Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Ultrasound Obstet Gynecol ; 61(6): 710-718, 2023 06.
Article in English | MEDLINE | ID: mdl-36647616

ABSTRACT

OBJECTIVES: Fetal endoscopic tracheal occlusion (FETO) improves neonatal survival of fetuses with congenital diaphragmatic hernia (CDH). However, FETO also increases the risk of preterm prelabor rupture of membranes (PPROM) and preterm delivery (PTD), as fetal membrane defects after fetoscopy do not heal. To solve this issue, an advanced sealing plug for closing the membrane defect is being developed. Using early-stage health economic modeling, we aimed to estimate the potential value of this innovative plug in terms of costs and effects, and to determine the properties required for it to become cost-effective. METHODS: Early-stage health economic modeling was applied to the case of performing FETO in women with a singleton pregnancy whose fetus is diagnosed prenatally with CDH. We simulated a cohort of patients using a state-transition model over a 45-year time horizon. In our best-case-scenario analysis, we compared the current-care strategy with the perfect-plug strategy, which reduces the risk of PPROM and PTD by 100%, to determine the maximum quality-adjusted life years (QALYs) gained and costs saved. Using threshold analysis, we determined the minimum percentage reduction in the risk of PPROM and PTD required for the plug to be considered cost-effective. The impact of model parameters on outcome was investigated using a sensitivity analysis. RESULTS: Our model indicated that a perfect-plug strategy would yield on average an additional 1.94 QALYs at a cost decrease of €2554 per patient. These values were influenced strongly by the percentage of cases with early PTD (27-34 weeks). Threshold analysis showed that, for €500 per plug, the plug strategy needs a minimum percentage reduction of 1.83% in the risk of PPROM and PTD (i.e. reduction in the risk from 47.50% to 46.63% for PPROM and from 71.50% to 70.19% for PTD) to be cost-effective. CONCLUSIONS: Our model-based approach showed clear potential of the plug strategy when applied in the context of FETO for CDH fetuses, as only a minor reduction in the risk of PPROM and PTD is needed for the plug to be cost-effective. Its value is expected to be even higher when used in conditions associated with a higher rate of early PTD. Continued investment in research and development of the plug strategy appears to provide value for money. © 2023 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.


Subject(s)
Fetal Membranes, Premature Rupture , Hernias, Diaphragmatic, Congenital , Premature Birth , Pregnancy , Infant, Newborn , Humans , Female , Hernias, Diaphragmatic, Congenital/surgery , Fetoscopy/adverse effects , Cost-Effectiveness Analysis , Fetal Membranes, Premature Rupture/etiology , Trachea
2.
Biomed Res Int ; 2018: 9873471, 2018.
Article in English | MEDLINE | ID: mdl-30228991

ABSTRACT

Glycosaminoglycans are important for cell signaling and therefore for proper embryonic development and adult homeostasis. Expressions of genes involved in proteoglycan/glycosaminoglycan (GAG) metabolism and of genes coding for growth factors known to bind GAGs were analyzed during skin development by microarray analysis and real time quantitative PCR. GAG related genes were organized in six categories based on their role in GAG homeostasis, viz. (1) production of precursor molecules, (2) production of core proteins, (3) synthesis of the linkage region, (4) polymerization, (5) modification, and (6) degradation of the GAG chain. In all categories highly dynamic up- and downregulations were observed during skin development, including differential expression of GAG modifying isoenzymes, core proteins, and growth factors. In two mice models, one overexpressing heparanase and one lacking C5 epimerase, differential expression of only few genes was observed. Data show that during skin development a highly dynamic and complex expression of GAG-associated genes occurs. This likely reflects quantitative and qualitative changes in GAGs/proteoglycans, including structural fine tuning, which may be correlated with growth factor handling.


Subject(s)
Gene Expression Regulation , Glycosaminoglycans/metabolism , Proteoglycans/metabolism , Skin/growth & development , Animals , Dermis , Female , Mice
3.
Acta Biomater ; 30: 265-276, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26555376

ABSTRACT

In skeletal muscle, the stem cell niche is important for controlling the quiescent, proliferation and differentiation states of satellite cells, which are key for skeletal muscle regeneration after wounding. It has been shown that type I collagen, often used as 3D-scaffolds for regenerative medicine purposes, impairs myoblast differentiation. This is most likely due to the absence of specific extracellular matrix proteins providing attachment sites for myoblasts and/or myotubes. In this study we investigated the differentiation capacity of primary murine myoblasts on type I collagen films either untreated or modified with elastin, laminin, type IV collagen, laminin/entactin complex, combinations thereof, and Matrigel as a positive control. Additionally, increased reactive oxygen species (ROS) and ROCK signaling might also be involved. To measure ROS levels with live-cell microscopy, fibronectin-coated glass coverslips were additionally coated with type I collagen and Matrigel onto which myoblasts were differentiated. On type I collagen-coated coverslips, myotube formation was impaired while ROS levels were increased. However, anti-oxidant treatment did not enhance myotube formation. ROCK inhibition, which generally improve cellular attachment to uncoated surfaces or type I collagen, enhanced myoblast attachment to type I collagen-coated coverslips and -films, but slightly enhanced myotube formation. Only modification of type I collagen films by Matrigel and a combination of laminin/entactin significantly improved myotube formation. Our results indicate that type I collagen scaffolds can be modified by satellite cell niche factors of which specifically laminin and entactin enhanced myotube formation. This offers a promising approach for regenerative medicine purposes to heal skeletal muscle wounds. STATEMENT OF SIGNIFICANCE: In this manuscript we show for the first time that impaired myotube formation on type I collagen scaffolds can be completely restored by modification with laminin and entactin, two extracellular proteins from the satellite cell niche. This offers a promising approach for regenerative medicine approaches to heal skeletal muscle wounds.


Subject(s)
Collagen Type I , Laminin , Membrane Glycoproteins , Membranes, Artificial , Muscle Fibers, Skeletal/metabolism , Wound Healing , Animals , Cattle , Collagen Type I/chemistry , Collagen Type I/pharmacology , Laminin/chemistry , Laminin/pharmacology , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/pharmacology , Mice , Muscle Fibers, Skeletal/pathology
4.
J Tissue Eng Regen Med ; 10(1): E34-44, 2016 Jan.
Article in English | MEDLINE | ID: mdl-23468399

ABSTRACT

Autologous skin grafts are the gold standard for the treatment of burn wounds. In a number of cases, treatment with autologous tissue is not possible and skin substitutes are used. The outcome, however, is not optimal and improvements are needed. Inspired by scarless healing in early embryonic development, we here set out a strategy for the design and construction of embryonic-like scaffolds for skin tissue engineering. This strategy may serve as a general approach in the construction of embryonic-like scaffolds for other tissues/organ. As a first step, key effector molecules upregulated during embryonic and neonatal skin formation were identified using a comparative gene expressing analysis. A set of 20 effector molecules was identified, from which insulin-like growth factor 2 (IGF2) and sonic hedgehog (SHH) were selected for incorporation into a type I collagen-heparin scaffold. Porous scaffolds were constructed using purified collagen fibrils and 6% covalently bound heparin (to bind and protect the growth factors), and IGF2 and SHH were incorporated either individually (~0.7 and 0.4 µg/mg scaffolds) or in combination (combined ~1.5 µg/mg scaffolds). In addition, scaffolds containing hyaluronan (up to 20 µg/mg scaffold) were prepared, based on the up- or downregulation of genes involved in hyaluronan synthesis/degradation and its suggested role in scarless healing. In conclusion, based on a comprehensive gene expression analysis, a set of effector molecules and matrix molecules was identified and incorporated into porous scaffolds. The scaffolds thus prepared may create an 'embryonic-like' environment for cells to recapitulate embryonic events and for new tissues/organs.


Subject(s)
Embryo, Mammalian/cytology , Skin/metabolism , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Blotting, Western , Cattle , Collagen/pharmacology , Collagen Type I/pharmacology , Embryo, Mammalian/drug effects , Gene Expression Regulation, Developmental/drug effects , Hedgehog Proteins/metabolism , Heparin/pharmacology , Hyaluronic Acid/pharmacology , Immunohistochemistry , Insulin-Like Growth Factor II/pharmacology , Mice, Inbred C57BL
5.
Organogenesis ; 11(3): 105-21, 2015.
Article in English | MEDLINE | ID: mdl-26060888

