contribute to disc pathology and are implicated in the catabolic phenotype associated with painful IDD. Whole bovine discs were cultured to examine cellular (anabolic/catabolic gene expression, cell viability and senescence using ß-galactosidase) and structural (histology and aggrecan degradation) changes in response to TNFα treatment. Control or TNFα cultures were assessed at 7 and 21 days; the 21 day group also included a recovery group with 7 days TNFα followed by 14 days in basal media. TNFα induced catabolic and anti-anabolic shifts in the nucleus pulposus (NP) and annulus fibrosus (AF) at 7 days and this persisted until 21 days however cell viability was not affected. Data indicates that TNFα increased aggrecan degradation products and suggests increased ß-galactosidase staining at 21 days without any recovery. TNFα treatment of whole bovine discs for 7 days induced changes similar to the degeneration processes that occur in human IDD: aggrecan degradation, increased catabolism, pro-inflammatory cytokines and nerve growth factor expression. TNFα significantly reduced anabolism in cultured IVDs and a possible mechanism may be associated with cell senescence. Results therefore suggest that successful treatments must promote anabolism and cell proliferation in addition to limiting inflammation.


Subject(s)
Intervertebral Disc Degeneration/etiology , Tumor Necrosis Factor-alpha/physiology , Animals , Cattle , Cell Survival , Cellular Senescence , Disease Models, Animal , Gene Expression , Humans , Inflammation Mediators/physiology , Intervertebral Disc/pathology , Intervertebral Disc/physiopathology , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Degeneration/physiopathology , Organ Culture Techniques , Tumor Necrosis Factor-alpha/administration & dosage
6.
Osteoarthritis Cartilage ; 19(8): 1011-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21549847

ABSTRACT

BACKGROUND: Complex loading develops in multiple spinal motions and in the case of hyperflexion is known to cause intervertebral disc (IVD) injury. Few studies have examined the interacting biologic and structural alterations associated with potentially injurious complex loading, which may be an important contributor to chronic progressive degeneration. OBJECTIVE: This study tested the hypothesis that low magnitudes of axial compression loading applied asymmetrically can induce IVD injury affecting cellular and structural responses in a large animal IVD ex-vivo model. METHODS: Bovine caudal IVDs were assigned to either a control or wedge group (15°) and placed in organ culture for 7 days under static 0.2MPa load. IVD tissue and cellular responses were assessed through confined compression, qRT-PCR, histology and structural and compositional measurements, including Western blot for aggrecan degradation products. RESULTS: Complex loading via asymmetric compression induced cell death, an increase in caspase-3 staining (apoptosis), a loss of aggrecan and an increase in aggregate modulus in the concave annulus fibrosis. While an up-regulation of MMP-1, ADAMTS4, IL-1ß, and IL-6 mRNA, and a reduced aggregate modulus were induced in the convex annulus. CONCLUSION: Asymmetric compression had direct deleterious effects on both tissue and cells, suggesting an injurious loading regime that could lead to a degenerative cascade, including cell death, the production of inflammatory mediators, and a shift towards catabolism. This explant model is useful to assess how injurious mechanical loading affects the cellular response which may contribute to the progression of degenerative changes in large animal IVDs, and results suggest that interventions should address inflammation, apoptosis, and lamellar integrity.


Subject(s)
Intervertebral Disc/physiopathology , Spinal Diseases/physiopathology , Weight-Bearing/physiology , ADAM Proteins/metabolism , ADAMTS4 Protein , Aggrecans/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Case-Control Studies , Caspase 3/metabolism , Cattle , Cell Death/physiology , Disease Models, Animal , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Intervertebral Disc/injuries , Intervertebral Disc/metabolism , Matrix Metalloproteinase 1/metabolism , Procollagen N-Endopeptidase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spinal Diseases/metabolism
7.
Eur Cell Mater ; 21: 31-45, 2011 Jan 12.
Article in English | MEDLINE | ID: mdl-21225593

