Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Placenta ; 76: 6-9, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30803713

ABSTRACT

Placental mediated fetal growth restriction (FGR) is a leading cause of perinatal morbidity and mortality. Heparan sulphate proteoglycans (HSPG) are highly expressed in placentae and regulate haemostasis. We hypothesise that altered expression of HSPGs, glypicans (GPC) may contribute to the development of FGR and small-for-gestational-age (SGA). GPC expression was determined in first-trimester chorionic villous samples collected from women with later SGA pregnancies and in placentae from third-trimester FGR and gestation-matched uncomplicated pregnancies. The expression of both GPC1 and GPC3 were significantly reduced in first-trimester SGA as well as in the third-trimester FGR placentae compared to controls. This is the first study to report a relationship between altered placental GPC expression and subsequent development of SGA/FGR.


Subject(s)
Fetal Growth Retardation/metabolism , Glypicans/metabolism , Placenta/metabolism , Adult , Case-Control Studies , Female , Humans , Infant, Newborn , Infant, Small for Gestational Age , Pregnancy , Pregnancy Trimester, First/metabolism , Pregnancy Trimester, Third/metabolism
2.
Biomater Sci ; 6(5): 947-957, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29560990

ABSTRACT

The development of bioscaffolds that incorporate chondroitin sulphate (CS) and their applications with progenitor and stem cells in cartilage, bone, cornea, skin, and neural repair are reviewed. CS is a heterogeneous structure due to the organisation of multiple CS disaccharide sulphation motifs, giving rise to a vast range of CS chain structures, and hence the wide range of biological activity. The incorporation of this biological molecule represents a significant advance in bioscaffold design and performance in tissue repair strategies. The intrinsic stem-cell directive properties of CS are covered in the context of tissue development, and the differing CS disaccharide motifs, referred to as the 'glyco-code'. These structural motifs contribute to stem cell proliferation and differentiation in the scaffold environment and improve outcomes in terms of tissue repair or regeneration worthy of future research.


Subject(s)
Chondroitin Sulfates/chemistry , Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Humans , Regeneration , Stem Cells/drug effects , Stem Cells/metabolism , Tissue Scaffolds/adverse effects
3.
Eur Cell Mater ; 32: 40-57, 2016 07 05.
Article in English | MEDLINE | ID: mdl-27377666

ABSTRACT

The aim of this study was to immunolocalise type VI collagen and perlecan and determine their interactive properties in the intervertebral disc (IVD). Confocal laser scanning microscopy co-localised perlecan with type VI collagen as pericellular components of IVD cells and translamellar cross-bridges in ovine and murine IVDs. These cross-bridges were significantly less abundant in the heparin sulphate deficient Hspg2 exon 3 null mouse IVD than in wild type. This association of type VI collagen with elastic components provides clues as to its roles in conveying elastic recoil properties to annular tissues. Perlecan and type VI collagen were highly interactive in plasmon resonance studies. Pericellular colocalisation of perlecan and type VI collagen provides matrix stabilisation and cell-matrix communication which allows IVD cells to perceive and respond to perturbations in their biomechanical microenvironment. Perlecan, at the cell surface, provides an adhesive interface between the cell and its surrounding extracellular matrix. Elastic microfibrillar structures regulate tensional connective tissue development and function. The 2010 Global Burden of Disease study examined 291 disorders and identified disc degeneration and associated low back pain as the leading global musculoskeletal disorder emphasising its massive socioeconomic impact and the need for more effective treatment strategies. A greater understanding of how the IVD achieves its unique biomechanical functional properties is of great importance in the development of such therapeutic measures.


Subject(s)
Collagen Type VI/metabolism , Heparan Sulfate Proteoglycans/metabolism , Intervertebral Disc/metabolism , Amino Acid Sequence , Animals , Fibronectins/metabolism , Heparan Sulfate Proteoglycans/chemistry , Intervertebral Disc/cytology , Laminin/metabolism , Mice, Inbred C57BL , Peptides/chemistry , Peptides/metabolism , Protein Transport , Sheep , Surface Plasmon Resonance
4.
Placenta ; 35(8): 596-605, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24947404

ABSTRACT

OBJECTIVE: Fetal growth restriction (FGR) is a key cause of adverse pregnancy outcome where maternal and fetal factors are identified as contributing to this condition. Idiopathic FGR is associated with altered vascular endothelial cell functions. Decorin (DCN) has important roles in the regulation of endothelial cell functions in vascular environments. DCN expression is reduced in FGR. The objectives were to determine the functional consequences of reduced DCN in a human microvascular endothelial cell line model (HMVEC), and to determine downstream targets of DCN and their expression in primary placental microvascular endothelial cells (PLECs) from control and FGR-affected placentae. APPROACH: Short-interference RNA was used to reduce DCN expression in HMVECs and the effect on proliferation, angiogenesis and thrombin generation was determined. A Growth Factor PCR Array was used to identify downstream targets of DCN. The expression of target genes in control and FGR PLECs was performed. RESULTS: DCN reduction decreased proliferation and angiogenesis but increased thrombin generation with no effect on apoptosis. The array identified three targets of DCN: FGF17, IL18 and MSTN. Validation of target genes confirmed decreased expression of VEGFA, MMP9, EGFR1, IGFR1 and PLGF in HMVECs and PLECs from control and FGR pregnancies. CONCLUSIONS: Reduction of DCN in vascular endothelial cells leads to disrupted cell functions. The targets of DCN include genes that play important roles in angiogenesis and cellular growth. Therefore, differential expression of these may contribute to the pathogenesis of FGR and disease states in other microvascular circulations.


Subject(s)
Decorin/metabolism , Endothelial Cells/metabolism , Fetal Growth Retardation/etiology , Gene Expression Regulation , Placenta/metabolism , Apoptosis , Case-Control Studies , Cell Line , Cell Proliferation , Female , Fetal Growth Retardation/metabolism , Humans , Pregnancy , RNA, Small Interfering , Thrombin/metabolism
5.
J Agric Food Chem ; 56(2): 343-8, 2008 Jan 23.
Article in English | MEDLINE | ID: mdl-18163570

ABSTRACT

Chondroitin sulfate (CS) is a glycosaminoglycan derived from cartilage and commonly used to treat osteoarthritis, psoriasis, and other conditions. The dimethylmethylene blue (DMMB) assay has been used often to measure glycosaminoglycan levels in relatively pure samples. In this study, we verified the accuracy of the DMMB assay in measuring CS levels in unpurified extract from bovine trachea and shark cartilage, despite potential interference from salts, proteins, and DNA. We found that the glycosaminoglycan signal obtained was due to CS and not to other glycosaminoglycan species. This was confirmed using fluorophore-assisted carbohydrate electrophoresis, which also revealed that the majority of the CS was monosulfated at the C4 or C6 position. Finally, we used anion-exchange chromatography to purify the bovine extract and obtained complete recovery of the glycosaminoglycans, with no contaminating protein. The results of this study should be very useful for future purification and analysis of this common supplement.


Subject(s)
Glycosaminoglycans/analysis , Glycosaminoglycans/isolation & purification , Animals , Cartilage/chemistry , Cattle , Chondroitin Sulfates/analysis , Chromatography, Ion Exchange , Methylene Blue/analogs & derivatives , Sharks , Trachea/chemistry
6.
Cell Mol Life Sci ; 63(21): 2435-45, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16952056

ABSTRACT

Perlecan is a large multi-domain extracellular matrix proteoglycan that plays a crucial role in tissue development and organogenesis. In vertebrates, perlecan functions in a diverse range of developmental and biological processes, from the establishment of cartilage to the regulation of wound healing. How can a single molecule modulate such a wide variety of processes? We suggest that perlecan employs the same basic mechanism, based on interactions with growth factors, morphogens and matrix proteins, to regulate each of these processes and that the local extracellular environment determines the function of perlecan and consequently its downstream effects on the structure and function of the organ. We discuss this hypothesis in relation to its role in three major vertebrate developmental processes: angiogenesis, chondrogenesis and endochondral ossification.


Subject(s)
Heparan Sulfate Proteoglycans/metabolism , Animals , Blood Vessels/growth & development , Chondrogenesis/physiology , Humans , Neovascularization, Physiologic , Osteogenesis/physiology
7.
Biomaterials ; 27(28): 4856-62, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16757021

ABSTRACT

Serum protein adsorption on colloidal silica surfaces was investigated using a quartz crystal microbalance with dissipation (QCM-D) monitoring. The amount of serum proteins adsorbed on colloidal silica-coated surfaces was not significantly different from the control silica surfaces, with the exception of 21nm colloidal silica which experienced significantly less (P<0.05) fibrinogen adsorption compared with control silica. The adhesion and proliferation of human endothelial cells (C11STH) on nano-scale colloidal silica surfaces were significantly reduced compared with control silica surfaces, suggesting that the conformation of adsorbed proteins on the colloidal silica surfaces plays a role in modulating the amount of cell binding. Fibronectin is one of the main extracellular matrix proteins involved in endothelial cell attachment to biomaterial surfaces. There was reduced binding of a monoclonal anti-fibronectin antibody, that reacted specifically with the cell-binding fragment, to fibronectin-coated colloidal silica surfaces compared with control silica surfaces. This suggests that the fibronectin adsorbed on the colloidal silica-coated surfaces was conformationally changed compared with control silica reducing the availability of the cell-binding domain of fibronectin.


Subject(s)
Blood Proteins/chemistry , Cell Proliferation/drug effects , Nanostructures/chemistry , Silicon Dioxide/pharmacology , Adsorption/drug effects , Animals , Cell Adhesion/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fibronectins/chemistry , Humans , Mice , NIH 3T3 Cells , Nanotechnology , Silicon Dioxide/chemistry , Surface Properties
8.
J Dent Res ; 85(2): 122-32, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16434729

ABSTRACT

Tissue engineering, grafting procedures, regeneration, and tissue remodeling are developing therapeutic modalities with great potential medical value, but these regenerative modalities are not as effective or predictable as clinicians and patients would like. Greater understanding of growth factors, cytokines, extracellular matrix molecules, and their roles in cell-mediated healing processes have made these regenerative therapies more clinically viable and will continue advancing the fields of tissue engineering and grafting. However, millions of oral and non-oral bone-grafting procedures are performed annually, and only a small percentage yield the most desirable results. Here we review the heparan-sulfate-decorated extracellular biomolecule named perlecan and the research relating to its potential as an adjunct in bone-regenerative procedures. The review includes an overview of bone graft substitutes and biological adjuncts to bone-regenerative procedures in medicine as they apply to periodontal disease, alveolar ridge augmentation, and barrier membrane therapy. Perlecan is discussed as a potential biological adjunct in terms of growth factor sequestration and delivery, and promoting cell adhesion, proliferation, differentiation, and angiogenesis. Further, we propose delivery and application schemes for perlecan and/or its domains in bone-regenerative procedures, with particular emphasis on its heparan-sulfate-decorated domain I. The perlecan molecule, with its heparan sulfate glycosylation, may provide a multi-faceted approach for the delivery of a more comprehensive stimulus than other single potential adjuncts currently available for bone-regenerative procedures.


Subject(s)
Alveolar Bone Loss/surgery , Bone Regeneration/drug effects , Heparan Sulfate Proteoglycans/pharmacology , Animals , Bone Substitutes , Cell Adhesion/drug effects , Cell Adhesion/physiology , Growth Substances/physiology , Guided Tissue Regeneration, Periodontal , Heparan Sulfate Proteoglycans/administration & dosage , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/physiology , Humans , Protein Structure, Tertiary
9.
FEBS Lett ; 579(22): 5019-23, 2005 Sep 12.
Article in English | MEDLINE | ID: mdl-16129435

ABSTRACT

Perlecan is a multidomain proteoglycan, usually substituted with heparan sulphate (HS), and sometimes substituted with both HS and chondroitin sulphate (CS). In this paper, we describe perlecan purified from HEK-293 cells substituted with HS, CS and keratan sulphate (KS). KS substitution was confirmed by immunoreactivity with antibody 5D4, sensitivity to keratanase treatment, and fluorophore-assisted carbohydrate electrophoresis. HEK-293 perlecan failed to promote FGF-dependent cell growth in an in vitro assay. This study is the first to report perlecan containing KS, and makes perlecan one of only a very few proteoglycans substituted with three distinct types of glycosaminoglycan chains.


Subject(s)
Chondroitin Sulfate Proteoglycans/analysis , Epithelial Cells/chemistry , Heparan Sulfate Proteoglycans/analysis , Keratan Sulfate/analysis , Cell Line , Chondroitin Sulfate Proteoglycans/metabolism , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factors/metabolism , Heparan Sulfate Proteoglycans/chemistry , Heparan Sulfate Proteoglycans/isolation & purification , Heparan Sulfate Proteoglycans/metabolism , Humans , Keratan Sulfate/metabolism , Lumican
10.
J Thromb Haemost ; 2(8): 1460-6, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15304055

ABSTRACT

von Willebrand factor (VWF) freshly released from endothelial cells is normally cleaved by the ADAMTS-13 metalloprotease to prevent the direct release of these ultra-large (UL) and hyper-reactive multimers into plasma. The balance of ULVWF proteolysis may be regulated by the amount of ULVWF released and the processing capacity of ADAMTS-13. The former associates with the size of ULVWF storage pool, sensitivity of vascular endothelial cells to stimulation, and the type of agonists, whereas the latter associates with the activity of ADAMTS-13. These parameters may vary significantly among individuals. We have determined the variations of ADAMTS-13 activity in 68 normal individuals by a flow-based assay and a static assay using ULVWF strings and recombinant VWF A2 domain as substrates, respectively. We found that the levels of ADAMTS-13 activity required to cleave the platelet-decorated ULVWF strings under flow is significantly higher than that of static assays. Normal plasma diluted to 25% significantly reduced its ability to cleave ULVWF strings under flow, whereas 2% plasma retained 48% enzyme activity in static assay. ADAMTS-13 activity varied from 33 to 100% among individuals and the variations were greater at shorter incubations of plasma with the substrate. Furthermore, the production of ULVWF from endothelial cells also varied among individuals. These results suggest that the commonly used static assays may underestimate the ADAMTS-13 activity required to cleave newly released ULVWF. They also demonstrated that the proteolysis of ULVWF may vary significantly among individuals, potentially contributing to the individual's vulnerability to thrombosis so that measurement of ADAMTS-13 may serve as a marker for TTP and other thrombotic diseases.


Subject(s)
Endothelium, Vascular/metabolism , von Willebrand Factor/metabolism , ADAM Proteins , ADAMTS13 Protein , Adult , Blood Platelets/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Metalloendopeptidases/metabolism , Middle Aged , Proteins/chemistry , Recombinant Proteins/chemistry , Reference Values , Risk Factors , Thrombosis , Time Factors
11.
J Thromb Haemost ; 2(3): 485-91, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15009467

ABSTRACT

The metalloprotease ADAMTS-13 cleaves von Willebrand factor (VWF) at the Y842/M843 peptide bond located in the A2 domain. Measurement of ADAMTS-13 activity is a clinical utility for thrombotic diseases, but the current assays used for diagnostic and clinical research are non-physiological and time consuming. We have expressed in bacteria a recombinant VWF-A2 peptide (aa 718-905) that contains both a 6xHis tag at the N-terminal end and a Tag-100 epitope at the C-terminal end. Diluted plasma was mixed with the VWF-A2 peptide and digestion was allowed to proceed in a Ni2+-coated microtiter well plate for 2 h. The immobilized Ni2+ captures the VWF-A2 peptide by its 6xHis tag and cleavage of the A2 peptide is measured by the removal of the C-terminus fragment of the A2 peptide that contains the Tag-100. The cleavage activity for this assay was defined by the low detection of A2 peptide containing the Tag-100 epitope by the antiTag-100 monoclonal antibody. The assay was completed in <5 h. We then used the assay to analyze ADAMTS-13 activity in plasma from 39 healthy donors and 16 samples from patients diagnosed as thrombotic thrombocytopenic purpura. The average of enzyme activity +/- SEM for normal plasmas diluted 1 : 50 was 40 +/- 4.2% while the value obtained for the patients was 2.4 +/- 0.7%. These results were validated by a traditional long incubation assay (24 h). Our assay provides significant advantages over currently used assays because it is quicker, reproducible, cost effective and measures ADAMTS-13 activity under physiological and non-denaturing conditions. This assay is clinically useful and significant in measuring ADAMTS-13 activity in plasma.


Subject(s)
Metalloendopeptidases/metabolism , von Willebrand Factor/metabolism , ADAM Proteins , ADAMTS13 Protein , Binding Sites , Enzyme-Linked Immunosorbent Assay/methods , Humans , Kinetics , Recombinant Proteins/metabolism , Substrate Specificity , von Willebrand Factor/chemistry
12.
Proteomics ; 1(12): 1534-41, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11747213

ABSTRACT

Three cellular sources of perlecan were examined in this study, namely human umbilical arterial endothelial cells (HUAEC), a transformed human umbilical venous endothelial cell line (C 1 1 STH) and a human colon carcinoma cell line (WiDr). Perlecans were immunopurified from conditioned media of the above cells and the purity of the perlecan preparations was examined by composite agarose polyacrylamide gel electrophoresis (CAPAGE) and semi-dry immunoblotting with monoclonal antibodies directed to either the perlecan core protein (mAb A76) or heparan sulphate (HS) side-chain (mAb10E4). The ability of each perlecan species to bind fibroblast growth factor-l (FGF-1) was examined using a biosensor (BIAcore). The bioactivity of perlecan FGF-1 interactions was also analysed using BaF3 cells transfected with fibroblast growth factor receptors FGFR1b and 1c. CAPAGE demonstrated subtle differences between the perlecans, indicating they had differing charge to mass ratios with C 11 STH perlecan being slightly more mobile in CAPAGE than the HUAEC and WiDr sample. BIAcore biosensor analysis demonstrated distinct differences in the ability of perlecan preparations to bind FGF-1; HUAEC and C 11 STH perlecan showed similar high binding responses as compared to WiDr perlecan, which bound FGF-1 very poorly. Binding of FGF-1 to endothelial perlecans was shown to be HS-dependent. Interestingly, HUAEC perlecan stimulated the growth of FGFR1b and FGFR1c expressing cells in the presence of FGF-1 comparable to heparin, whereas C 11 STH perlecan showed only very limited stimulation of FGFR 1b cells and was incapable of stimulating FGFR1c cells. WiDr perlecan exhibited no stimulation of growth in either cell line. Collectively the data presented herein indicate that. different cell types express perlecans which vary in the growth factor binding capabilities, which may suggest differences in their HS chain substructure. This may represent a subtle mechanism whereby cells can modulate the responsiveness of perlecan to a range of biologically important ligands and thus in a broader context may have important implications for cell signalling.


Subject(s)
Biosensing Techniques , Electrophoresis, Agar Gel/methods , Electrophoresis, Polyacrylamide Gel/methods , Heparan Sulfate Proteoglycans/physiology , Cell Line , Heparan Sulfate Proteoglycans/isolation & purification , Heparan Sulfate Proteoglycans/metabolism , Humans , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/metabolism
14.
J Immunol Methods ; 247(1-2): 217-24, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11150552

ABSTRACT

Serum is a common component of most in vitro cell culture media, particularly of primary cells. Studies of cellular responses to particular adhesion molecules or growth factors are often confounded by the presence of these molecules in the serum supplement. We describe a combined affinity protocol for removing vitronectin and fibronectin from serum. This protocol can also be used to purify these molecules. We also describe the removal of growth-promoting elements using heparin-Sepharose. As vitronectin and fibronectin each bind to heparin, these molecules are removed first and the heparin-Sepharose depletion occurs last in the sequence. This protocol provides a detailed step-by-step guide to achieve quantitative depletion of serum in an optimised format, with additional information on pitfalls and problems. It should be of use to people who wish to accurately determine the relationship between cells, extracellular matrix molecules and growth factors.


Subject(s)
Fibronectins/isolation & purification , Vitronectin/isolation & purification , Animals , Cattle , Cell Adhesion Molecules/blood , Cell Adhesion Molecules/isolation & purification , Fibronectins/blood , Growth Substances/blood , Growth Substances/isolation & purification , Vitronectin/blood
15.
J Biol Chem ; 275(10): 7095-100, 2000 Mar 10.
Article in English | MEDLINE | ID: mdl-10702276

ABSTRACT

Perlecan is a multifaceted heparan sulfate proteoglycan that is expressed not only as an intrinsic constituent of basement membranes but also as a cell-surface and pericellular proteoglycan. Perlecan functions as a ligand reservoir for various growth factors that become stabilized against misfolding or proteolysis and acts as a co-receptor for basic fibroblast growth factor by augmenting high affinity binding and receptor activation. These biological properties are mediated by the heparan sulfate moiety. Rather little is known about the protein core's mediation of functions. We have recently discovered that fibroblast growth factor-7 (FGF7) binds to perlecan protein core and that exogenous perlecan efficiently reconstitutes FGF7 mitogenic activity in perlecan-deficient cells. In this report we examined the specific binding of FGF7 to various domains and subdomains of perlecan protein core. Using several experimental approaches including overlay protein assays, radioligand binding experiments, and the yeast two-hybrid system, we demonstrate that FGF7 binds specifically to the N-terminal half of domain III and to a lesser extent to domain V, with affinity constants in the range of 60 nM. Thus, perlecan protein core should be considered a novel biological ligand for FGF7, an interaction that could influence cancer growth and tissue remodeling.


Subject(s)
Fibroblast Growth Factors , Growth Substances/metabolism , Heparan Sulfate Proteoglycans , Heparitin Sulfate/metabolism , Proteoglycans/metabolism , Cells, Cultured , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Humans , Recombinant Proteins/metabolism
16.
Matrix Biol ; 18(2): 163-78, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10372557

ABSTRACT

Perlecan, a major heparan sulfate proteoglycan of vascularized tissues, was immunopurified from media conditioned by human endothelial cells of both arterial and venous origin. The heparan sulfate moiety of perlecan from cultured arterial cells differed in amount and/or composition from that produced by a transformed cell line of venous origin. Both forms of perlecan bound basic fibroblast growth factor with Kd approximately 70 nM. In ELISA experiments, perlecan and its protein core bound to various extracellular matrix components in a manner that was strongly influenced by the format of the assay. Human vascular smooth muscle cells and human endothelial cells adhered to perlecan-coated surfaces, and both cell types adhered better to the venous cell-derived than to the arterial cell-derived perlecan. Removal of the heparan sulfate chains abolished this difference and increased the ability of both types of perlecan to adhere vascular cells. Denaturation of perlecan and its protein core also rendered each of them more adhesive, indicating the presence of conformation-independent adhesion determinants in the polypeptide sequence. Their location was investigated using recombinant perlecan domains. Overall, our results represent the first demonstration of human perlecan acting as an adhesive molecule for human vascular cells and suggest that it may play a role in vascular wound healing.


Subject(s)
Endothelium, Vascular/chemistry , Heparan Sulfate Proteoglycans , Heparitin Sulfate/metabolism , Proteoglycans/metabolism , Animals , Binding Sites , Cattle , Cell Adhesion , Cell Line, Transformed , Cells, Cultured , Collagen/metabolism , Endothelium, Vascular/cytology , Fibroblast Growth Factor 2/metabolism , Fibronectins/metabolism , Heparitin Sulfate/immunology , Heparitin Sulfate/isolation & purification , Humans , Mice , Muscle, Smooth, Vascular/cytology , Proteoglycans/immunology , Proteoglycans/isolation & purification , Rats , Swine
17.
Biochem Biophys Res Commun ; 256(3): 542-8, 1999 Mar 24.
Article in English | MEDLINE | ID: mdl-10080934

ABSTRACT

Recombinant forms of human perlecan domain I were secreted as proteoglycans by stably transfected human 293 cells. A recombinant domain I-only proteoglycan spanned the 95- to 265-kDa region in SDS-PAGE and appeared to be 160 kDa in denaturing gel filtration. Its glycosaminoglycan (GAG) content was approximately 67% heparan sulfate, and its average GAG chain size of 20 kDa suggested that the true molecular mass of the proteoglycan was 90 kDa. Domain I with enhanced green fluorescent protein fused to its C-terminus had an apparent molecular mass of 210-220 kDa and contained approximately 100% heparan sulfate. Its average GAG chain size (also 20 kDa) suggested a true molecular mass of 117 kDa for this proteoglycan. Its sulfate content of 53-77 mol SO2-4 per mole of protein indicated the presence of one sulfate group per 4-7 GAG sugar residues.


Subject(s)
Glycosaminoglycans/analysis , Heparan Sulfate Proteoglycans , Heparin/analogs & derivatives , Heparitin Sulfate/biosynthesis , Proteoglycans/biosynthesis , Amino Acid Sequence , Blotting, Western , Cell Line , Chromatography, Gel , Culture Media, Conditioned , Electrophoresis, Polyacrylamide Gel , Gene Expression , Glycosaminoglycans/biosynthesis , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Heparin/analysis , Heparin/biosynthesis , Heparin/chemistry , Heparin/genetics , Heparin/metabolism , Heparitin Sulfate/chemistry , Heparitin Sulfate/genetics , Heparitin Sulfate/metabolism , Humans , Molecular Weight , Polysaccharide-Lyases/metabolism , Protein Biosynthesis , Protein Sorting Signals/genetics , Protein Sorting Signals/metabolism , Proteoglycans/chemistry , Proteoglycans/genetics , Proteoglycans/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sulfates/analysis , Transcription, Genetic , Transfection
18.
Atherosclerosis ; 141(1): 141-52, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9863547

ABSTRACT

Endothelial cells recovering from damage due to disease or surgical procedures come into close contact with extracellular matrix (ECM) secreted by intimal vascular smooth muscle cells (VSMCs). We have investigated these relationships using human umbilical artery endothelial cells (HUAECs) and human mammary artery VSMC in vitro. HUAEC adhesion and proliferation were significantly lower on ECM secreted by VSMC compared with HUAEC ECM or surface-coated fibronectin. Characterisation of the ECM of both cell types with monoclonal antibodies showed that the ECM secreted by VSMC contained significantly more elastin, chondroitin sulphate and collagen types I, III and V than that from HUAECs. HUAECs adhered poorly to collagen type V coated on plastic and not at all to elastin. When these proteins were co-coated with fibronectin, elastin did not inhibit migration or proliferation compared to the response on fibronectin but collagen type V significantly inhibited both. Treatment of VSMC ECM with enzymes which selectively depleted the matrix of collagen types I, III and IV, or chondroitin sulphate, had no effect on HUAEC responses to the ECM, suggesting that these molecules did not contribute to the inhibition of HUAECs. Treatment of VSMC ECM with a mixture of collagenases, selectively depleted the matrix of collagen type V, as well as types I, III and IV. Such depleted ECMs supported increased proliferation of HUAECs compared to buffer controls. Overall these results suggest that collagen V secreted into the ECM of VSMC may inhibit the recovery of adjacent endothelium.


Subject(s)
Collagen/physiology , Endothelium, Vascular/physiology , Extracellular Matrix/physiology , Muscle, Smooth, Vascular/metabolism , Cell Adhesion/physiology , Cell Division/physiology , Cells, Cultured , Chondroitinases and Chondroitin Lyases/pharmacology , Collagen/analysis , Collagenases/pharmacology , Endothelium, Vascular/cytology , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Humans , Pancreatic Elastase/pharmacology , Umbilical Arteries
19.
J Vasc Res ; 35(6): 449-60, 1998.
Article in English | MEDLINE | ID: mdl-9858870

ABSTRACT

The proliferation of vascular smooth muscle cells (VSMC) plays a significant part in both the developing atherosclerotic lesion and in restenosis. Heparin has been widely reported to inhibit the growth of VSMC in culture and intimal VSMC in some animal models of vascular hyperplasia. Clinical trials with heparin, however, have failed to inhibit restenosis following angioplasty. Bovine serum is normally used as a growth supplement in in vitro VSMC growth assays. We have compared the effects of human serum with those of bovine serum on the cellular response to heparin in human VSMC culture. While heparin inhibited the proliferation of human VSMC in the presence of bovine serum, it was totally ineffective in the presence of human serum. These observations were consistent over a wide range of serum and VSMC samples. Experiments utilizing neutralizing antibodies to a number of growth factors showed that cells in either serum were similarly dependent on platelet-derived growth factor for proliferation. In contrast, proliferation in the presence of bovine serum was shown to be dependent on extracellular basic fibroblast growth factor, whereas that in human serum was not. Direct binding of [3H]-heparin to VSMC was significantly reduced in the presence of human serum compared with bovine serum, and the former contained twice the concentration of heparin-binding factors of the latter. Removal of heparin-binding factors from either serum type significantly reduced the proliferation potential. Fractionation of heparin-binding factors from human serum showed that the major growth-promoting activity, together with heparin resistance, was contained within a fraction excluded by a 100,000 molecular weight membrane. We conclude that the mechanism of resistance to heparin in human serum is likely to be due to a combination of differential growth factor binding and interference with heparin interaction with cellular receptors by a high molecular weight heparin-binding factor. This phenomenon may significantly contribute to the lack of success of heparin as an antirestenotic agent in clinical trials.


Subject(s)
Heparin/pharmacology , Muscle, Smooth, Vascular/drug effects , Animals , Antibodies , Becaplermin , Blood , Cattle , Cell Division/drug effects , Cells, Cultured , Culture Media , Heparin/blood , Humans , Muscle, Smooth, Vascular/cytology , Platelet-Derived Growth Factor/immunology , Proto-Oncogene Proteins c-sis
20.
J Vasc Surg ; 28(4): 676-86, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9786264

ABSTRACT

PURPOSE: In vivo and in vitro observations strongly suggest that marked differences exist in the phenotype, growth, and matrix-producing capabilities of distinct smooth muscle cell subpopulations. An earlier study from our laboratory showed differences in matrix metalloproteinase expression patterns in cultures of medial smooth muscle cells from tissue affected by abdominal aortic aneurysm (AAA) or atherosclerotic occlusive disease and from normal arterial tissue. In this study we were interested in ascertaining whether smooth muscle cells from the same sample groups also synthesized different proteoglycan profiles that correlated with vascular disease. METHODS: Proteoglycans from smooth muscle cell monolayer cultures from tissue affected by AAA or atherosclerotic occlusive disease and from normal arterial tissue were examined by means of immunoblotting and affinity-blotting composite agarose polyacrylamide gel electrophoresis (CAPAGE) and sodium dodecyl sulphate PAGE. Enzyme-linked immunosorbent assay (ELISA) was used to quantitate perlecan levels in smooth muscle cell monolayer media samples. RESULTS: Versican, perlecan, and biglycan levels were significantly elevated in AAA smooth muscle cell cultures. Two populations of smooth muscle cell versican were identified by means of CAPAGE-immunoblotting and by means of a novel affinity-blotting technique with biotinylated hyaluronan. A small keratan sulfate-substituted proteoglycan was present in similar levels in all smooth muscle cell cultures. This proteoglycan had a free core protein of about 55 kd after keratanase digestion and had a relatively high charge-to-mass ratio, as was evident from its electrophoretic mobility in CAPAGE; this proteoglycan was tentatively identified as keratocan. Immunoblotting with monoclonal antibodies 3-G-10 (anti-delta heparan sulfate, heparan sulfate stubs generated by heparitinase treatment) and 10-E-4 (anti-native heparan sulfate chains) helped identify several smooth muscle cell heparan sulfate-substituted proteoglycans. Elevated levels of intact and processed perlecan core protein were identified in AAA cultures by means of immunoblotting with a monoclonal antibody to perlecan core protein (A76). ELISA measurements confirmed that perlecan levels were significantly higher in AAA smooth muscle cell cultures compared with the normal arterial tissue and tissue affected by atherosclerotic occlusive disease. CONCLUSIONS: Because heparan sulfate proteoglycans can bind growth factors, their elevated synthesis by AAA smooth muscle cells in combination with an increased expression of matrix metalloproteinases may at least partly explain the differential proliferative capacity of the AAA smooth muscle cells examined and may govern the pattern of abnormal cellular proliferation and matrix protein synthesis observed in the pathogenesis of vascular disease.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Heparan Sulfate Proteoglycans , Muscle, Smooth, Vascular/metabolism , Proteoglycans/biosynthesis , Aged , Aged, 80 and over , Aorta, Abdominal/metabolism , Arteriosclerosis/metabolism , Biglycan , Cells, Cultured , Chondroitin Sulfate Proteoglycans/analysis , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix Proteins , Heparitin Sulfate/analysis , Humans , Immunoblotting , Lectins, C-Type , Middle Aged , Proteoglycans/analysis , Versicans
SELECTION OF CITATIONS
SEARCH DETAIL
...