Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mater Sci Eng C Mater Biol Appl ; 97: 567-575, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30678943

ABSTRACT

The biofunctionalization of scaffolds for tissue engineering is crucial to improve the results of regenerative therapies. This study compared the effect of platelet-functionalization of 2D electrospun and 3D centrifugal spun scaffolds on the osteogenic potential of hMSCs. Scaffolds prepared from poly-ε-caprolactone, using electrospinning and centrifugal spinning technology, were functionalized using five different concentrations of platelets. Cell proliferation, metabolic activity and osteogenic differentiation were tested using hMSCs cultured in differential and non-differential medium. The porous 3D structure of the centrifugal spun fibers resulted in higher cell proliferation. Furthermore, the functionalization of the scaffolds with platelets resulted in a dose-dependent increase in cell metabolic activity, proliferation and production of an osteogenic marker - alkaline phosphatase. The effect was further promoted by culture in an osteogenic differential medium. The increase in combination of both platelets and osteogenic media shows an improved osteoinduction by platelets in environments rich in inorganic phosphate and ascorbate. Nevertheless, the results of the study showed that the optimal concentration of platelets for induction of hMSC osteogenesis is in the range of 900-3000 × 109 platelets/L. The study determines the potential of electrospun and centrifugal spun fibers with adhered platelets, for use in bone tissue engineering.


Subject(s)
Blood Platelets/metabolism , Polyesters/chemistry , Tissue Engineering , Tissue Scaffolds/chemistry , Alkaline Phosphatase/metabolism , Blood Platelets/cytology , Cell Adhesion , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Elastic Modulus , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis , Porosity
2.
Cell Adh Migr ; 12(3): 271-285, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29130836

ABSTRACT

Additive manufacturing, also called 3D printing, is an effective method for preparing scaffolds with defined structure and porosity. The disadvantage of the technique is the excessive smoothness of the printed fibers, which does not support cell adhesion. In the present study, a 3D printed scaffold was combined with electrospun classic or structured nanofibers to promote cell adhesion. Structured nanofibers were used to improve the infiltration of cells into the scaffold. Electrospun layers were connected to 3D printed fibers by gluing, thus enabling the fabrication of scaffolds with unlimited thickness. The composite 3D printed/nanofibrous scaffolds were seeded with primary chondrocytes and tested in vitro for cell adhesion, proliferation and differentiation. The experiment showed excellent cell infiltration, viability, and good cell proliferation. On the other hand, partial chondrocyte dedifferentiation was shown. Other materials supporting chondrogenic differentiation will be investigated in future studies.


Subject(s)
Cell Adhesion/physiology , Chondrocytes/cytology , Nanofibers , Printing, Three-Dimensional , Tissue Scaffolds , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured/physiology , Humans , Nanofibers/chemistry , Tissue Engineering/methods
3.
Biomed Mater ; 13(2): 025004, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29084934

ABSTRACT

Fibrous scaffolds are desired in tissue engineering applications for their ability to mimic extracellular matrix. In this study we compared fibrous scaffolds prepared from polycaprolactone using three different fabrication methods, electrospinning (ES), electro-blowing and melt-blown combined with ES. Scaffolds differed in morphology, fiber diameters and pore sizes. Mesenchymal stem cell adhesion, proliferation and osteogenic differentiation on scaffolds was evaluated. The most promising scaffold was shown to be melt-blown in combination with ES which combined properties of both technologies. Microfibers enabled good cell infiltration and nanofibers enhanced cell adhesion. This scaffold was used for further testing in critical sized defects in rabbits. New bone tissue formation occurred from the side of the treated defects, compared to a control group where only fat tissue was present. Polycaprolactone fibrous scaffold prepared using a combination of melt-blown and ES technology seems to be promising for bone regeneration. The practical application of results is connected with enormous production capacity and low cost of materials produced by melt-blown technology, compared to other bone scaffold fabrication methods.


Subject(s)
Bone and Bones/pathology , Nanofibers/chemistry , Osteogenesis/drug effects , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Bone Regeneration , Cell Adhesion , Cell Proliferation , Cell Survival , Femur/pathology , Male , Mesenchymal Stem Cells/cytology , Microscopy, Electron, Scanning , Polymers/chemistry , Rabbits
4.
J Biomed Mater Res A ; 103(2): 671-82, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24838634