ABSTRACT

The relative contribution of a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)4 and ADAMTS5 to aggrecan degradation under oncostatin M (OSM) stimulation, the role of the ancillary domains of the aggrecanases on their ability to cleave within the chondroitin sulfate (CS)-2 region, the role of hyaluronidases (HYAL) in stimulating aggrecan release in the absence of proteolysis, and the identity of the hyaluronidase involved in OSM-mediated cartilage breakdown were investigated. Bovine articular cartilage explants were cultured in the presence of interleukin-1beta (IL-1beta), tumor necrosis factor alpha (TNFalpha) and/or OSM, or treated with trypsin and/or hyaluronidase. Aggrecan was digested with various domain-truncated isoforms of ADAMTS4 and ADAMTS5. Aggrecan and link protein degradation and release were analyzed by immunoblotting. Aggrecanase and HYAL gene expression were determined. ADAMTS4 was the most inducible aggrecanase upon cytokine stimulation, whereas ADAMTS5 was the most abundant aggrecanase. ADAMTS5 was the most active aggrecanase and was responsible for the generation of an OSM-specific degradation pattern in the CS-2 region. Its ability to cleave at the OSM-specific site adjacent to the aggrecan G3 region was enhanced by truncation of the C-terminal thrombospondin domain, but reduced by further truncation of both the spacer and cysteine-rich domains of the enzyme. OSM has the ability to mediate proteoglycan release through hyaluronan degradation, under conditions where HYAL-2 is the predominant hyaluronidase being expressed. Compared to other catabolic cytokines, OSM exhibits a unique potential at degrading the proteoglycan aggregate, by promoting early robust aggrecanolysis, primarily through the action of ADAMTS5, and hyaluronan degradation.


Subject(s)
ADAM Proteins/metabolism , Aggrecans/metabolism , Cartilage, Articular/metabolism , Chondroitin Sulfates/metabolism , Hyaluronoglucosaminidase/metabolism , Oncostatin M/metabolism , Animals , Cattle , Cells, Cultured , Cytokines/metabolism , Electrophoresis, Polyacrylamide Gel , Hyaluronoglucosaminidase/genetics , Immunoblotting , Interleukin-1beta/metabolism , Metalloproteases/genetics , Metalloproteases/metabolism , Osteoarthritis/metabolism , Protein Isoforms , Thrombospondins/genetics , Thrombospondins/metabolism , Tissue Culture Techniques , Trypsin/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Osteoarthritis Cartilage ; 16(10): 1245-52, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18372197

ABSTRACT

OBJECTIVE: In a previous study, we identified a 50-kDa G3-containing aggrecan degradation product in bovine cartilage, released from the tissue after interleukin-1 (IL-1) stimulation in the presence of oncostatin M (OSM). Our objective was to purify, determine the N-terminal sequence of this fragment and verify whether this cleavage could be attributed to a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4 and ADAMTS-5 action in vitro. METHODS: Collected media from bovine cartilage explant cultures stimulated with IL-1+OSM were subjected to anion-exchange chromatography. The N-terminal sequence of the fragment of interest in the purified fractions was determined by automated Edman sequencing. Fetal bovine aggrecan was digested with full-length recombinant ADAMTS-4 and ADAMTS-5 and resulting degradation products were analyzed by sodium dodecyl sulfate/polyacrylamide gel electrophoresis (SDS/PAGE) and immunoblotting using an anti-G3 antiserum and an anti-neoepitope antibody that had been generated to the new N-terminus of the G3 fragment. RESULTS: Characterization of the 50-kDa fragment showed that it possesses chondroitin sulfate (CS) and is the result of a cleavage within the C-terminal portion of the CS-2 domain, adjacent to the G3 region. Sequence analysis identified the cleavage region as TQRPAE(2047)-(2048)ARLEIE, suggesting an aggrecanase-derived product. Using an anti-neoepitope antibody specific for the additional cleavage site, it was shown that the product is generated in vitro upon digestion of aggrecan by ADAMTS-5 and, to a much lesser extent, by ADAMTS-4. CONCLUSIONS: The abundance and rapid rate of release of this degradation product in organ cultures in the presence of OSM suggest that it could result from a unique aggrecan proteolysis mediated by aggrecanases.


Subject(s)
ADAM Proteins/metabolism , Aggrecans/metabolism , Proteoglycans/metabolism , ADAM Proteins/chemistry , Aggrecans/chemistry , Animals , Cartilage, Articular/metabolism , Cattle , Chondrocytes/metabolism , Chondroitin Sulfates/chemistry , Interleukin-1 , Oncostatin M , Proteoglycans/chemistry
9.
Osteoarthritis Cartilage ; 16(1): 98-104, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17574450

ABSTRACT

OBJECTIVE: To investigate the potential synergistic and differential effects of cytokine combinations on proteoglycan aggregate catabolism in cartilage. METHODS: Bovine articular cartilage explants were maintained in organ culture and subjected to stimulation with cytokine combinations including interleukin-1alpha (IL-1alpha), IL-1beta, IL-6, IL-17, tumor necrosis factor-alpha (TNFalpha) and oncostatin M (OSM). Aggrecan, link protein and hyaluronan (HA) release and degradation were analyzed, and the effect of the hyaluronidase inhibitor apigenin was investigated. RESULTS: For all cytokine mixtures studied cleavage of aggrecan only by aggrecanase action was apparent. However, OSM acting synergistically with IL-1 or TNFalpha produced a rapid release of all proteoglycan aggregate components due to both aggrecan and HA degradation. This was abolished by the hyaluronidase inhibitor, apigenin. In addition, in the presence of OSM a low molecular weight aggrecan G3 product was observed, suggesting altered aggrecanase cleavage activity is induced by this cytokine. CONCLUSIONS: Under cytokine stimulation, aggrecan release from cartilage may take place via proteolysis of the aggrecan core protein or via depolymerization of HA, with the latter mechanism being induced by OSM. OSM is associated with joint inflammation and its participation may account for the more rapid loss of aggrecan from articular cartilage in the inflammatory arthritides, compared to osteoarthritis.


Subject(s)
Cartilage, Articular/metabolism , Extracellular Matrix Proteins/metabolism , Interleukins/pharmacology , Oncostatin M/pharmacology , Proteoglycans/drug effects , Aggrecans/metabolism , Animals , Apigenin/pharmacology , Cartilage, Articular/drug effects , Cattle , Cytokines/pharmacology , Drug Synergism , Enzyme Inhibitors/pharmacology , Glycosaminoglycans/metabolism , Hyaluronic Acid/metabolism , Proteoglycans/metabolism , Tumor Necrosis Factor-alpha/pharmacology
10.
Matrix Biol ; 26(2): 140-3, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17123807

ABSTRACT

PRELP is a member of the small leucine-rich repeat proteoglycan family that is abundantly expressed in many cartilages compared to other connective tissues. To study the consequence of PRELP overexpression in tissues where it is normally expressed at low abundance, transgenic mice were generated in which the human PRELP transgene was placed under control of the CMV promoter. A connective tissue phenotype was observed in the skin, where the organization of collagen fibrils in the dermis was perturbed and the thickness of the hypodermal fat layer was diminished.


Subject(s)
Collagen/metabolism , Dermis/cytology , Extracellular Matrix Proteins/metabolism , Gene Expression , Glycoproteins/metabolism , Skin/metabolism , Adipose Tissue/metabolism , Animals , Collagen/physiology , Collagen/ultrastructure , DNA Primers , Dermis/metabolism , Humans , Mice , Mice, Transgenic , Microscopy, Electron , Plasmids/genetics , Skin/ultrastructure
11.
Eur Cell Mater ; 12: 92-101, 2006 Nov 30.
Article in English | MEDLINE | ID: mdl-17136680

ABSTRACT

Cartilage contains a variety of proteoglycans that are essential for its normal function. These include aggrecan, decorin, biglycan, fibromodulin and lumican. Each proteoglycan serves several functions that are determined by both its core protein and its glycosaminoglycan chains. This review discusses the structure/function relationships of the cartilage proteoglycans, and the manner in which perturbations in proteoglycan structure or abundance can adversely affect tissue function.


Subject(s)
Cartilage/chemistry , Proteoglycans/chemistry , Aggrecans/chemistry , Amino Acid Sequence , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation
12.
Osteoarthritis Cartilage ; 14(11): 1147-54, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16806997

ABSTRACT

OBJECTIVE: Aggrecanase-1 [a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-4] and aggrecanase-2 (ADAMTS-5) have been named for their ability to degrade the proteoglycan aggrecan. While this may be the preferred substrate for these enzymes, they are also able to degrade other proteins. The aim of this work was to determine whether the aggrecanases could degrade biglycan and decorin. METHODS: Biglycan, decorin and aggrecan were purified from human and bovine cartilage and subjected to degradation by recombinant aggrecanase-1 or aggrecanase-2. In vitro degradation was assessed by sodium dodecyl sulfate/polyacrylamide gel electrophoresis (SDS/PAGE) and immunoblotting, and the cleavage site in biglycan was determined by N-terminal amino acid sequencing. SDS/PAGE and immunoblotting were also used to assess in situ degradation in both normal and arthritic human articular cartilage. RESULTS: Both aggrecanase-1 and aggrecanase-2 are able to cleave bovine and human biglycan at a site within their central leucine-rich repeat regions. Cleavage occurs at an asparagine-cysteine bond within the fifth leucine-rich repeat. In contrast, the closely related proteoglycan decorin is not a substrate for the aggrecanases. Analysis of human articular cartilage from osteoarthritic (OA) and rheumatoid arthritic (RA) joints showed that a biglycan degradation product of equivalent size is present in the extracellular matrix. No equivalent degradation product was, however, detectable in normal adult human articular cartilage. CONCLUSION: Biglycan, which is structurally unrelated to aggrecan, can act as a substrate for aggrecanase-1 and aggrecanase-2, and these proteinases may account for at least part of the biglycan degradation that is present in arthritic cartilage.


Subject(s)
Endopeptidases/metabolism , Extracellular Matrix Proteins/metabolism , Proteoglycans/metabolism , ADAM Proteins/metabolism , ADAMTS4 Protein , ADAMTS5 Protein , Adolescent , Adult , Aged , Aggrecans/metabolism , Animals , Arthritis, Rheumatoid/metabolism , Biglycan , Cartilage/metabolism , Cartilage, Articular/metabolism , Cattle , Child , Child, Preschool , Decorin , Electrophoresis, Polyacrylamide Gel/methods , Humans , Infant , Middle Aged , Osteoarthritis/metabolism , Procollagen N-Endopeptidase/metabolism , Protein Denaturation
13.
Clin Exp Allergy ; 35(10): 1361-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16238797

ABSTRACT

BACKGROUND: Inflammatory and structural changes of the airway mucosa are chronic features of asthma. The mechanisms underlying these changes and their modulation by steroid prophylaxis have not been clarified. OBJECTIVE: We postulated that asymptomatic ongoing allergen exposure could drive airway inflammation as well as changes in the extracellular matrix (ECM), and that inhaled steroids could prevent this. METHODS: Therefore, we exposed patients with mild asthma to 2 weeks of repeated low-dose allergen, with concomitant inhaled steroid or placebo treatment. Bronchial biopsies, which were taken before and after this exposure, were stained and digitally analysed. The ECM proteins in asthmatics were also compared with a normal control group. RESULTS: Low-dose allergen exposure alone resulted in a significant increase of bronchial epithelial macrophages. Despite ongoing allergen exposure, inhaled steroids reduced the numbers of mucosal eosinophils, neutrophils and T lymphocytes. At baseline, the mean density of the proteoglycans (PGS) biglycan and decorin were, respectively, higher and lower in the bronchial mucosa of asthmatics as compared with normal controls. Steroid treatment, during allergen exposure, increased the mean density of the PGS biglycan and versican. CONCLUSION: We conclude that chronic allergen exposure induces inflammatory changes in the bronchial mucosa. Despite ongoing allergen exposure, steroid treatment decreases mucosal inflammatory cells while altering PG density. The latter observation highlights the need to examine steroid-induced changes closely in the airway structure in patients with asthma.