ABSTRACT

A persistent clinical demand exists for a suitable arterial prosthesis. In this study, a vascular conduit mimicking the native 3-layered artery, and constructed from the extracellular matrix proteins type I collagen and elastin, was evaluated for its performance as a blood vessel equivalent. A tubular 3-layered graft (elastin-collagen-collagen) was prepared using highly purified type I collagen fibrils and elastin fibers, resembling the 3-layered native blood vessel architecture. The vascular graft was crosslinked and heparinised (37 ± 4 µg heparin/mg graft), and evaluated as a vascular graft using a porcine bilateral iliac artery model. An intra-animal comparison with clinically-used heparinised ePTFE (Propaten®) was made. Analyses included biochemical characterization, duplex scanning, (immuno)histochemistry and scanning electron microscopy. The tubular graft was easy to handle with adequate suturability. Implantation resulted in pulsating grafts without leakage. One week after implantation, both ePTFE and the natural acellular graft had 100% patencies on duplex scanning. Grafts were partially endothelialised (Von Willebrand-positive endothelium with a laminin-positive basal membrane layer). After one month, layered thrombi were found in the natural (4/4) and ePTFE graft (1/4), resulting in occlusion which in case of the natural graft is likely due to the porosity of the inner elastin layer. In vivo application of a molecularly-defined tubular graft, based on nature's matrix proteins, for vascular surgery is feasible.


Subject(s)
Arterial Occlusive Diseases/physiopathology , Blood Vessel Prosthesis/adverse effects , Collagen/chemistry , Elastin/chemistry , Iliac Artery/physiology , Vascular Patency/physiology , Animals , Arterial Occlusive Diseases/etiology , Bioprosthesis , Equipment Failure Analysis , Extracellular Matrix Proteins/chemistry , Female , Graft Rejection , Iliac Artery/surgery , Prosthesis Design , Swine , Treatment Outcome , Vascular Grafting/adverse effects , Vascular Grafting/instrumentation
6.
Eur J Vasc Endovasc Surg ; 43(1): 66-72, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22001149

ABSTRACT

In recent years, ample attention has been directed towards the mechanisms that play a major role in the process of vascular graft failure, especially graft thrombosis and intimal narrowing have been highlighted. In this article, a survey is conducted into the key mechanisms of the biological processes of intimal hyperplasia and ultimate graft failure. The sequence of biochemical events that lead to thrombosis of grafts is used as a guideline to describe possible counteracting prosthetic surface interventions in each separate phase of the process.


Subject(s)
Blood Vessel Prosthesis Implantation/instrumentation , Blood Vessel Prosthesis , Coated Materials, Biocompatible , Graft Occlusion, Vascular/prevention & control , Thrombosis/prevention & control , Vascular Patency , Animals , Blood Vessel Prosthesis Implantation/adverse effects , Graft Occlusion, Vascular/etiology , Graft Occlusion, Vascular/pathology , Graft Occlusion, Vascular/physiopathology , Humans , Hyperplasia , Prosthesis Design , Thrombosis/etiology , Thrombosis/pathology , Thrombosis/physiopathology
7.
Acta Biomater ; 7(3): 1063-71, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20965285

ABSTRACT

Skin substitutes are of great benefit in the treatment of patients with full thickness wounds, but there is a need for improvement with respect to wound closure with minimal contraction, early vascularisation, and elastin formation. In this study we designed and developed an acellular double-layered skin construct, using matrix molecules and growth factors to target specific biological processes. The epidermal layer was prepared using type I collagen, heparin and fibroblast growth factor 7 (FGF7), while the porous dermal layer was prepared using type I collagen, solubilised elastin, dermatan sulfate, heparin, fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor (VEGF). The construct was biochemically and morphologically characterised and evaluated in vivo using a rat full thickness wound model. The results were compared with the commercial skin substitute IntegraDRT and untreated wounds. The double-layered construct was prepared according to the design specifications. The epidermal layer was about 40 µm thick, containing 9% heparin and 0.2 µg FGF7 mg per layer, localised at the periphery. The dermal layer was 2.5 mm thick, had rounded pores and contained 10% dermatan sulfate+heparin, and 0.7 µg FGF2+VEGF mg per layer. The double-layered skin construct was implanted in a skin defect and on day 7, 14, 28 and 112 the (remaining) wound area was photographed, excised and (immuno) histologically evaluated. The double-layered skin construct showed more cell influx, significantly less contraction and increased blood vessel formation at early time points in comparison with IntegraDRT and/or the untreated wound. On day 14 the double-layered skin construct also had the fewest myofibroblasts present. On day 112 the double-layered skin construct contained more elastic fibres than IntegraDRT and the untreated wound. Structures resembling hair follicles and sebaceous glands were found in the double-layered skin construct and the untreated wound, but hardly any were found in IntegraDRT. The results provide new opportunities for the application of acellular skin constructs in the treatment of surgical wounds.