ABSTRACT

A three-dimensional scaffold of type I collagen and hydroxyapatite enriched with polycaprolactone nanofibers (Coll/HA/PCL), autologous mesenchymal stem cells (MSCs) in osteogenic media, and thrombocyte-rich solution (TRS) was an optimal implant for bone regeneration in vivo in white rabbits. Nanofibers optimized the viscoelastic properties of the Coll/HA scaffold for bone regeneration. MSCs and TRS in the composite scaffold improved bone regeneration. Three types of Coll/HA/PCL scaffold were prepared: an MSC-enriched scaffold, a TRS-enriched scaffold, and a scaffold enriched with both MSCs and TRS. These scaffolds were implanted into femoral condyle defects 6 mm in diameter and 10-mm deep. Untreated defects were used as a control. Macroscopic and histological analyses of the regenerated tissue from all groups were performed 12 weeks after implantation. The highest volume and most uniform distribution of newly formed bone occurred in defects treated with scaffolds enriched with both MSCs and TRS compared with that in defects treated with scaffolds enriched by either component alone. The modulus of elasticity in compressive testing was significantly higher in the Coll/HA/PCL scaffold than those without nanofibers. The composite Coll scaffold functionalized with PCL nanofibers and enriched with MSCs and TRS appears to be a novel treatment for bone defects.


Subject(s)
Blood Platelets/chemistry , Bone Regeneration , Collagen/chemistry , Durapatite/chemistry , Mesenchymal Stem Cells/metabolism , Nanofibers/chemistry , Polyesters/chemistry , Tissue Scaffolds/chemistry , Animals , Cells, Cultured , Mesenchymal Stem Cells/cytology , Rabbits
5.
Article in Czech | MEDLINE | ID: mdl-23452427

ABSTRACT

PURPOSE OF THE STUDY: Articular cartilage defects arise due to injury or osteochondral disease such as osteonecrosis or osteochondritis dissecans. In adult patients cartilage has minimal ability to repair itself and the lesions develop into degenerative arthritis. Overcoming the low regenerative capacity of the cartilage cells and the Hayflick limit poses a challenge for the therapy of osteochondral defects. Composite scaffolds with appropriate biomechanical properties combined with a suitable blend of proliferation and differentiation factors could be a solution. The aim of this in vitro study was to develop a novel functionalised hydrogel with an integrated drug delivery system stimulating articular cartilage regeneration. MATERIAL AND METHODS: Injectable collagen/ hyaluronic acid/fibrin composite hydrogel was mixed with nanofibre-based microparticles. These were loaded with ascorbic acid and dexamethasone. In addition, the effect of thrombocyte-rich solution (TRS) was studied. The gels seeded with mesenchymal stem cells (MSCs) were cultivated for 14 days. The viability, proliferation and morphology of the cells were evaluated using molecular and microscopic methods. Scaffold degradation was also assessed. RESULTS: The cultivation study showed that MSCs remained viable in all experimental groups, which indicated good biocompatibility of the gel. However, the number of cells in the groups enriched with microparticles was lower than in the other groups. On the other hand, confocal microscopy showed higher cell viability and rounded morphology of the cells, which can be associated with chodrogenic differentiation. The scaffolds containing microparticles showed significantly higher stability during the 14-day experiment. DISCUSSION: Our results suggest that the addition of microparticles to the scaffold improved cell differentiation into the chondrogenic lineage, resulting in a lower proliferation rate. Cell viability was better in the groups enriched with microparticles that served as an efficient drug delivery system. In addition, the presence of microparticles slowed down gel degradation which can help achieve sufficient stability of the system for the time frame required for cartilage regeneration. CONCLUSIONS: The novel approach described here produced an efficient system where microparticles served as a drug delivery system and stabilised the gel for prolonged periods of time. These characteristics play an important role in the development of scaffolds for cartilage regeneration. In the future the results of these in vitro experiments will be verified in an in vivo study.


Subject(s)
Ascorbic Acid/pharmacology , Cartilage, Articular/physiology , Dexamethasone/pharmacology , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Regeneration/drug effects , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Blood Platelets/physiology , Cells, Cultured , Drug Delivery Systems , Humans , Injections , Materials Testing , Mesenchymal Stem Cells , Osteochondritis Dissecans/therapy , Osteonecrosis/therapy , Tissue Engineering/methods
6.
Cell Prolif ; 46(1): 23-37, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23216517

ABSTRACT

OBJECTIVES: We prepared 3D poly (ε-caprolactone) (PCL) nanofibre scaffolds and tested their use for seeding, proliferation, differentiation and migration of mesenchymal stem cell (MSCs). MATERIALS AND METHODS: 3D nanofibres were prepared using a special collector for common electrospinning; simultaneously, a 2D PCL nanofibre layer was prepared using a classic plain collector. Both scaffolds were seeded with MSCs and biologically tested. MSC adhesion, migration, proliferation and osteogenic differentiation were investigated. RESULTS: The 3D PCL scaffold was characterized by having better biomechanical properties, namely greater elasticity and resistance against stress and strain, thus this scaffold will be able to find broad applications in tissue engineering. Clearly, while nanofibre layers of the 2D scaffold prevented MSCs from migrating through the conformation, cells infiltrated freely through the 3D scaffold. MSC adhesion to the 3D nanofibre PCL layer was also statistically more common than to the 2D scaffold (P < 0.05), and proliferation and viability of MSCs 2 or 3 weeks post-seeding, were also greater on the 3D scaffold. In addition, the 3D PCL scaffold was also characterized by displaying enhanced MSC osteogenic differentiation. CONCLUSIONS: We draw the conclusion that all positive effects observed using the 3D PCL nanofibre scaffold are related to the larger fibre surface area available to the cells. Thus, the proposed 3D structure of the nanofibre layer will find a wide array of applications in tissue engineering and regenerative medicine.


Subject(s)
Cell Culture Techniques/instrumentation , Cell Differentiation , Mesenchymal Stem Cells/cytology , Nanofibers/chemistry , Polyesters/chemistry , Tissue Scaffolds , Cell Culture Techniques/methods , Cell Movement , Cell Proliferation , Cell Survival , Cells, Cultured , Elasticity , Humans , Integrin-Binding Sialoprotein/metabolism , Mesenchymal Stem Cells/metabolism , Nanofibers/ultrastructure , Osteocalcin/metabolism , Osteogenesis , Regenerative Medicine , Surface Properties , Tissue Engineering
7.
J Biomed Mater Res A ; 99(2): 307-15, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21858919

ABSTRACT

Collagen/hydroxyapatite (HA) composite scaffolds are known to be suitable scaffolds for seeding with mesenchymal stem cells (MSCs) differentiated into osteoblasts and for the in vitro production of artificial bones. However, the optimal collagen/HA ratio remains unclear. Our study confirmed that a higher collagen content increased scaffold stiffness but that a greater stiffness was not sufficient for bone tissue formation, a complex process evidently also dependent on scaffold porosity. We found that the scaffold pore diameter was dependent on the concentration of collagen and HA and that it could play a key role in cell seeding. In conclusion, the optimal scaffold for new bone formation and cell proliferation was found to be a composite scaffold formed from 50 wt % HA in 0.5 wt % collagen I solution.


Subject(s)
Cell Differentiation/physiology , Collagen/chemistry , Extracellular Matrix/chemistry , Hydroxyapatites/chemistry , Mesenchymal Stem Cells/physiology , Osteoblasts/physiology , Tissue Scaffolds/chemistry , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Biomarkers/metabolism , Cattle , Cell Adhesion , Cell Proliferation , Collagen/metabolism , Elastic Modulus , Extracellular Matrix/metabolism , Humans , Hydroxyapatites/metabolism , Materials Testing , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Porosity
8.
Cell Prolif ; 44(2): 183-91, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21401760

ABSTRACT

OBJECTIVES: The aim of this study was to develop functionalized nanofibres as a simple delivery system for growth factors (GFs) and make nanofibre cell-seeded scaffold implants a one-step intervention. MATERIALS AND METHODS: We have functionalized polycaprolactone (PCL) nanofibres with thrombocytes adherent on them. Immobilized, these thrombocytes attached to nanofibre scaffolds were used as a nanoscale delivery system for native (autologous) proliferation and differentiation factors, in vitro. Pig chondrocytes were seeded on the thrombocyte-coated scaffolds and levels of proliferation and differentiation of these cells were compared with those seeded on non-coated scaffolds. RESULTS: Immobilized thrombocytes on PCL nanofibres effectively enhanced chondrocyte proliferation due to time-dependent degradation of thrombocytes and release of their GFs. CONCLUSIONS: These simply functionalized scaffolds present new possibilities for nanofibre applications, as smart cell scaffolds equipped with a GF delivery tool.


Subject(s)
Blood Platelets/metabolism , Chondrocytes/cytology , Nanofibers/chemistry , Polyesters/chemistry , Animals , Blood Platelets/cytology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cells, Immobilized/metabolism , Drug Carriers/chemistry , Intercellular Signaling Peptides and Proteins/administration & dosage , Swine
9.
Physiol Res ; 59(5): 773-781, 2010.
Article in English | MEDLINE | ID: mdl-20406034

ABSTRACT

Non-woven textile mesh from polyglycolic acid (PGA) was found as a proper material for chondrocyte adhesion but worse for their proliferation. Neither hyaluronic acid nor chitosan nor polyvinyl alcohol (PVA) increased chondrocyte adhesion. However, chondrocyte proliferation suffered from acidic byproducts of PGA degradation. However, the addition of PVA and/or chitosan into a wet-laid non-woven textile mesh from PGA improved chondrocyte proliferation seeded in vitro on the PGA-based composite scaffold namely due to a diminished acidification of their microenvironment. This PVA/PGA composite mesh used in combination with a proper hydrogel minimized the negative effect of PGA degradation without dropping positive parameters of the PGA wet-laid non-woven textile mesh. In fact, presence of PVA and/or chitosan in the PGA-based wet-laid non-woven textile mesh even advanced the PGA-based wet-laid non-woven textile mesh for chondrocyte seeding and artificial cartilage production due to a positive effect of PVA in such a scaffold on chondrocyte proliferation.


Subject(s)
Chondrocytes/cytology , Polyglycolic Acid , Polyvinyl Alcohol , Tissue Culture Techniques/methods , Tissue Scaffolds , Animals , Cartilage/cytology , Cell Adhesion , Cell Division , Hyaluronic Acid , Hydrogel, Polyethylene Glycol Dimethacrylate , Microscopy, Confocal , Rabbits , Textiles , Water
10.
Physiol Res ; 56 Suppl 1: S61-S68, 2007.
Article in English | MEDLINE | ID: mdl-17552893

ABSTRACT

The present article introduces a novel method of characterizing the macromechanical cartilage properties based on dynamic testing. The proposed approach of instrumented impact testing shows the possibility of more detailed investigation of the acting dynamic forces and corresponding deformations within the wide range of strain rates and loads, including the unloading part of stress-strain curves and hysteresis loops. The presented results of the unconfined compression testing of both the native joint cartilage tissues and potential substitute materials outlined the opportunity to measure the dissipation energy and thus to identify the initial mechanical deterioration symptoms and to introduce a better definition of material damage. Based on the analysis of measured specimen deformation, the intact and pathologically changed cartilage tissue can be distinguished and the differences revealed.


Subject(s)
Cartilage, Articular/physiology , Materials Testing , Tissue Engineering , Tissue Scaffolds , Animals , Cells, Cultured , Chondrocytes/physiology , Compressive Strength , Elasticity , Humans , Models, Biological , Prosthesis Failure , Stress, Mechanical , Time Factors
11.
Physiol Res ; 56 Suppl 1: S5-S16, 2007.
Article in English | MEDLINE | ID: mdl-17552900

ABSTRACT

The potential of novel scaffold containing sodium hyaluronate, type I collagen, and fibrin was investigated in the regeneration of osteochondral defects in miniature pigs. Both autologous chondrocyte-seeded scaffolds and non-seeded scaffolds were implanted into two defects located in the non-weight-bearing zone of the femoral trochlea (defect A was located more distally and medially, defect B was located more proximally and laterally). Control defects were left untreated. Twelve weeks after the operation, the knees were evaluated in vivo using MRI. Six months after the implantation, the defects were analyzed using MRI, histological, and immunohistochemical analysis. In the A defects of chondrocyte-seeded scaffold group, hyaline cartilage and fibrocartilage was formed, containing type II collagen, acidic and neutral glycosaminoglycans while the non-seeded scaffold group was predominantly filled with fibrocartilage. Defects in the control group were predominantly filled with fibrous tissue. Histomorphometric analysis of photomicrographs revealed a significantly higher amount of hyaline cartilage in the cell-seeded scaffold group in A defects than in other groups. Both scaffold groups in A defects showed significantly less fibrous tissue than cell-seeded defects B and the control group. Both histological and MRI analysis proved that the novel composite scaffold has a potential to regenerate osteochondral defects within six months.


Subject(s)
Biocompatible Materials , Cartilage Diseases/surgery , Chondrocytes/transplantation , Collagen Type I/chemistry , Fibrin/chemistry , Hyaluronic Acid/chemistry , Stifle/surgery , Tissue Scaffolds , Animals , Cartilage Diseases/metabolism , Cartilage Diseases/pathology , Cartilage Diseases/physiopathology , Cells, Cultured , Chondrocytes/metabolism , Collagen Type II/metabolism , Disease Models, Animal , Fibrocartilage/metabolism , Fibrocartilage/pathology , Fibrocartilage/surgery , Glycosaminoglycans/metabolism , Hyaline Cartilage/metabolism , Hyaline Cartilage/pathology , Hyaline Cartilage/surgery , Immunohistochemistry , Magnetic Resonance Imaging , Stifle/metabolism , Stifle/pathology , Stifle/physiopathology , Swine , Swine, Miniature , Time Factors , Tissue Engineering
SELECTION OF CITATIONS
SEARCH DETAIL
...