Subject(s)
Allergens/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Asthma/drug therapy , Bronchi/pathology , Glucocorticoids/therapeutic use , Administration, Inhalation , Allergens/adverse effects , Asthma/etiology , Asthma/immunology , Asthma/pathology , Biopsy , Bronchoscopy , Budesonide/therapeutic use , Double-Blind Method , Extracellular Matrix/metabolism , Fibronectins/analysis , Goblet Cells/pathology , Humans , Inhalation Exposure/adverse effects , Mucous Membrane/immunology , Mucous Membrane/metabolism , Proteoglycans/analysis , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology
14.
Eur Cell Mater ; 5: 41-7; discussion 47, 2003 Jun 30.
Article in English | MEDLINE | ID: mdl-14562271

ABSTRACT

Osteogenesis imperfecta is a heritable disorder of bone formation resulting in low bone mass and a propensity to fracture. It exhibits a broad range of clinical severity, ranging from multiple fracturing in utero and perinatal death to normal adult stature and a low fracture incidence. The disorder is currently classified into seven types based on differences in clinical presentation and bone architecture. Mutation in one of the type I collagen genes is commonly associated with osteogenesis imperfecta, but is not a prerequisite for the diagnosis. Indeed, the newer forms of osteogenesis imperfecta (types V, VI and VII) are not associated with type I collagen gene defects. Amongst the type I collagen gene mutations that can occur, missense base substitutions involving glycine codons in the exons encoding the central triple-helix forming domain predominate. Such mutations can occur in all the classical forms of osteogenesis imperfecta (types I-IV), but genotype/phenotype correlations are complex and often unpredictable. Treatment of osteogenesis imperfecta by bisphosphonate therapy can improve bone mass in all types of the disorder, and while not being a cure for the disorder does improve the quality of life of the patient.


Subject(s)
Genetic Variation/genetics , Osteogenesis Imperfecta/diagnosis , Osteogenesis Imperfecta/genetics , Humans , Mutation, Missense , Osteogenesis Imperfecta/classification
15.
Osteoarthritis Cartilage ; 11(3): 167-76, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12623288

ABSTRACT

OBJECTIVE: Small proteoglycans (PGs) may accumulate in late stage osteoarthritis even as aggrecan is lost. It is not clear what role transforming growth factor (TGF) beta has in this accumulation. Our goal was to investigate the ability of TGF beta 1 to modulate the synthesis and accumulation of decorin, biglycan, and fibromodulin in cartilage explants cultured under conditions in which aggrecan synthesis remains relatively constant. DESIGN: Articular cartilage was cultured in the presence or absence of 4 ng/ml TGF beta 1 for up to 16 days. Material extracted from cartilage was assayed for 35SO(4)-large and small PGs and for total endogenous decorin, biglycan and fibromodulin. RESULTS: The synthesis of 35SO(4)-small PGs increased during the 16 days in culture in response to TGF beta 1, but declined in control cultures. The difference in 35SO(4)-decorin between TGF beta 1 and control samples reached nine-fold after 16 days, while the difference in total endogenous decorin was less than 1.5-fold. 35SO(4)-decorin, which was present in TGF beta 1-treated cultures had an identical core protein, but a longer glycosaminoglycan chain than that of decorin in control cultures. No significant differences in endogenous biglycan were detected, but accumulation of fibromodulin in TGF beta 1 explants exceeded fibromodulin in controls, on average, by 3.8-fold. Fibromodulin was present in cartilage in both keratan sulfate- and non-sulfated oligosaccharide-substituted forms. CONCLUSIONS: The accumulation of each of the three small PGs was affected to a different extent in response to TGF beta 1. Of the three, fibromodulin content was most rapidly augmented in response to TGF beta 1.


Subject(s)
Carrier Proteins/metabolism , Cartilage, Articular/drug effects , Extracellular Matrix Proteins , Osteoarthritis/metabolism , Proteoglycans/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Autoradiography , Biglycan , Blotting, Western/methods , Carrier Proteins/analysis , Carrier Proteins/biosynthesis , Cartilage, Articular/metabolism , Cells, Cultured , Decorin , Dogs , Electrophoresis, Polyacrylamide Gel/methods , Fibromodulin , Glycosylation , Luminescent Measurements , Proteoglycans/analysis , Proteoglycans/biosynthesis
16.
Biochem Soc Trans ; 30(Pt 6): 869-74, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12440935

ABSTRACT

The ability of the nucleus pulposus of the intervertebral disc to resist compressive loads is due to its high content of the proteoglycan aggrecan. Degeneration of the intervertebral disc is preceded and accompanied by a loss of aggrecan due to proteolysis. Biological repair of intervertebral disc degeneration should strive to restore aggrecan content to its optimal functional level. One approach to such repair is to supplement the degenerate nucleus with cells that are capable of aggrecan synthesis. Such cells can be supported in a biomolecular scaffold, but it is essential that the scaffold is compatible with high aggrecan retention if a functional tissue is to be attained.


Subject(s)
Aging , Extracellular Matrix Proteins , Intervertebral Disc/metabolism , Intervertebral Disc/physiology , Proteoglycans/physiology , Aggrecans , Animals , Humans , Lectins, C-Type , Polymorphism, Genetic , Proteoglycans/chemistry , Proteoglycans/genetics
17.
Eur Spine J ; 11 Suppl 2: S215-20, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12384747

ABSTRACT

The intervertebral disc unites the vertebrae in the spine, providing the flexibility required for bending and twisting and resisting the compression inflicted by gravity when in an upright posture. The discs have a complex structure, with the outer annulus fibrosus having lamellae of organized collagen fibrils and the inner nucleus pulposus having a more random collagen organization and an abundance of aggregating proteoglycans. This composite nature endows the disc with both the tension-resisting properties of a ligament and the compression-resisting properties of articular cartilage. Unfortunately, disc structure and function does not remain optimal throughout life, but undergoes progressive degeneration, commencing in the young adult, and is particularly evident in the nucleus pulposus. With time, disc degeneration may result in clinical symptoms, such as low back pain, and require medical intervention. Such treatment may involve removal of the offending disc by surgery rather than its repair, which would be the preferred course of action. In the near future, current bioengineering techniques may offer the possibility of repairing the damaged disc, if an engineered tissue with the appropriate functional properties can be generated to augment the ailing disc. In this report, we summarized our recent results, in which disc cells were implanted into a scaffold of collagen and hyaluronan, or entrapped into a chitosan gel, and growth factors were used to modulate matrix synthesis in an attempt to produce a tissue with a similar molecular composition to native nucleus pulposus tissue.


Subject(s)
Cell Transplantation/methods , Chitin/analogs & derivatives , Intervertebral Disc Displacement/therapy , Chitosan , Collagen , Humans , Hyaluronic Acid
18.
Bone ; 31(1): 12-8, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12110406

ABSTRACT

Osteogenesis imperfecta (OI) is a heritable disease of bone with low bone mass and bone fragility. The disease is generally classified into four types based on clinical features and disease severity, although recently fifth and sixth forms have also been reported. Most forms of OI are autosomal dominant. Rarely, autosomal recessive disease has been described. We report the clinical, radiological, and histological features of four children (age 3.9-8.6 years at last follow-up; all girls) and four adults (age 28-33 years; two women) with a novel form of autosomal recessive OI living in an isolated First Nations community in northern Quebec. In keeping with the established numeric classification for OI forms, we have called this form of the disease OI type VII. The phenotype is moderate to severe, characterized by fractures at birth, bluish sclerae, early deformity of the lower extremities, coxa vara, and osteopenia. Rhizomelia is a prominent clinical feature. Histomorphometric analyses of iliac crest bone samples revealed findings similar to OI type I, with decreased cortical width and trabecular number, increased bone turnover, and preservation of the birefringent pattern of lamellar bone. The disease has subsequently been localized to chromosome 3p22-24.1, which is outside the loci for type I collagen genes. The underlying genetic basis for the disease remains to be determined.


Subject(s)
Collagen Type I , Genes, Recessive , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/pathology , Bone Density/genetics , Child , Child, Preschool , Collagen/genetics , Collagen Type I, alpha 1 Chain , Female , Humans , Male , Osteogenesis Imperfecta/diagnostic imaging , Pedigree , Radiography
19.
Bone ; 31(1): 19-25, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12110407

ABSTRACT

We have identified a novel form of autosomal recessive osteogenesis imperfecta (OI) in a small First Nations community from northern Quebec. Mutation screening of the COL1A1/COL1A2 genes revealed no detectable mutations, and type I collagen protein analyses were also normal. By linkage analysis, we mapped this unique autosomal recessive variant of osteogenesis imperfecta to chromosome 3p22-24.1. Based on the assumption of a founder effect, genome-wide screening was performed on a DNA sample pooled from seven affected individuals. Familial as well as historical recombinations identified within an extended haplotype of 19 markers localized the disease between markers D3S2324 and D3S1561, separated by <5 cM. Based on chromosomal localization to 3p22-24.1, the transforming growth factor-beta receptor 2 gene and the parathyroid hormone/parathyroid hormone-related peptide receptor were tested, but were excluded as being associated with the phenotype. This study excludes type I collagen mutations in the pathogenesis of the disease and assigns this form of OI to a locus other than the ones containing the type I collagen genes.


Subject(s)
Chromosome Mapping/methods , Chromosomes, Human, Pair 3/genetics , Osteogenesis Imperfecta/genetics , Female , Humans , Male , Pedigree
20.
Matrix Biol ; 20(8): 555-64, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11731272

ABSTRACT

The cDNA sequence of the murine proline/arginine-rich end leucine-rich repeat protein (PRELP) gene was cloned by PCR-based techniques. The gene encodes a protein of 378 amino acids, which is four amino acid residues shorter than its human counterpart. This difference resides mainly in the amino terminal region of the mature protein, which is five amino acids shorter in the mouse than the human and has a lower arginine content. The remainder of the protein, including the structure of the leucine-rich repeats, the potential sites for N-linked glycosylation, and the disulfide-bonded domains are well conserved between species. In common with humans, the murine gene possesses three exons, with the translation initiation codon residing in exon 2 and the termination codon in exon 3. Exons 1 and 2 are separated by an intron of approximately 6.7 kbp, whereas exons 2 and 3 are separated by an intron of approximately 1.7 kbp. Western blot analysis of mouse cartilage extracts indicates that PRELP exists as a glycoprotein of approximately 55 kDa, as in human cartilage. Immunohistochemical and in situ hybridization analysis reveal that PRELP is expressed in cartilage throughout both fetal development and post-natal life, in contrast to the human where expression in cartilage is not apparent prior to birth. Northern blot analysis indicates that PRELP mRNA is also expressed in the developing embryo prior to skeletogenesis. The promoter region of the mouse PRELP gene possesses no TATA box in its proximal region, in common with humans, and shows differences in the conservation of elements known to be involved in regulating expression of the human PRELP gene.


Subject(s)
Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression , Gene Expression Regulation, Developmental , Glycoproteins/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Species Specificity , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...