Subject(s)
Blood Vessels/growth & development , Skin, Artificial , Skin/growth & development , Animals , Blood Vessels/metabolism , Collagen/metabolism , Drug Evaluation, Preclinical , Elastin/metabolism , Male , Microscopy, Electron, Scanning , Rats , Rats, Wistar , Skin/metabolism
8.
Acta Biomater ; 6(12): 4666-74, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20619367

ABSTRACT

There is a consistent need for a suitable natural biomaterial to function as an arterial prosthesis in achieving arterial regeneration. Natural grafts are generally obtained by decellularization of native blood vessels, but batch to batch variations may occur and the nature/content of remaining contaminants is generally unknown. In this study we fabricated a molecularly defined natural arterial graft from scratch resembling the native three layered architecture from the fibrillar extracellular matrix components collagen and elastin. Using casting, moulding, freezing and lyophilization techniques, a triple layered construct was prepared consisting of an inner layer of elastin fibres, a middle (porous) film layer of collagen fibrils and an outer scaffold layer of collagen fibrils. The construct was carbodiimide cross-linked and heparinized. Characterization included biochemical/biophysical analyses, scanning electron microscopy, micro-computed tomography, (immuno)histology and haemocompatibility. Burst pressures were up to 400mm Hg and largely conferred by the intermediate porous collagen film layer. The highly purified type I collagen fibrils and elastin fibres used did not evoke platelet aggregation in vitro. Suturability of the graft in end to side anastomosis was successful and considered adequate for in vivo application.


Subject(s)
Blood Vessel Prosthesis , Blood Vessels/physiology , Collagen/chemistry , Elastin/chemistry , Materials Testing/methods , Tissue Scaffolds/chemistry , Animals , Cattle , Collagen/ultrastructure , Elastin/ultrastructure , Horses , Humans , Immunohistochemistry , Mechanical Phenomena , Microscopy, Electron, Scanning , X-Ray Microtomography
10.
Biomaterials ; 30(31): 6213-20, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19699517

ABSTRACT

Large-scale in vivo evaluation of biomaterials is time-consuming and limited by ethical considerations. The availability of a library of biomaterials would allow a fast and rational in vitro selection of those biomaterials to be evaluated in vivo. For this reason, we developed an array of 48 different, molecularly-defined films based on native fibrillar collagen. The films differed in the type and amount of extracellular matrix components (type I/IV collagens, fibrous/solubilised elastin, glycosaminoglycans, heparin, chondroitin sulfate or dermatan sulfate), method of preparation (homogenisation) and method and extent of crosslinking (carbodiimide (EDC/NHS) or glutaraldehyde). The array was evaluated by studying morphology, proliferation and differentiation of primary human keratinocytes/fibroblasts. Major differences were observed. Only a small selection of films (especially those containing elastin fibres) specifically stimulated the proliferation of keratinocytes, but not fibroblasts. Such films may be the biomaterials of choice for in vivo evaluation for skin tissue engineering and regenerative medicine.


Subject(s)
Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Fibrillar Collagens/chemistry , Skin/cytology , Tissue Engineering/methods , Cell Proliferation/drug effects , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Immunohistochemistry , Microscopy, Electron, Scanning
11.
Biomaterials ; 28(30): 4378-98, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17631957

ABSTRACT

Biomaterials based upon elastin and elastin-derived molecules are increasingly investigated for their application in tissue engineering. This interest is fuelled by the remarkable properties of this structural protein, such as elasticity, self-assembly, long-term stability, and biological activity. Elastin can be applied in biomaterials in various forms, including insoluble elastin fibres, hydrolysed soluble elastin, recombinant tropoelastin (fragments), repeats of synthetic peptide sequences and as block copolymers of elastin, possibly in combination with other (bio)polymers. In this review, the properties of various elastin-based materials will be discussed, and their current and future applications evaluated.


Subject(s)
Biocompatible Materials/chemistry , Elastin/chemistry , Tissue Engineering/methods , Animals , Elastin/physiology , Elastin/ultrastructure , Humans , Models, Biological , Molecular Structure , Protein Structure, Tertiary
15.
J Biomed Mater Res B Appl Biomater ; 77(2): 357-68, 2006 May.
Article in English | MEDLINE | ID: mdl-16362956

ABSTRACT

Porous scaffolds composed of collagen or collagen and elastin were prepared by freeze drying at temperatures between -18 and -196 degrees C. All scaffolds had a porosity of 90-98% and a homogeneous distribution of pores. Freeze drying at -18 degrees C afforded collagen and collagen/elastin matrices with average pore sizes of 340 and 130 mum, respectively. After 20 successive cycles up to 10% of strain, collagen/elastin dense films had a total degree of strain recovery of 70% +/- 5%, which was higher than that of collagen films (42% +/- 6%). Crosslinking of collagen/elastin matrices either in water or ethanol/water (40% v/v) was carried out using a carbodiimide (N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride, EDC) in combination with a succinimide (N-hydroxysuccinimide, NHS) in the presence or absence of a diamine (J230) or by reaction with butanediol diglycidylether (BDGE), followed by EDC/NHS. Crosslinking with EDC/NHS or EDC/NHS/J230 resulted in matrices with increased stiffness as compared to noncrosslinked matrices, whereas sequential crosslinking with the diglycidylether and EDC/NHS yielded very brittle scaffolds. Ethanol/water was the preferred solvent in the crosslinking process because of its ability to preserve the open porous structure during crosslinking. Smooth muscle cells were seeded on the (crosslinked) scaffolds and could be expanded during 14 days of culturing.


Subject(s)
Blood Vessel Prosthesis , Collagen/therapeutic use , Elastin/therapeutic use , Tissue Engineering/methods , Biomechanical Phenomena , Cell Culture Techniques , Cross-Linking Reagents , Freeze Drying , Humans , Myocytes, Smooth Muscle/cytology , Porosity , Solvents
16.
Biomaterials ; 26(1): 81-92, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15193883

ABSTRACT

Collagen-elastin scaffolds may be valuable biomaterials for tissue engineering because they combine tensile strength with elasticity. In this study, the tissue response to and the calcification of these scaffolds were evaluated. In particular, the hypothesis was tested that calcification, a common phenomenon in biomaterials, may be due to microfibrils within the elastic fibre, and that these microfibrils might generate a tissue response. Four scaffolds were subcutaneously implanted, viz. collagen, collagen + pure elastin, collagen+microfibril-containing, and collagen + pulverised elastic ligament (the source for elastin). Explants were evaluated at day 3, 7 and 21. In young Sprague Dawley rats, collagen + ligament calcified substantially, whereas collagen + elastin (with and without microfibrils) calcified less, and collagen did not. Calcification started at elastic fibres. In both Sprague Dawley and Wistar adult rats, however, none of the scaffolds calcified. Mononuclear cell infiltration was prominent in young and adult Sprague Dawley rats. In adult Wistar rats, this infiltration was associated with the presence of microfibrils. Degradation of scaffolds and new matrix formation were related with cellular influx and degree of vascularisation. In conclusion, absence of microfibrils from the elastic fibre does not prevent calcification in young Sprague Dawley rats, but does reduce the tissue response in adult Wistar rats. Cellular response and calcification differs with age and strain and therefore the choice of animal model is of key importance in biomaterial evaluation.


Subject(s)
Aging/pathology , Biocompatible Materials/adverse effects , Calcinosis/pathology , Collagen/adverse effects , Elastin/adverse effects , Foreign-Body Reaction/pathology , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Calcinosis/etiology , Calcinosis/prevention & control , Collagen/chemistry , Elastin/chemistry , Foreign-Body Reaction/etiology , Implants, Experimental/adverse effects , Male , Materials Testing , Rats , Rats, Sprague-Dawley , Rats, Wistar , Species Specificity
17.
Biomaterials ; 24(22): 4001-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12834595

ABSTRACT

Extracellular matrix components are valuable building blocks for the preparation of biomaterials involved in tissue engineering, especially if their biological, chemical and physical characteristics can be controlled. In this study, isolated type I collagen fibrils, elastin fibres and chondroitin sulphate (CS) were used for the preparation of molecularly-defined collagen-elastin-glycosaminoglycan scaffolds. A total of 12 different scaffolds were prepared with four different ratios of collagen and elastin (1:9, 1:1, 9:1 and 1:0), with and without chemical crosslinking, and with and without CS. Collagen was essential to fabricate coherent, porous scaffolds. Electron microscopy showed that collagen and elastin physically interacted with each other and that elastin fibres were enveloped by collagen. By carbodiimide-crosslinking, amine groups were coupled to carboxylic groups and CS could be incorporated. More CS could be bound to collagen scaffolds (10%) than to collagen-elastin scaffolds (2.4-8.5% depending on the ratio). The attachment of CS increased the water-binding capacity to up to 65%. Scaffolds with a higher collagen content had a higher tensile strength whereas addition of elastin increased elasticity. Scaffolds were cytocompatible as was established using human myoblast and fibroblast culture systems. It is concluded that molecularly-defined composite scaffolds can be composed from individual, purified, extracellular matrix components. Data are important in the design and application of tailor-made biomaterials for tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Chondroitin Sulfates/chemistry , Collagen/chemistry , Elastin/chemistry , Glycosaminoglycans/chemistry , Cell Division , Cross-Linking Reagents/pharmacology , Fibroblasts/metabolism , Humans , Microscopy, Electron , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Muscles/cytology , Myoblasts/metabolism , Tissue Engineering , Water/chemistry
18.
Biomaterials ; 22(14): 1997-2005, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11426877

ABSTRACT

Elastin is an insoluble, highly cross-linked protein, providing elasticity to organs like lung. aorta, and ligaments. Despite its remarkable mechanical properties. elastin has found little use as a biomaterial. Purification of intact elastin from elastic fibres presents a major challenge, among others for the intimate interwoveness of elastin and microfibrils. Insoluble elastin preparations tend to calcify, which may be due to calcium-binding microfibrillar (e.g. fibrillin). In this study, elastin was purified from horse ligamentum nuchae using five different procedures. One procedure is based on treatment with 0.1 M NaOH, another on autoclaving and treatment with cyanogen bromide. Three other procedures are based on combinations of extraction steps and enzyme digestions. Purity of preparations was assessed by sodium dodecyl sulphate polyacrylamide gel electrophoresis, amino acid analysis, bright field immunofluorescence and transmission electron microscopy. The procedure involving extractions/enzymes combined with an early application of 2-mercaptoethanol and cyanogen bromide gives a highly pure elastin preparation. Electron microscopic analysis showed that this preparation is devoid of microfibrillar components. This procedure is therefore the method of choice for preparation of insoluble elastin as a biomaterial for tissue engineering.


Subject(s)
Chemical Fractionation/methods , Elastin/isolation & purification , Acetone/pharmacology , Amino Acids/analysis , Animals , Collagenases/pharmacology , Cyanogen Bromide/pharmacology , Elastin/chemistry , Elastin/immunology , Guanidine/pharmacology , Horses , Hot Temperature , Ligaments/chemistry , Ligaments/drug effects , Mercaptoethanol/pharmacology , Microscopy, Electron , Microscopy, Fluorescence , Sodium Hydroxide/pharmacology , Solubility , Solvents/pharmacology , Trypsin/pharmacology
19.
J Agric Food Chem ; 47(8): 2980-90, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10552597

ABSTRACT

beta-Lactoglobulin (betaLg) was hydrolyzed by plasmin to a degree of hydrolysis of 4%. The hydrolysate was fractionated by ion-exchange chromatography and subsequent hydrophobic-interaction chromatography. The betaLg peptide fraction consisting of smaller peptides (mostly <2 kDa) had poor foam- and emulsion-forming and -stabilizing properties. Most of the betaLg peptides were identified (in either the nonreduced or reduced form) by mass spectrometry on the basis of the known primary structure of the intact protein and the specificity of the enzyme. The peptides formed during betaLg/plasmin-hydrolysis were (1) peptides lacking a cysteyl residue, (2) peptides composed of a single amino acid chain containing intramolecular disulfide bonds, and (3) peptides composed of two amino acid chains linked by an intermolecular disulfide bond. It appeared that significant SH/SS-exchange had taken place during hydrolysis. Many of the peptides present in the peptide fraction that exhibited good functional properties were disulfide-linked fragments.


Subject(s)
Fibrinolysin/metabolism , Lactoglobulins/chemistry , Peptide Fragments/chemistry , Amino Acid Sequence , Chromatography, High Pressure Liquid , Disulfides/analysis , Hydrolysis , Lactoglobulins/metabolism , Peptide Fragments/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL