Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Biomed Mater Res B Appl Biomater ; 110(8): 1862-1875, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35233920

RESUMO

Bioreactors have been used for bone graft engineering in pre-clinical investigations over the past 15 years. The ability of bioreactor-incubated bone marrow nuclear cells (BMNCs) to enhance bone-forming potential varies significantly, and the three-dimensional (3D) distribution of BMNCs within the scaffold is largely unknown. The aims of this study were (1) to investigate the efficacy of a carbonated hydroxyapatite (CHA) with/without BMNCs on spine fusion rate and fusion mass microarchitecture using a highly challenging two-level posterolateral spine fusion without instrumentation; and (2) to evaluate 3D distribution of BMNCs within scaffolds characterized by immunohistochemistry. Fusion rate and fusion mass were quantified by micro-CT, microarchitectural analysis, and histology. While the homogenous 3D distribution of BMNCs was not observed, BMNCs were found to migrate towards a substitute core. In the autograft group, the healing rate was 83.3%, irrespective of the presence of BMNCs. In the CHA group, also 83.3% was fused in the presence of BMNCs, and 66.7% fused without BMNCs. A significant decrease in the fusion mass porosity (p = .001) of the CHA group suggested the deposition of mineralized bone. The autograft group revealed more bone, thicker trabeculae, and better trabecular orientation but less connection compared to the CHA group. Immunohistochemistry confirmed the ability of bioreactors to incubate a large-sized substitute coated with viable BMNCs with the potential for proliferation and differentiation. These findings suggested that a bioreactor-activated substitute is comparable to autograft on spine fusion and that new functional bone regeneration could be achieved by a combination of BMNCs, biomaterials, and bioreactors.


Assuntos
Substitutos Ósseos , Fusão Vertebral , Animais , Reatores Biológicos , Medula Óssea , Células da Medula Óssea , Substitutos Ósseos/química , Substitutos Ósseos/farmacologia , Transplante Ósseo/métodos , Ovinos , Fusão Vertebral/métodos
2.
Biosens Bioelectron ; 166: 112467, 2020 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-32805618

RESUMO

Nasal chondrocyte-derived engineered cartilage has been demonstrated to be safe and feasible for the treatment of focal cartilage lesions with promising preliminary evidences of efficacy. To ensure the quality of the products and processes, and to meet regulatory requirements, quality controls for identity, purity, and potency need to be developed. We investigated the use of Raman spectroscopy, a nondestructive analytical method that measures the chemical composition of samples, and statistical learning methods for the development of quality controls to quantitatively characterize the starting biopsy and final grafts. We provide a proof-of-concept to show how Raman spectroscopy can be used to identify the types of tissues found in a nasal septal biopsy, i.e., hyaline cartilage and perichondrium, for a novel tissue identity assay. The tissues could be classified with a sensitivity of 89% and specificity of 77%. We also show how clinically relevant and mature nasal chondrocyte-derived engineered cartilage can be assessed with Raman spectroscopy for the development of potency assays. The maturity of engineered grafts, based on the quantified ratio of glycosaminoglycans to DNA and histological score, could be accurately assessed (R2 = 0.78 and 0.89, respectively, between predicted and measured values). Our results demonstrate the potential of Raman spectroscopy for the development of characterization assays for regenerative therapies that could be integrated into a good manufacturing practice-compliant process.


Assuntos
Técnicas Biossensoriais , Análise Espectral Raman , Cartilagem , Condrócitos , Engenharia Tecidual
3.
Cell Prolif ; 52(6): e12653, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31489992

RESUMO

OBJECTIVES: Bioreactor-based production systems have the potential to overcome limitations associated with conventional tissue engineering manufacturing methods, facilitating regulatory compliant and cost-effective production of engineered grafts for widespread clinical use. In this work, we established a bioreactor-based manufacturing system for the production of cartilage grafts. MATERIALS & METHODS: All bioprocesses, from cartilage biopsy digestion through the generation of engineered grafts, were performed in our bioreactor-based manufacturing system. All bioreactor technologies and cartilage tissue engineering bioprocesses were transferred to an independent GMP facility, where engineered grafts were manufactured for two large animal studies. RESULTS: The results of these studies demonstrate the safety and feasibility of the bioreactor-based manufacturing approach. Moreover, grafts produced in the manufacturing system were first shown to accelerate the repair of acute osteochondral defects, compared to cell-free scaffold implants. We then demonstrated that grafts produced in the system also facilitated faster repair in a more clinically relevant chronic defect model. Our data also suggested that bioreactor-manufactured grafts may result in a more robust repair in the longer term. CONCLUSION: By demonstrating the safety and efficacy of bioreactor-generated grafts in two large animal models, this work represents a pivotal step towards implementing the bioreactor-based manufacturing system for the production of human cartilage grafts for clinical applications. Read the Editorial for this article on doi:10.1111/cpr.12625.


Assuntos
Reatores Biológicos , Condrócitos/citologia , Engenharia Tecidual , Alicerces Teciduais , Doença Aguda , Animais , Cartilagem Articular/patologia , Doença Crônica , Feminino , Modelos Animais , Ovinos , Engenharia Tecidual/métodos
4.
Biotechnol J ; 12(12)2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28881093

RESUMO

Bone marrow-derived mesenchymal stromal cells (BMSC), when expanded directly within 3D ceramic scaffolds in perfusion bioreactors, more reproducibly form bone when implanted in vivo as compared to conventional expansion on 2D polystyrene dishes/flasks. Since the bioreactor-based expansion on 3D ceramic scaffolds encompasses multiple aspects that are inherently different from expansion on 2D polystyrene, we aimed to decouple the effects of specific parameters among these two model systems. We assessed the effects of the: 1) 3D scaffold vs. 2D surface; 2) ceramic vs. polystyrene materials; and 3) BMSC niche established within the ceramic pores during in vitro culture, on subsequent in vivo bone formation. While BMSC expanded on 3D polystyrene scaffolds in the bioreactor could maintain their in vivo osteogenic potential, results were similar as BMSC expanded in monolayer on 2D polystyrene, suggesting little influence of the scaffold 3D environment. Bone formation was most reproducible when BMSC are expanded on 3D ceramic, highlighting the influence of the ceramic substrate. The presence of a pre-formed niche within the scaffold pores had negligible effects on the in vivo bone formation. The results of this study allow a greater understanding of the parameters required for perfusion bioreactor-based manufacturing of osteogenic grafts for clinical applications.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Cultura de Células/instrumentação , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Alicerces Teciduais , Adolescente , Adulto , Reatores Biológicos , Técnicas de Cultura de Células/métodos , Proliferação de Células , Cerâmica/química , Humanos , Pessoa de Meia-Idade , Perfusão , Adulto Jovem
5.
J Biomed Mater Res B Appl Biomater ; 104(3): 532-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25952142

RESUMO

A computer-controlled perfusion bioreactor was developed for the streamlined production of engineered osteogenic grafts. This system automated the required bioprocesses, from the initial filling of the system through the phases of cell seeding and prolonged cell/tissue culture. Flow through chemo-optic micro-sensors allowed to non-invasively monitor the levels of oxygen and pH in the perfused culture medium throughout the culture period. To validate its performance, freshly isolated ovine bone marrow stromal cells were directly seeded on porous scaffold granules (hydroxyapatite/ß-tricalcium-phosphate/poly-lactic acid), bypassing the phase of monolayer cell expansion in flasks. Either 10 or 20 days after culture, engineered cell-granule grafts were implanted in an ectopic mouse model to quantify new bone formation. After four weeks of implantation, histomorphometry showed more bone in bioreactor-generated grafts than cell-free granule controls, while bone formation did not show significant differences between 10 days and 20 days of incubation. The implanted granules without cells had no bone formation. This novel perfusion bioreactor has revealed the capability of activation larger viable bone graft material, even after shorter incubation time of graft material. This study has demonstrated the feasibility of engineering osteogenic grafts in an automated bioreactor system, laying the foundation for a safe, regulatory-compliant, and cost-effective manufacturing process.


Assuntos
Reatores Biológicos , Células da Medula Óssea/metabolismo , Substitutos Ósseos/química , Fosfatos de Cálcio/química , Durapatita/química , Osteogênese , Poliésteres/química , Engenharia Tecidual , Animais , Células da Medula Óssea/citologia , Feminino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Ovinos
6.
J Tissue Eng Regen Med ; 9(12): 1394-403, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23225781

RESUMO

Co-culture of mesenchymal stromal cells (MSCs) with articular chondrocytes (ACs) has been reported to improve the efficiency of utilization of a small number of ACs for the engineering of implantable cartilaginous tissues. However, the use of cells of animal origin and the generation of small-scale micromass tissues limit the clinical relevance of previous studies. Here we investigated the in vitro and in vivo chondrogenic capacities of scaffold-based constructs generated by combining primary human ACs with human bone marrow MSCs (BM-MSCs). The two cell types were cultured in collagen sponges (2 × 6 mm disks) at the BM-MSCs:ACs ratios: 100:0, 95:5, 75:25 and 0:100 for 3 weeks. Scaffolds freshly seeded or further precultured in vitro for 2 weeks were also implanted subcutaneously in nude mice and harvested after 8 or 6 weeks, respectively. Static co-culture of ACs (25%) with BM-MSCs (75%) in scaffolds resulted in up to 1.4-fold higher glycosaminoglycan (GAG) content than what would be expected based on the relative percentages of the different cell types. In vivo GAG induction was drastically enhanced by the in vitro preculture and maximal at the ratio 95:5 (3.8-fold higher). Immunostaining analyses revealed enhanced accumulation of type II collagen and reduced accumulation of type X collagen with increasing ACs percentage. Constructs generated in the perfusion bioreactor system were homogeneously cellularized. In summary, human cartilage grafts were successfully generated, culturing BM-MSCs with a relatively low fraction of non-expanded ACs in porous scaffolds. The proposed co-culture strategy is directly relevant towards a single-stage surgical procedure for cartilage repair.


Assuntos
Células da Medula Óssea/metabolismo , Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Engenharia Tecidual/métodos , Adulto , Idoso , Animais , Células da Medula Óssea/citologia , Cartilagem Articular/citologia , Condrócitos/citologia , Condrócitos/transplante , Técnicas de Cocultura , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Células Estromais/citologia , Células Estromais/metabolismo
7.
J Tissue Eng ; 5: 2041731414540674, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25383165

RESUMO

While calcium phosphate-based ceramics are currently the most widely used materials in bone repair, they generally lack tensile strength for initial load bearing. Bulk titanium is the gold standard of metallic implant materials, but does not match the mechanical properties of the surrounding bone, potentially leading to problems of fixation and bone resorption. As an alternative, nickel-titanium alloys possess a unique combination of mechanical properties including a relatively low elastic modulus, pseudoelasticity, and high damping capacity, matching the properties of bone better than any other metallic material. With the ultimate goal of fabricating porous implants for spinal, orthopedic and dental applications, nickel-titanium substrates were fabricated by means of selective laser melting. The response of human mesenchymal stromal cells to the nickel-titanium substrates was compared to mesenchymal stromal cells cultured on clinically used titanium. Selective laser melted titanium as well as surface-treated nickel-titanium and titanium served as controls. Mesenchymal stromal cells had similar proliferation rates when cultured on selective laser melted nickel-titanium, clinically used titanium, or controls. Osteogenic differentiation was similar for mesenchymal stromal cells cultured on the selected materials, as indicated by similar gene expression levels of bone sialoprotein and osteocalcin. Mesenchymal stromal cells seeded and cultured on porous three-dimensional selective laser melted nickel-titanium scaffolds homogeneously colonized the scaffold, and following osteogenic induction, filled the scaffold's pore volume with extracellular matrix. The combination of bone-related mechanical properties of selective laser melted nickel-titanium with its cytocompatibility and support of osteogenic differentiation of mesenchymal stromal cells highlights its potential as a superior bone substitute as compared to clinically used titanium.

8.
PLoS One ; 9(7): e102359, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25020062

RESUMO

Mesenchymal stromal/stem cell (MSC) expansion in conventional monolayer culture on plastic dishes (2D) leads to progressive loss of functionality and thus challenges fundamental studies on the physiology of skeletal progenitors, as well as translational applications for cellular therapy and molecular medicine. Here we demonstrate that 2D MSC expansion can be entirely bypassed by culturing freshly isolated bone marrow nucleated cells within 3D porous scaffolds in a perfusion-based bioreactor system. The 3D-perfusion system generated a stromal tissue that could be enzymatically treated to yield CD45- MSC. As compared to 2D-expanded MSC (control), those derived from 3D-perfusion culture after the same time (3 weeks) or a similar extent of proliferation (7-8 doublings) better maintained their progenitor properties, as assessed by a 4.3-fold higher clonogenicity and the superior differentiation capacity towards all typical mesenchymal lineages. Transcriptomic analysis of MSC from 5 donors validated the robustness of the process and indicated a reduced inter-donor variability and a significant upregulation of multipotency-related gene clusters following 3D-perfusion--as compared to 2D-expansion. Interestingly, the differences in functionality and transcriptomics between MSC expanded in 2D or under 3D-perfusion were only partially captured by cytofluorimetric analysis using conventional surface markers. The described system offers a multidisciplinary approach to study how factors of a 3D engineered niche regulate MSC function and, by streamlining conventional labor-intensive processes, is prone to automation and scalability within closed bioreactor systems.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Mesenquimais/citologia , Perfusão/métodos , Reatores Biológicos , Técnicas de Cultura de Células/instrumentação , Proliferação de Células , Separação Celular , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Perfusão/instrumentação , Fenótipo
9.
Eur Cell Mater ; 24: 224-36, 2012 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-23007908

RESUMO

Inflammatory cytokines present in the milieu of the fracture site are important modulators of bone healing. Here we investigated the effects of interleukin-1ß (IL-1ß) on the main events of endochondral bone formation by human bone marrow mesenchymal stromal cells (BM-MSC), namely cell proliferation, differentiation and maturation/remodelling of the resulting hypertrophic cartilage. Low doses of IL-1ß (50 pg/mL) enhanced colony-forming units-fibroblastic (CFU-f) and -osteoblastic (CFU-o) number (up to 1.5-fold) and size (1.2-fold) in the absence of further supplements and glycosaminoglycan accumulation (1.4-fold) upon BM-MSC chondrogenic induction. In osteogenically cultured BM-MSC, IL-1ß enhanced calcium deposition (62.2-fold) and BMP-2 mRNA expression by differential activation of NF-κB and ERK signalling. IL-1ß-treatment of BM-MSC generated cartilage resulted in higher production of MMP-13 (14.0-fold) in vitro, mirrored by an increased accumulation of the cryptic cleaved fragment of aggrecan, and more efficient cartilage remodelling/resorption after 5 weeks in vivo (i.e., more TRAP positive cells and bone marrow, less cartilaginous areas), resulting in the formation of mature bone and bone marrow after 12 weeks. In conclusion, IL-1ß finely modulates early and late events of the endochondral bone formation by BM-MSC. Controlling the inflammatory environment could enhance the success of therapeutic approaches for the treatment of fractures by resident MSC and as well as improve the engineering of implantable tissues.


Assuntos
Condrogênese , Interleucina-1beta/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Adulto , Animais , Proteína Morfogenética Óssea 2/genética , Cálcio/metabolismo , Cartilagem/crescimento & desenvolvimento , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Masculino , Metaloproteinase 13 da Matriz/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Pessoa de Meia-Idade , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , RNA Mensageiro/biossíntese
10.
Eur Spine J ; 21(9): 1740-7, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22777077

RESUMO

PURPOSE: To evaluate the effect of a large perfusion-bioreactor cell-activated bone substitute, on a two-level large posterolateral spine fusion sheep model. METHODS: A 50 mm long porous biphasic-calcium-phosphate bone substitute reinforced with poly(D,L-lactide) and, activated with bone marrow derived mononuclear-cells (BMNC) was used. Eighteen sheep were divided into two groups and one group (n = 9) had BMNC-activated bone substitutes and cell-free substitutes implanted. The second group (n = 9) had autograft supplemented with BMNC and regular autograft implanted. The implant material was alternated between spine level L2-L3 and L4-L5 in both groups. MicroCT was used to compare the spine fusion efficacy and bone structure of the two groups as well as the implanted bone substitutes and non-implanted substitutes. RESULTS: After 4½ months six sheep survived in both groups and we found five spine levels were fused when using activated bone substitute compared to three levels with cell-free bone substitute (p = 0.25). Five sheep fused at both levels in the autograft group. A significant increased bone density (p < 0.05) and anisotropy (p < 0.05) was found in the group of activated bone substitutes compared to cell-free bone substitute and no difference existed on the other parameters. The implanted bone substitutes had a significant higher bone density and trabecular thickness than non-implanted bone substitutes, thus indicating that the PLA reinforced BCP had osteoconductive properties (p < 0.05). No effect of the supplemented BMNC to autograft was observed. The autograft group had a significant higher bone density, trabecular thickness and degree of anisotropy than the implanted bone substitutes (p < 0.05), but a lower connectivity density existed (p < 0.05). This indicates that though the activated substitute might have a similar fusion efficacy to autograft, the fusion bridge is not of equal substance. CONCLUSION: We found that bioreactor-generated cell-based bone substitutes seemed superior in fusion ability when compared to cell-free bone substitute and comparable to autograft in fusion ability, but not in bone structure. This combined with the favorable biocompatible abilities and strength comparable to human cancellous bone indicates that it might be a suitable bone substitute in spine fusion procedures.


Assuntos
Reatores Biológicos , Substitutos Ósseos/uso terapêutico , Leucócitos Mononucleares/transplante , Vértebras Lombares/diagnóstico por imagem , Fusão Vertebral/métodos , Animais , Células da Medula Óssea , Substitutos Ósseos/química , Fosfatos de Cálcio/uso terapêutico , Durapatita/uso terapêutico , Feminino , Vértebras Lombares/cirurgia , Poliésteres/uso terapêutico , Ovinos , Tomografia Computadorizada por Raios X
11.
Biomaterials ; 33(20): 5085-93, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22510434

RESUMO

Materials based on synthetic polymers can be extensively tailored in their physical properties but often suffer from limited biological functionality. Here we tested the hypothesis that the biological performance of 3D synthetic polymer-based scaffolds can be enhanced by extracellular matrix (ECM) deposited by cells in vitro and subsequently decellularized. The hypothesis was tested in the context of bone graft substitutes, using polyesterurethane (PEU) foams and mineralized ECM laid by human mesenchymal stromal cells (hMSC). A perfusion-based bioreactor system was critically employed to uniformly seed and culture hMSC in the scaffolds and to efficiently decellularize (94% DNA reduction) the resulting ECM while preserving its main organic and inorganic components. As compared to plain PEU, the decellularized ECM-polymer hybrids supported the osteoblastic differentiation of newly seeded hMSC by up-regulating the mRNA expression of typical osteoblastic genes (6-fold higher bone sialoprotein; 4-fold higher osteocalcin and osteopontin) and increasing calcium deposition (6-fold higher), approaching the performance of ceramic-based materials. After ectopic implantation in nude mice, the decellularized hybrids induced the formation of a mineralized matrix positively immunostained for bone sialoprotein and resembling an immature osteoid tissue. Our findings consolidate the perspective of bioreactor-based production of ECM-decorated polymeric scaffolds as off-the-shelf materials combining tunable physical properties with the physiological presentation of instructive biological signals.


Assuntos
Materiais Biocompatíveis , Matriz Extracelular , Polímeros , Engenharia Tecidual , Animais , Diferenciação Celular , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Osteoblastos/citologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Biomaterials ; 32(2): 321-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20952054

RESUMO

In the bone marrow, specialized microenvironments, called niches, regulate hematopoietic stem cell (HSC) maintenance and function through a complex crosstalk between different cell types. Although in vivo studies have been instrumental to elucidate some of the mechanisms by which niches exert their function, the establishment of an in vitro model that recapitulates the fundamental interactions of the niche components in a controlled setting would be of great benefit. We have previously shown that freshly harvested bone marrow- or adipose tissue-derived cells can be cultured under perfusion within porous scaffolds, allowing the formation of an organized 3D stromal tissue, composed by mesenchymal and endothelial progenitors and able to support hematopoiesis. Here we describe 3D scaffold-based perfusion systems as potential models to reconstruct ex vivo the bone marrow stem cell niche. We discuss how several culture parameters, including scaffold properties, cellular makeup and molecular signals, can be varied and controlled to investigate the role of specific cues in affecting HSC fate. We then provide a perspective of how the system could be exploited to improve stem cell-based therapies and how the model can be extended toward the engineering of other specialized stromal niches.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Cultura de Células/métodos , Nicho de Células-Tronco/citologia , Alicerces Teciduais , Animais , Humanos
13.
J Cell Mol Med ; 12(4): 1238-49, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18782188

RESUMO

Biological substitutes for autologous bone flaps could be generated by combining flap pre-fabrication and bone tissue engineering concepts. Here, we investigated the pattern of neotissue formation within large pre-fabricated engineered bone flaps in rabbits. Bone marrow stromal cells from 12 New Zealand White rabbits were expanded and uniformly seeded in porous hydroxyapatite scaffolds (tapered cylinders, 10-20 mm diameter, 30 mm height) using a perfusion bioreactor. Autologous cell-scaffold constructs were wrapped in a panniculus carnosus flap, covered by a semipermeable membrane and ectopically implanted. Histological analysis, substantiated by magnetic resonance imaging (MRI) and micro-computerized tomography scans, indicated three distinct zones: an outer one, including bone tissue; a middle zone, formed by fibrous connective tissue; and a central zone, essentially necrotic. The depths of connective tissue and of bone ingrowth were consistent at different construct diameters and significantly increased from respectively 3.1+/-0.7 mm and 1.0+/-0.4 mm at 8 weeks to 3.7+/-0.6 mm and 1.4+/-0.6 mm at 12 weeks. Bone formation was found at a maximum depth of 1.8 mm after 12 weeks. Our findings indicate the feasibility of ectopic pre-fabrication of large cell-based engineered bone flaps and prompt for the implementation of strategies to improve construct vascularization, in order to possibly accelerate bone formation towards the core of the grafts.


Assuntos
Substitutos Ósseos/metabolismo , Osteogênese , Engenharia Tecidual , Alicerces Teciduais , Animais , Células da Medula Óssea/citologia , Osso e Ossos/citologia , Células Cultivadas , Cerâmica , Tecido Conjuntivo , Implantes Experimentais , Imageamento por Ressonância Magnética , Porosidade , Coelhos , Células Estromais/citologia , Fatores de Tempo , Tomografia Computadorizada por Raios X
14.
J Tissue Eng Regen Med ; 1(1): 60-5, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18038393

RESUMO

Reproducible osteogenicity is a key requirement for the clinical use of bone substitutes based on bone marrow stromal cells (BMSCs) and three-dimensional (3D) scaffolds. In this study we addressed whether a minimal cell density is required for ectopic osteogenicity of constructs generated using a recently developed perfusion system for seeding and culturing human BMSCs on 3D scaffolds. Cells from human bone marrow aspirates were directly seeded and expanded for 3 weeks within the pores of ceramic-based scaffolds, using a perfusion bioreactor. The resulting constructs were either implanted subcutaneously in nude mice, to determine their capacity to generate bone tissue, or digested to retrieve the expanded cells and assess their number, phenotype and clonogenic capacity. The final number of BMSCs in the constructs was correlated neither to the initial number of seeded cells, nor to the subsequent bone formation. Instead, the final number of clonogenic BMSCs in the constructs was positively correlated to the initial number of BMSCs seeded, and was significantly higher in osteogenic than in non-osteogenic constructs. These results indicate that clonogenic cells play a crucial role in determining the osteogenicity of engineered bone substitutes. Possible ways to quantify the density of clonogenic cells as a quality control parameter to predict potency of BMSC-based constructs are discussed.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular , Osteogênese , Células Estromais/citologia , Adulto , Idoso , Materiais Biocompatíveis , Células Cultivadas , Humanos , Pessoa de Meia-Idade , Fenótipo
15.
J Biosci Bioeng ; 100(5): 489-94, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16384786

RESUMO

Osteochondral defects (i.e., those that affect both the articular cartilage and underlying subchondral bone) are often associated with mechanical instability of the joint, and therefore with the risk of inducing osteoarthritic degenerative changes. The in vitro fabrication of osteochondral grafts of predefined size and shape, starting from autologous cells combined with three-dimensional porous biomaterials, is a promising approach for the treatment of osteochondral defects. However, the quality of ex vivo generated cartilage and bone-like tissues is currently restricted by a limited understanding of the regulatory role of physicochemical culture parameters on tissue development. By allowing reproducible and controlled changes in specific biochemical and biomechanical factors, bioreactor systems provide the technological means to reveal fundamental mechanisms of cell function in a three-dimensional environment and the potential to improve the quality of engineered tissues. In addition, by automating and standardizing the manufacturing process in controlled closed systems, bioreactors could reduce production costs and thus facilitate broader clinical impact of engineered osteochondral grafts.


Assuntos
Reatores Biológicos , Doenças das Cartilagens/terapia , Cartilagem Articular , Condrogênese/fisiologia , Próteses e Implantes , Animais , Materiais Biocompatíveis , Cartilagem Articular/anormalidades , Condrócitos/citologia , Condrócitos/fisiologia , Humanos , Osteoblastos/citologia , Osteoblastos/fisiologia , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos , Transplante Autólogo
16.
Stem Cells ; 23(8): 1066-72, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16002780

RESUMO

Three-dimensional (3D) culture systems are critical to investigate cell physiology and to engineer tissue grafts. In this study, we describe a simple yet innovative bioreactor-based approach to seed, expand, and differentiate bone marrow stromal cells (BMSCs) directly in a 3D environment, bypassing the conventional process of monolayer (two-dimensional [2D]) expansion. The system, based on the perfusion of bone marrow-nucleated cells through porous 3D scaffolds, supported the formation of stromal-like tissues, where BMSCs could be cocultured with hematopoietic progenitor cells in proportions dependent on the specific medium supplements. The resulting engineered constructs, when implanted ectopically in nude mice, generated bone tissue more reproducibly, uniformly, and extensively than scaffolds loaded with 2D-expanded BMSCs. The developed system may thus be used as a 3D in vitro model of bone marrow to study interactions between BMSCs and hematopoietic cells as well as to streamline manufacture of osteoinductive grafts in the context of regenerative medicine.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Cultura de Células , Osteogênese , Adulto , Animais , Células da Medula Óssea/ultraestrutura , Proliferação de Células , Células Cultivadas , Meios de Cultura/química , Durapatita , Células-Tronco Hematopoéticas/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Perfusão , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA