Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Biomaterials ; 34(17): 4259-65, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23489921

ABSTRACT

An important tenet in designing scaffolds for regenerative medicine consists in mimicking the dynamic mechanical properties of the tissues to be replaced to facilitate patient rehabilitation and restore daily activities. In addition, it is important to determine the contribution of the forming tissue to the mechanical properties of the scaffold during culture to optimize the pore network architecture. Depending on the biomaterial and scaffold fabrication technology, matching the scaffolds mechanical properties to articular cartilage can compromise the porosity, which hampers tissue formation. Here, we show that scaffolds with controlled and interconnected pore volume and matching articular cartilage dynamic mechanical properties, are indeed effective to support tissue regeneration by co-cultured primary and expanded chondrocyte (1:4). Cells were cultured on scaffolds in vitro for 4 weeks. A higher amount of cartilage specific matrix (ECM) was formed on mechanically matching (M) scaffolds after 28 days. A less protein adhesive composition supported chondrocytes rounded morphology, which contributed to cartilaginous differentiation. Interestingly, the dynamic stiffness of matching constructs remained approximately at the same value after culture, suggesting a comparable kinetics of tissue formation and scaffold degradation. Cartilage regeneration in matching scaffolds was confirmed subcutaneously in vivo. These results imply that mechanically matching scaffolds with appropriate physico-chemical properties support chondrocyte differentiation.


Subject(s)
Cartilage/physiology , Chemical Phenomena , Regeneration/physiology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cattle , DNA/metabolism , Extracellular Matrix/metabolism , Glycosaminoglycans/metabolism , Materials Testing , Mice , Mice, Nude , Microscopy, Electron, Scanning , Subcutaneous Tissue/metabolism
2.
J Tissue Eng Regen Med ; 7(9): 751-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-22438217

ABSTRACT

Cartilage has a poor regenerative capacity. Tissue-engineering approaches using porous scaffolds seeded with chondrocytes may improve cartilage repair. The aim of this study was to examine the effect of pore size and pore interconnectivity on cartilage repair in osteochondral defects treated with different scaffolds seeded with allogenic chondrocytes. Scaffolds consisting of 55 wt% poly(ethylene oxide terephthalate) and 45 wt% poly(butylene terephthalate) (PEOT/PBT) with different pore sizes and interconnectivities were made, using a compression moulding (CM) and a three-dimensional fibre (3DF) deposition technique. In these scaffolds, allogenic chondrocytes were seeded, cultured for 3 weeks and implanted in osteochondral defects of skeletally mature rabbits. At 3 weeks no difference in cartilage repair between an empty osteochondral defect, CM or 3DF scaffolds was found. Three months post-implantation, cartilage repair was significantly improved after implantation of a 3DF scaffold compared to a CM scaffold. Although not significant, Mankin scores for osteoarthritis (OA) indicated less OA in the 3DF scaffold group compared to empty defects and CM-treated defects. It is concluded that scaffold pore size and pore interconnectivity influences osteochondral repair and a decreased pore interconnectivity seems to impair osteochondral repair.


Subject(s)
Bone and Bones/pathology , Cartilage, Articular/pathology , Regeneration , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Chondrocytes/cytology , Female , Osteoarthritis/pathology , Polyesters/chemistry , Polyethylene Glycols/chemistry , Polyethylene Terephthalates , Polymers/chemistry , Porosity , Rabbits , Wound Healing
3.
Cell Prolif ; 42(4): 485-97, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19486014

ABSTRACT

BACKGROUND: Preliminary studies investigated advanced scaffold design and tissue engineering approaches towards restoring congruent articulating surfaces in small joints. MATERIALS AND METHODS: Anatomical femoral and tibial cartilage constructs, fabricated by three-dimensional fibre deposition (3DF) or compression moulding/particulate leaching (CM), were evaluated in vitro and in vivo in an autologous rabbit model. Effects of scaffold pore architecture on rabbit chondrocyte differentiation and mechanical properties were evaluated following in vitro culture and subcutaneous implantation in nude mice. After femoral and tibial osteotomy and autologous implantation of tissue-engineered constructs in rabbit knee joints, implant fixation and joint articulation were evaluated. RESULTS: Rapid prototyping of 3DF architectures with 100% interconnecting pores promoted homogeneous distribution of viable cells, glycosaminoglycan (GAG) and collagen type II; significantly greater GAG content and differentiation capacity (GAG/DNA) in vitro compared to CM architectures; and higher mechanical equilibrium modulus and dynamic stiffness (at 0.1 Hz). Six weeks after implantation, femoral and tibial constructs had integrated with rabbit bone and knee flexion/extension and partial load bearing were regained. Histology demonstrated articulating surfaces between femoral and tibial constructs for CM and 3DF architectures; however, repair tissue appeared fibrocartilage-like and did not resemble implanted cartilage. CONCLUSIONS: Anatomically shaped, tissue-engineered constructs with designed mechanical properties and internal pore architectures may offer alternatives for reconstruction or restoration of congruent articulating surfaces in small joints.


Subject(s)
Chondrocytes/cytology , Chondrocytes/transplantation , Tissue Engineering/methods , Tissue Scaffolds , Animals , Cartilage/anatomy & histology , Cartilage/cytology , Cell Differentiation , Cell Survival , Cells, Cultured , Chondrocytes/chemistry , DNA/analysis , Femur/anatomy & histology , Femur/cytology , Glycosaminoglycans/analysis , Joints/anatomy & histology , Joints/cytology , Materials Testing , Mechanical Phenomena , Mice , Mice, Nude , Microscopy, Electron, Scanning , Rabbits , Tibia/anatomy & histology , Tibia/cytology , Transplantation, Autologous
4.
Tissue Eng ; 11(9-10): 1297-311, 2005.
Article in English | MEDLINE | ID: mdl-16259586

ABSTRACT

The zonal organization of cells and extracellular matrix (ECM) constituents within articular cartilage is important for its biomechanical function in diarthroidal joints. Tissue-engineering strategies adopting porous three-dimensional (3D) scaffolds offer significant promise for the repair of articular cartilage defects, yet few approaches have accounted for the zonal structural organization as in native articular cartilage. In this study, the ability of anisotropic pore architectures to influence the zonal organization of chondrocytes and ECM components was investigated. Using a novel 3D fiber deposition (3DF) technique, we designed and produced 100% interconnecting scaffolds containing either homogeneously spaced pores (fiber spacing, 1 mm; pore size, about 680 microm in diameter) or pore-size gradients (fiber spacing, 0.5-2.0 mm; pore size range, about 200-1650 microm in diameter), but with similar overall porosity (about 80%) and volume fraction available for cell attachment and ECM formation. In vitro cell seeding showed that pore-size gradients promoted anisotropic cell distribution like that in the superficial, middle, and lower zones of immature bovine articular cartilage, irrespective of dynamic or static seeding methods. There was a direct correlation between zonal scaffold volume fraction and both DNA and glycosaminoglycan (GAG) content. Prolonged tissue culture in vitro showed similar inhomogeneous distributions of zonal GAG and collagen type II accumulation but not of GAG:DNA content, and levels were an order of magnitude less than in native cartilage. In this model system, we illustrated how scaffold design and novel processing techniques can be used to develop anisotropic pore architectures for instructing zonal cell and tissue distribution in tissue-engineered cartilage constructs.


Subject(s)
Cartilage, Articular/cytology , Cartilage, Articular/growth & development , Chondrocytes/cytology , Chondrocytes/physiology , Polymers/chemistry , Tissue Engineering/methods , Animals , Anisotropy , Biocompatible Materials/chemistry , Cattle , Cell Adhesion , Cell Culture Techniques , Cells, Cultured , Chondrocytes/ultrastructure , Collagen Type I/metabolism , Collagen Type I/ultrastructure , Collagen Type II/biosynthesis , Collagen Type II/ultrastructure , DNA/analysis , Extracellular Matrix/physiology , Extracellular Matrix/ultrastructure , Glycosaminoglycans/analysis , Histocytochemistry , Immunohistochemistry , Materials Testing , Models, Biological , Phthalic Acids/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Porosity , Surface Properties , Time Factors
5.
Biomaterials ; 26(1): 63-72, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15193881

ABSTRACT

A highly interconnecting and accessible pore network has been suggested as one of a number of prerequisites in the design of scaffolds for tissue engineering. In the present study, two processing techniques, compression-molding/particulate-leaching (CM), and 3D fiber deposition (3DF), were used to develop porous scaffolds from biodegradable poly(ethylene glycol)-terephthalate/poly(butylene terephthalate) (PEGT/PBT) co-polymers with varying pore architectures. Three-dimensional micro-computed tomography (microCT) was used to characterize scaffold architectures and scaffolds were seeded with articular chondrocytes to evaluate tissue formation. Scaffold porosity ranged between 75% and 80%. Average pore size of tortuous CM scaffolds (182 microm) was lower than those of organized 3DF scaffolds (525 microm). The weight ratio of glycosaminoglycans (GAG)/DNA, as a measure of cartilage-like tissue formation, did not change after 14 days of culture whereas, following subcutaneous implantation, GAG/DNA increased significantly and was significantly higher in 3DF constructs than in CM constructs, whilst collagen type II was present within both constructs. In conclusion, 3DF PEGT/PBT scaffolds create an environment in vivo that enhances cartilaginous matrix deposition and hold particular promise for treatment of articular cartilage defects.


Subject(s)
Cartilage, Articular/cytology , Cartilage, Articular/growth & development , Chondrocytes/cytology , Chondrocytes/physiology , Polyesters/chemistry , Polyethylene Glycols/chemistry , Tissue Engineering/methods , Animals , Bioartificial Organs , Biocompatible Materials/chemistry , Cartilage, Articular/diagnostic imaging , Cattle , Cell Culture Techniques/methods , Cell Proliferation , Cell Survival , Cells, Cultured , Chondrocytes/diagnostic imaging , Compressive Strength , Elasticity , Materials Testing , Porosity , Radiography , Surface Properties
6.
Tissue Eng ; 10(7-8): 987-94, 2004.
Article in English | MEDLINE | ID: mdl-15363156

ABSTRACT

Tissue-engineering approaches for cartilage repair hold promise for the treatment of cartilage defects. Various methods to prevent or reduce dedifferentiation during chondrocyte expansion are currently under investigation. In the present study we evaluated the effect of oxygen on chondrocyte proliferation, as oxygen has received increased attention as a possible regulator of chondrocyte differentiation and its effect during expansion is uncertain. Therefore, the effect of three oxygen tensions (4, 10.5, and 21%) was investigated in a bioreactor microcarrier culture, which allows precise control of the oxygen tension in the liquid phase. During culture cells acquired a round shape on microcarriers. No differences in proliferation rate of chondrocytes were observed within the range of oxygen tensions evaluated. Cells exhibited predominantly anaerobic metabolism and, per mole of glucose, approximately 2 mol of lactate was produced independent of oxygen tension. Cellular oxygen consumption was comparable for all bioreactor cultures. Nevertheless, specific consumption rates were relatively high (2-4 x 10(-17) mol. cell(-1). s(-1)), in comparison with chondrocytes in cartilage (0.8-2.2 x 10(-18) mol. cell(-1)). Subsequent cartilaginous tissue formation in pellets was not affected as qualitatively assessed by safranin-O staining. At the oxygen concentrations evaluated, no effect of oxygen tension was observed on proliferation, oxygen consumption, and yield of lactate on glucose administration. For future investigations of chondrocytes and oxygen, the bioreactor system, which allows precise control and monitoring of oxygen tension, holds promise.


Subject(s)
Bioreactors , Cartilage, Articular/cytology , Cartilage, Articular/growth & development , Chondrocytes/cytology , Chondrocytes/physiology , Oxygen/metabolism , Tissue Engineering/methods , Adult , Animals , Cattle , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Differentiation , Cell Proliferation , Humans , Miniaturization
7.
Tissue Eng ; 10(5-6): 893-903, 2004.
Article in English | MEDLINE | ID: mdl-15265307

ABSTRACT

Anterior cruciate ligament (ACL) reconstruction surgery still has important problems to overcome, such as "donor site morbidity" and the limited choice of grafts in revision surgery. Tissue engineering of ligaments may provide a solution for these problems. Little is known about the optimal cell source for tissue engineering of ligaments. The aim of this study is to determine the optimal cell source for tissue engineering of the anterior cruciate ligament. Bone marrow stromal cells (BMSCs), ACL, and skin fibroblasts were seeded onto a resorbable suture material [poly(L-lactide/glycolide) multifilaments] at five different seeding densities, and cultured for up to 12 days. All cell types tested attached to the suture material, proliferated, and synthesized extracellular matrix rich in collagen type I. On day 12 the scaffolds seeded with BMSCs showed the highest DNA content (p < 0.01) and the highest collagen production (p < 0.05 for the two highest seeding densities). Scaffolds seeded with ACL fibroblasts showed the lowest DNA content and collagen production. Accordingly, BMSCs appear to be the most suitable cells for further study and development of tissue-engineered ligament.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Fibroblasts/cytology , Fibroblasts/physiology , Ligaments/cytology , Ligaments/physiology , Tissue Engineering/methods , Animals , Anterior Cruciate Ligament/cytology , Anterior Cruciate Ligament/physiology , Cell Culture Techniques/methods , Cell Differentiation , Cell Proliferation , Cell Survival/physiology , Cells, Cultured , Extracellular Matrix/physiology , Goats , Skin/cytology , Skin Physiological Phenomena , Stromal Cells/cytology , Stromal Cells/physiology
8.
Biomaterials ; 25(26): 5773-80, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15147823

ABSTRACT

Repair of articular cartilage defects using tissue engineered constructs composed of a scaffold and cultured autologous cells holds promise for future treatments. However, nutrient limitation (e.g. oxygen) has been suggested as a cause of the onset of chondrogenesis solely within the peripheral boundaries of larger constructs. In the present study, oxygen gradients were evaluated by microelectrode measurements in two porous polyethylene glycol terephthalate/polybutylene terephthalate (PEGT/PBT) scaffold architectures, a compression-molded and particle-leached sponge (CM) and a 3D-deposited fiber (3DF) scaffold. During the first 14 days in vitro, gradients intensified, after which a gradual decrease of the gradients was observed in vitro. In vivo, however, gradients changed instantly and became less pronounced. Although similar gradients were observed regardless of scaffold type, significantly more cells were present in the center of 3DF constructs after 2 weeks of in vivo culture. Our results stress the importance of a rationally designed scaffold for tissue-engineering applications. Organized structures, such as the 3DF PEGT/PBT polymer scaffolds, offer possibilities for regulation of nutrient supply and, therefore, hold promise for clinical approaches for cartilage repair.


Subject(s)
Cell Culture Techniques/methods , Chondrocytes/cytology , Chondrocytes/metabolism , Oxygen/chemistry , Oxygen/metabolism , Polyesters/chemistry , Polyethylene Glycols/chemistry , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Cartilage/cytology , Cartilage/metabolism , Cattle , Cells, Cultured , Materials Testing , Mice , Mice, Nude , Molecular Conformation , Oxygen/analysis , Polyesters/analysis , Polyethylene Glycols/analysis , Surface Properties
9.
Biomaterials ; 25(18): 4149-61, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15046905

ABSTRACT

In this study, we present and characterize a fiber deposition technique for producing three-dimensional poly(ethylene glycol)-terephthalate-poly(butylene terephthalate) (PEGT/PBT) block co-polymer scaffolds with a 100% interconnecting pore network for engineering of articular cartilage. The technique allowed us to "design-in" desired scaffold characteristics layer by layer by accurately controlling the deposition of molten co-polymer fibers from a pressure-driven syringe onto a computer controlled x-y-z table. By varying PEGT/PBT composition, porosity and pore geometry, 3D-deposited scaffolds were produced with a range of mechanical properties. The equilibrium modulus and dynamic stiffness ranged between 0.05-2.5 and 0.16-4.33 MPa, respectively, and were similar to native articular cartilage explants (0.27 and 4.10 MPa, respectively). 3D-deposited scaffolds seeded with bovine articular chondrocytes supported a homogeneous cell distribution and subsequent cartilage-like tissue formation following in vitro culture as well as subcutaneous implantation in nude mice. This was demonstrated by the presence of articular cartilage extra cellular matrix constituents (glycosaminoglycan and type II collagen) throughout the interconnected pore volume. Similar results were achieved with respect to the attachment of expanded human articular chondrocytes, resulting in a homogeneous distribution of viable cells after 5 days dynamic seeding. The processing methods and model scaffolds developed in this study provide a useful method to further investigate the effects of scaffold composition and pore architecture on articular cartilage tissue formation.


Subject(s)
Cartilage, Articular/cytology , Cartilage, Articular/growth & development , Chondrocytes/cytology , Chondrocytes/physiology , Polyesters/chemistry , Polyethylene Glycols/chemistry , Polyethylene Terephthalates/analogs & derivatives , Polyethylene Terephthalates/chemistry , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Cattle , Cell Adhesion , Cell Division , Cell Survival , Compressive Strength , Crystallization/methods , Elasticity , Extracellular Matrix/physiology , Humans , Materials Testing , Porosity , Surface Properties , Viscosity
10.
Biotechnol Bioeng ; 86(1): 9-18, 2004 Apr 05.
Article in English | MEDLINE | ID: mdl-15007836

ABSTRACT

The supply of oxygen within three-dimensional tissue-engineered (TE) cartilage polymer constructs is mainly by diffusion. Oxygen consumption by cells results in gradients in the oxygen concentration. The aims of this study were, firstly, to identify the gradients within TE cartilage polymer constructs and, secondly, to predict the profiles during in vitro culture. A glass microelectrode system was adapted and used to penetrate cartilage and TE cartilaginous constructs, yielding reproducible measurements with high spatial resolution. Cartilage polymer constructs were cultured for up to 41 days in vitro. Oxygen concentrations, as low as 2-5%, were measured within the center of these constructs. At the beginning of in vitro culture, the oxygen gradients were steeper in TE constructs in comparison to native tissue. Nevertheless, during the course of culture, oxygen concentrations approached the values measured in native tissue. A mathematical model was developed which yields oxygen profiles within cartilage explants and TE constructs. Model input parameters were assessed, including the diffusion coefficient of cartilage (2.2 x 10(-9)) + (0.4 x 10(-9) m(2) s(-1)), 70% of the diffusion coefficient of water and the diffusion coefficient of constructs (3.8 x 10(-10) m(2) s(-1)). The model confirmed that chondrocytes in polymer constructs cultured for 27 days have low oxygen requirements (0.8 x 10(-19) mol m(-3) s(-1)), even lower than chondrocytes in native cartilage. The ability to measure and predict local oxygen tensions offers new opportunities to obtain more insight in the relation between oxygen tension and chondrogenesis.


Subject(s)
Chondrocytes/cytology , Chondrocytes/metabolism , Models, Biological , Oxygen Consumption/physiology , Oxygen/metabolism , Polyesters/chemistry , Polyethylene Glycols/chemistry , Tissue Engineering/methods , Animals , Biocompatible Materials/chemistry , Cattle , Cell Culture Techniques/methods , Cell Division/physiology , Cells, Cultured , Chondrogenesis/physiology , Diffusion , Materials Testing , Models, Chemical , Oxygen/chemistry , Tissue Distribution
11.
Osteoarthritis Cartilage ; 12(4): 306-13, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15023382

ABSTRACT

OBJECTIVE: To determine the effect of dissolved oxygen tension (DO) on the redifferentiation of dedifferentiated adult human nasal septum chondrocytes cultured as pellets. DESIGN: After isolation, human nasal chondrocytes were expanded in monolayer culture, which resulted in their dedifferentiation. Dedifferentiated cells were pelleted, transferred to a bioreactor and maintained for up to 21 days at 100% DO (21% oxygen), 25% DO (5.25% oxygen) or 5% DO (1% oxygen), which was controlled in the liquid phase. Redifferentiation was assessed by staining the extracellular matrix with safranin-O and by the immunolocalization of collagen types I, II, IX and of a fibroblast membrane marker (11-fibrau). In addition, glycosaminoglycans (GAG) and DNA content were determined spectrophotometrically. RESULTS: In monolayer culture, cells dedifferentiated and multiplied 90- to 100-fold. Cell pellets cultured in a bioreactor under conditions of low oxygen tension (25% DO or 5% DO) stained intensely for GAGs and for collagen type II, but very weakly for collagen type I. After 14 days of culturing, cell pellets maintained at 5% DO stained more intensely for collagen IX and more weakly for 11-fibrau than did those incubated at 25% DO. After 21 days of culturing the GAG content of cell pellets maintained at 5% DO was significantly greater than that of those incubated at 25% DO. Under air-saturated conditions (100% DO), the DNA and GAG contents of cell pellets decreased with time in culture. After 21 days of culturing, both parameters were substantially lower in cell pellets maintained at 100% DO than in those incubated at low oxygen tensions. The staining signals for collagen types II and IX were much weaker, and those for the markers of dedifferentiation (collagen type I and 11-fibrau) much stronger under air-saturated conditions than at low oxygen tensions. CONCLUSION: These observations demonstrate that using the present set-up, low oxygen tension stimulates the redifferentiation of dedifferentiated adult human nasal chondrocytes in pellet cultures.


Subject(s)
Cell Differentiation/physiology , Chondrocytes/physiology , Nasal Septum/physiology , Oxygen/physiology , Adult , Antigens, Surface/analysis , Biomarkers/analysis , Cells, Cultured , Collagen Type I/analysis , Collagen Type II/analysis , Collagen Type IX/analysis , DNA/analysis , Glycosaminoglycans/analysis , Humans
12.
Tissue Eng ; 9(5): 939-48, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14633378

ABSTRACT

Functional cartilage implants for orthopedic surgery or in vitro tissue evaluation can be created from expanded chondrocytes and biodegradable scaffolds. Expansion of chondrocytes in two-dimensional culture systems results in their dedifferentiation. The hallmark of this process is the switch of collagen synthesis from type II to type I. The aim of this study was to evaluate the postexpansion chondrogenic potential of microcarrier-expanded bovine articular chondrocytes in pellet cultures. A selection of microcarriers was screened for initial attachment of chondrocytes. On the basis of those results and additional selection criteria related to clinical application, Cytodex-1 microcarriers were selected for further investigation. Comparable doubling times were obtained in T-flask and microcarrier cultures. During propagation on Cytodex-1 microcarriers, cells acquired a spherical-like morphology and the presence of collagen type II was detected. Both observations are indicative of a differentiated chondrocyte. Pellet cultures of microcarrier-expanded cells showed cartilage-like morphology and staining for proteoglycans and collagen type II after 14 days. In contrast, pellets of T-flask-expanded cells had a fibrous appearance and showed abundant staining only for collagen type I. Therefore, culture of chondrocytes on microcarriers may offer useful and cost-effective cell expansion opportunities in the field of cartilage tissue engineering.


Subject(s)
Cell Differentiation/physiology , Chondrocytes/physiology , Tissue Engineering/methods , Animals , Cattle , Cell Adhesion/physiology , Cell Division/physiology , Collagen Type I/immunology , Collagen Type I/metabolism , Collagen Type II/immunology , Collagen Type II/metabolism , Immunohistochemistry
13.
Biomaterials ; 24(28): 5153-61, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14568432

ABSTRACT

Articular cartilage has a limited capacity for self-repair. To overcome this problem, it is expected that functional cartilage replacements can be created from expanded chondrocytes seeded in biodegradable scaffolds. Expansion of chondrocytes in two-dimensional culture systems often results in dedifferentiation. This investigation focuses on the post-expansion phenotype of human nasal chondrocytes expanded on macroporous gelatin CultiSpher G microcarriers. Redifferentiation was evaluated in vitro via pellet cultures in three different culture media. Furthermore, the chondrogenic potential of expanded cells seeded in polyethylene glycol terephthalate/ polybuthylene terephthalate (PEGT/PBT) scaffolds, cultured for 14 days in vitro, and subsequently implanted subcutaneously in nude mice, was assessed. Chondrocytes remained viable during microcarrier culture and yielded doubling times (1.07+/-0.14 days) comparable to T-flask expansion (1.20+/-0.36 days). Safranin-O staining from pellet culture in different media demonstrated that production of GAG per cell was enhanced by microcarrier expansion. Chondrocyte-polymer constructs with cells expanded on microcarriers contained significantly more proteoglycans after subcutaneous implantation (288.5+/-29.2 microg) than those with T-flask-expanded cells (164.0+/-28.7 microg). Total collagen content was similar between the two groups. This study suggests that macroporous gelatin microcarriers are effective matrices for nasal chondrocyte expansion, while maintaining the ability of chondrocyte differentiation. Although the exact mechanism by which chondrocyte redifferentiation is induced through microcarrier expansion has not yet been elucidated, this technique shows promise for cartilage tissue engineering approaches.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/physiology , Chondrocytes/cytology , Chondrocytes/physiology , Gelatin , Polyethylene Terephthalates , Tissue Engineering/methods , Transplants , Adolescent , Adult , Animals , Biomimetic Materials/metabolism , Cell Culture Techniques/instrumentation , Cell Division/physiology , Cell Survival/physiology , Cells, Cultured , Extracellular Matrix/physiology , Humans , Materials Testing , Membranes, Artificial , Mice , Mice, Nude , Middle Aged , Nasal Cavity/cytology , Nasal Cavity/physiology , Porosity , Prostheses and Implants , Tissue Engineering/instrumentation
14.
J Mater Sci Mater Med ; 14(3): 235-40, 2003 Mar.
Article in English | MEDLINE | ID: mdl-15348469

ABSTRACT

Previously, it was found that chondrocytes and fibroblasts could be efficiently seeded onto three-dimensional scaffolds in spinner flasks. In this study different culture conditions were compared to create a living dermal substitute as rapidly as possible. Human dermal fibroblasts were dynamically seeded onto biodegradable porous PEGT/PBT copolymer (PolyActive) scaffolds for 24 h in spinner flasks. Subsequently, the cell-seeded scaffolds were cultured in two conditions: statically (without medium flow, S) and dynamically (with slow medium flow, D). Qualitative analyses (scanning electron microscopy and histology) and quantitative assays for DNA, total collagen (hydroxyproline) and glycosaminoglycans were done with samples cultured for 3, 7, 14 and 21 days. In dynamically cultured constructs, human dermal fibroblasts were uniformly distributed throughout the pores of the scaffolds and had deposited higher amounts of extracellular matrix (ECM). Significantly higher numbers of fibroblasts were found (p<0.001), and significantly more collagen (hydroxyproline content) (p<0.001) and glycosaminoglycan (GAG) (p<0.05) were deposited at all the investigated time points when compared to static cultured constructs. In conclusion, medium flow stimulated the proliferation of human dermal fibroblasts and accelerated the ECM deposition in PolyActive dermal substitutes when compared to static culture. Dynamic culture reduced the time to create a dermal substitute containing autologous fibroblasts.

15.
Crit Rev Eukaryot Gene Expr ; 12(3): 209-36, 2002.
Article in English | MEDLINE | ID: mdl-12449344

ABSTRACT

Articular cartilage lesions resulting from trauma or degenerative diseases are commonly encountered clinical problems. It is well-established that adult articular cartilage has limited regenerative capacity, and, although numerous treatment protocols are currently employed clinically, few approaches exist that are capable of consistently restoring long-term function to damaged articular cartilage. Tissue engineering strategies that focus on the use of three-dimensional scaffolds for repairing articular cartilage lesions offer many advantages over current treatment strategies. Appropriate design of biodegradable scaffold conduits (either preformed or injectable) allow for the delivery of reparative cells bioactive factors, or gene factors to the defect site in an organized manner. This review seeks to highlight pertinent design considerations and limitations related to the development, material selection, and processing of scaffolds for articular cartilage tissue engineering, evidenced over the last decade. In particular, considerations for novel repair strategies that use scaffolds in combination with controlled release of bioactive factors or gene therapy are discussed, as are scaffold criteria related to mechanical stimulation of cell-seeded constructs. Furthermore, the subsequent impact of current and future aspects of these multidisciplinary scaffold-based approaches related to in vitro and in vivo cartilage tissue engineering are reported herein.


Subject(s)
Cartilage , Tissue Engineering , Animals , Cartilage/injuries , Cartilage/physiology , Chondroitin Sulfates/metabolism , Collagen Type I/metabolism , Humans , Manufactured Materials , Rats
16.
Cell Tissue Bank ; 3(4): 245-53, 2002.
Article in English | MEDLINE | ID: mdl-15256873

ABSTRACT

Skin defects left after excision of hypertrophic scars were treated with a dermal substitute and split-thickness skin grafts transplanted after vascularisation of the substitute. The used substitute was a synthetic porous scaffold made from the biodegradable copolymer polyethyleneglycol-terephtalate and polybuthylene-terephtalate. The study was designed to assess the rate of granulation tissue formation, graft take, and after 3 and 12 months the quality of life (pain, comfort of treatment, cosmetic or functional nuisance), scar formation and wound contraction. In addition, scaffold biodegradation and scar tissue formation were evaluated histologically.Seven patients with different causes of burn injury were enrolled, of which 5 completed the study. In the first 4 patients the time between scaffold application and split-thickness skin overgrafting was in between 17 and 24 days. The time point of overgrafting was significantly reduced to 10-12 days by meshing of the dermal scaffold as evidenced in the last 3 patients. Histological evaluation at 3 months revealed normal generation of dermal tissue, however, the collagen bundles were parallel organized like in scar tissue. In the deeper layers of the neodermis, fragments of the dermal substitute were present, causing a mild inflammatory response. One year post-treatment, some fragments of the copolymer were still observed. The extent of wound contraction after successful overgrafting ranged from 30% to 57% after 1 year. All 5 patients showed an improvement in the total Vancouver Scar Score compared to the value before scar removal being similar to what can be expected when treated with split-thickness skin grafts alone. No unanticipated adverse effects due to application of the substitute were observed.We conclude that although this synthetic dermal substitute can be safely used in humans, the presence of 3D dermal template in a full-thickness skin defect will not automatically improve the skin tissue regeneration process or inhibit wound contraction.

17.
J Biomed Mater Res ; 54(1): 47-58, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11077402

ABSTRACT

The attachment, proliferation, morphology, and differentiation of two cell types-skeletal muscle cells and chondrocytes-were investigated on different compositions of poly(ethylene glycol) and poly(butylene terephthalate) segmented block copolymers. Four weight percentages (40, 55, 60, and 70%) and two different molecular weights (300 and 1000 Da) of poly(ethylene glycol) were tested. Varying the weight percentage and molecular weight of poly(ethylene glycol) resulted in different behaviors for skeletal muscle cells and chondrocytes. The attachment of skeletal muscle was the highest (similar to tissue culture polystyrene) when copolymers containing 55 wt % of poly(ethylene glycol) were used, regardless of the poly(ethylene glycol) molecular weight. Maximum proliferation and differentiation of skeletal muscle cells was achieved when copolymers containing 55 wt % and 300 Da molecular weight of poly(ethylene glycol) were used. In contrast, the weight percentage and molecular weight of poly(ethylene glycol) had no significant effect on chondrocyte attachment and proliferation; the attached chondrocytes retained a differentiated phenotype only when a 70 wt % of poly(ethylene glycol) was used. Cell behavior was correlated with the surface properties of the copolymer films, as indicated by contact-angle measurements. These results suggest that an optimized wt % and molecular weight of poly(ethylene glycol) will be useful depending on the specific cell type.


Subject(s)
Biocompatible Materials , Chondrocytes/physiology , Muscle, Skeletal/cytology , Polyesters , Polyethylene Glycols , Animals , Blotting, Western , Cattle , Cell Adhesion , Cell Differentiation/physiology , Cell Line , Cell Separation , Chondrocytes/ultrastructure , Cytological Techniques , Materials Testing , Mice , Microscopy, Electron, Scanning , Molecular Weight , Muscle, Skeletal/ultrastructure , Surface Properties
19.
Biophys J ; 76(5): 2702-10, 1999 May.
Article in English | MEDLINE | ID: mdl-10233084

ABSTRACT

The sensitivity of a nitroxide spin label to the polarity of its environment has been used to estimate the hydrophobic barrier of the proton channel of the transmembrane proton pump bacteriorhodopsin. By means of site-specific mutagenesis, single cysteine residues were introduced at 10 positions located at the protein surface, in the protein interior, and along the proton pathway. After reaction with a methanethiosulfonate spin label, the principle values of the hyperfine tensor A and the g-tensor were determined from electron paramagnetic resonance spectra measured at 170 K. The shape of the hydrophobic barrier of the proton channel is characterized in terms of a polarity index, DeltaA, determined from the variation of the hyperfine coupling constant Azz. The maximum of the hydrophobic barrier is found to be close to the retinal chromophore in the proton uptake pathway. The effect of the asymmetric distribution of charged and polar residues in the proton release and uptake pathways is clearly reflected in the behavior of the hydrophobic barrier. The presence of azide reduces the barrier height of both the cytoplasmic and extracellular channels. This finding supports the view of azide and other weakly acidic anions as catalysts for the formation of hydrogen-bonded networks in proton pathways of proteins.


Subject(s)
Bacteriorhodopsins/chemistry , Azides/pharmacology , Bacteriorhodopsins/genetics , Bacteriorhodopsins/radiation effects , Biophysical Phenomena , Biophysics , Cysteine/chemistry , Electron Spin Resonance Spectroscopy , Hydrogen Bonding , Models, Molecular , Mutagenesis, Site-Directed , Nitrogen Oxides/chemistry , Oxalates/pharmacology , Protein Conformation , Protons , Spectroscopy, Fourier Transform Infrared , Spectrum Analysis, Raman , Spin Labels
20.
J Mater Sci Mater Med ; 10(12): 773-7, 1999 Dec.
Article in English | MEDLINE | ID: mdl-15347949

ABSTRACT

The present study aims at optimizing dermal fibroblast seeding and cultivation in Polyactive scaffolds in order to limit the biopsy size needed for autologous treatment of full-thickness skin defects and chronic wounds. Three methods for seeding and cultivation of fibroblasts in porous scaffolds were compared: dynamic seeding followed by static cultivation (DS), static seeding followed by static cultivation (SS) and dynamic seeding followed by dynamic cultivation (DD). Human dermal fibroblasts isolated from cultured explants were seeded in porous PEO/PBT (Polyactive) scaffolds. Samples were taken from 6 h to 21 days post-seeding for both histological analysis (cell distribution and extracellular matrix (ECM) formation), and quantitative cell number assay. The seeding efficiency 24 h post-seeding was 76% (+/-3.6%) for dynamically seeded matrices, whereas it was only 30% (+/-5%) for statically seeded matrices (p<0.001). ECM formation was abundant in DS samples already at day 10, while even after 21 days ECM formation was less pronounced in SS samples. Surprisingly, cells detached from DD samples as aggregates, starting from day 10. Cell numbers as assayed quantitatively correlated with the histological results. At all timepoints cell numbers found for DS samples were significantly higher as compared to SS samples. At day 21, DS samples contained approximately twofold more cells as compared to SS and DD samples and comprised ECM consisting of collagen types I and III. Our results indicate that the combination of dynamic seeding and static cultivation assures efficient utilization of isolated fibroblasts and improved neodermis formation, thereby allowing a reduction in the skin biopsy size needed for the engineering of living skin substitute.

SELECTION OF CITATIONS
SEARCH DETAIL
...