Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Pediatr Otorhinolaryngol ; 176: 111807, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38134588

RESUMO

OBJECTIVE: The aim of this study was to assess the in vivo efficacy of a novel regenerative collagen-based scaffold developed by the Royal College of Surgeons in Ireland in a chronic tympanic membrane perforation (TMP) using a chinchilla model. METHODS: Bilateral TMPs were induced in 17 mixed gender chinchillas using tympanic membrane resection followed by a mixture of topical Mitomycin C and dexamethasone for 3 days. These were monitored with weekly otoscopy for 8 weeks. Animals were excluded if signs of infection developed in the follow up period (n = 8). At 8 weeks, intervention began and 18 TMPs were assigned to either treatment with the collagen-based scaffold (treated group) or spontaneous healing (control group). Animals were euthanized 6 weeks post-intervention. Otoscopic imaging and auditory brain response (ABR) were conducted at baseline, 8 weeks post-TMP induction and 6 weeks post-intervention. All TMPs were then evaluated at 6 weeks post-intervention and bullae underwent histologic evaluation. RESULTS: At 6 weeks post-intervention, otoscopic imaging demonstrated various degrees of healing in the treated ears. The treated group was noted to have an increased rate of healing when compared to the control group. Histologic evaluation demonstrated a variation in the degree of perforation healing within groups, with some animals in the treated group showing high levels of perforation healing. At 8 weeks after the TMP procedure, most of the animals had worsened hearing response. At 6-week post the collagen-based scaffold treatment, about 50 % (4/8) of the treated ears had improved in hearing response as compared to those of non-treated ears. CONCLUSION: Given the initial histologic evidence of partial healing in scaffold-treated ears, the post-intervention period should be extended to monitor the potential for complete healing. Given the overall positive findings related to healing with the scaffold-treated ears, this material warrants further investigation.


Assuntos
Perfuração da Membrana Timpânica , Humanos , Animais , Perfuração da Membrana Timpânica/cirurgia , Perfuração da Membrana Timpânica/patologia , Cicatrização , Membrana Timpânica/patologia , Colágeno , Mitomicina/farmacologia
2.
Acta Biomater ; 162: 164-181, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36967054

RESUMO

Despite the glimmer of hope provided by the discovery and commercialization of bone morphogenetic protein-2 (BMP-2) as a bone graft substitute, side effects related to the use of supraphysiological doses have hindered its clinical usage. In this study, we compared the osteoinductive potential of BMP-2 homodimer with a heterodimer of BMP-2/7, both delivered via a collagen-hydroxyapatite (CHA) scaffold delivery system, with the aim to reduce the overall therapeutic BMP doses and the associated side-effects. We first show that the incorporation of hydroxyapatite in collagen-based BMP delivery systems is pivotal for achieving efficient BMP sequestration and controlled release. Using an ectopic implantation model, we then showed that the CHA+BMP-2/7 was more osteoinductive than CHA+BMP-2. Further evaluation of the molecular mechanisms responsible for this increased osteoinductivity at an early stage in the regeneration process indicated that the CHA+BMP-2/7 enhanced progenitor cell homing at the implantation site, upregulated the key transcriptomic determinants of bone formation, and increased the production of bone extracellular matrix components. Using fluorescently labelled BMP-2/7 and BMP-2, we demonstrated that the CHA scaffold provided a long-term delivery of both molecules for at least 20 days. Finally, using a rat femoral defect model, we showed that an ultra-low dose (0.5 µg) of BMP-2/7 accelerated fracture healing and performed at a level comparable to 20-times higher BMP-2 dose. Our results indicate that the sustained delivery of BMP-2/7 via a CHA scaffold could bring us a step closer in the quest for the use of physiological growth factor doses in fracture healing. STATEMENT OF SIGNIFICANCE: • Incorporation of hydroxyapatite (HA) in a collagen scaffold dramatically improves bone morphogenic protein (BMP) sequestration via biophysical interactions with BMP, thereby providing more controlled BMP release compared with pristine collagen. • We then investigate the molecular mechanisms responsible for increased osteoinductive potential of a heterodimer BMP-2/7 with is clinically used counterpart, the BMP-2 homodimer. • The superior osteoinductive properties of BMP-2/7 are a consequence of its direct positive effect on progenitor cell homing at the implantation site, which consequently leads to upregulation of cartilage and bone related genes and biochemical markers. • An ultra-low dose of BMP-2/7 delivered via a collagen-HA (CHA) scaffold leads to accelerated healing of a critical femoral defect in rats while a 20-times higher BMP-2 dose was required to achieve comparable results.


Assuntos
Substitutos Ósseos , Durapatita , Ratos , Animais , Durapatita/farmacologia , Colágeno/farmacologia , Colágeno/química , Osteogênese , Osso e Ossos , Consolidação da Fratura , Substitutos Ósseos/farmacologia , Proteína Morfogenética Óssea 2/farmacologia , Proteína Morfogenética Óssea 2/química , Regeneração Óssea
3.
Biomater Biosyst ; 8: 100066, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36824377

RESUMO

Cartilage has poor regenerative capacity and thus damage to the joint surfaces presents a major clinical challenge. Recent research has focussed on the development of tissue-engineered and cell-based approaches for the treatment of cartilage and osteochondral injuries, with current clinically available cell-based approaches including autologous chondrocyte implantation and matrix-assisted autologous chondrocyte implantation. However, these approaches have significant disadvantages due to the requirement for a two-stage surgical procedure and an in vitro chondrocyte expansion phase which increases logistical challenges, hospital times and costs. In this study, we hypothesized that seeding biomimetic tri-layered scaffolds, with proven regenerative potential, with chondrocyte/infrapatellar fat pad stromal cell co-cultures would improve their regenerative capacity compared to scaffolds implanted cell-free. Rapid cell isolation techniques, without the requirement for long term in vitro culture, were utilised to achieve co-cultures of chondrocytes and stromal cells and thus overcome the limitations of existing cell-based techniques. Cell-free and cell-seeded scaffolds were implanted in osteochondral defects, created within the femoral condyle and trochlear ridge, in a translational large animal goat model. While analysis showed trends towards delayed subchondral bone healing in the cell-seeded scaffold group, by the 12 month timepoint the cell-free and cell-seeded groups yield cartilage and bone tissue with comparable quality and quantity. The results of the study reinforce the potential of the biomimetic tri-layered scaffold to repair joint defects but failed to demonstrate a clear benefit from the addition of the CC/FPMSC co-culture to this scaffold. Taking into consideration the additional cost and complexity associated with the cell-seeded scaffold approach, this study demonstrates that the treatment of osteochondral defects using cell-free tri-layered scaffolds may represent a more prudent clinical approach.

4.
J Control Release ; 334: 96-105, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33811984

RESUMO

Leveraging the differential response of genes to mechanical loading may allow for the identification of novel therapeutics and we have recently established placental growth factor (PGF) as a mechanically augmented gene which promotes angiogenesis at higher doses and osteogenesis at lower doses. Herein, we sought to execute a mechanobiology-informed approach to regenerative medicine by designing a functionalized scaffold for the dose-controlled delivery of PGF which we hypothesized would be capable of promoting regeneration of critically-sized bone defects. Alginate microparticles and collagen/hydroxyapatite scaffolds were shown to be effective PGF-delivery platforms, as demonstrated by their capacity to promote angiogenesis in vitro. A PGF release profile consisting of an initial burst release to promote angiogenesis followed by a lower sustained release to promote osteogenesis was achieved by incorporating PGF-loaded microparticles into a collagen/hydroxyapatite scaffold already containing directly incorporated PGF. Although this PGF-functionalized scaffold demonstrated only a modest increase in osteogenic capacity in vitro, robust bone regeneration was observed after implantation into rat calvarial defects, indicating that the dose-dependent effect of PGF can be harnessed as an alternative to multi-drug systems for the delivery of both pro-angiogenic and pro-osteogenic cues. This mechanobiology-informed approach provides a framework for strategies aimed at identifying and evaluating novel scaffold-based systems for regenerative applications.


Assuntos
Medicina Regenerativa , Alicerces Teciduais , Animais , Biofísica , Regeneração Óssea , Colágeno , Preparações de Ação Retardada , Feminino , Osteogênese , Fator de Crescimento Placentário , Ratos , Ratos Wistar
5.
J Mech Behav Biomed Mater ; 118: 104445, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33740688

RESUMO

Extracellular matrix (ECM)-derived scaffolds have shown promise as tissue-engineered grafts for promoting cartilage repair. However, there has been a lack of focus on fine-tuning the frictional properties of scaffolds for cartilage tissue engineering as well as understanding their interactions with synovial fluid constituents. Proteoglycan-4 (PRG4) and hyaluronan (HA) are macromolecules within synovial fluid that play key roles as boundary mode lubricants during cartilage surface interactions. The overall objective of this study was to characterize the role PRG4 and HA play in the lubricating function of collagen-glycosaminoglycan (GAG) scaffolds for cartilage repair. As a first step towards this goal, we aimed to develop a suitable in vitro friction test to establish the boundary mode lubrication parameters for collagen-GAG scaffolds articulated against glass in a phosphate buffered saline (PBS) bath. Subsequently, we sought to leverage this system to determine the effect of physiological synovial fluid lubricants, PRG4 and HA, on the frictional properties of collagen-GAG scaffolds, with scaffolds hydrated in PBS and bovine synovial fluid (bSF) serving as negative and positive controls, respectively. At all compressive strains examined (ε = 0.1-0.5), fluid depressurization within hydrated collagen-GAG scaffolds was >99% complete at ½ minute. The coefficient of friction was stable at all compressive strains (ranging from a low 0.103 ± 0.010 at ε = 0.3 up to 0.121 ± 0.015 at ε = 0.4) and indicative of boundary-mode conditions. Immunohistochemistry demonstrated that PRG4 from recombinant human (rh) and bovine sources adsorbed to collagen-GAG scaffolds and the coefficient of friction for scaffolds immersed in rhPRG4 (0.067 ± 0.027) and normal bSF (0.056 ± 0.020) solution decreased compared to PBS (0.118 ± 0.21, both p < 0.05, at ε = 0.2). The ability of the adsorbed rhPRG4 to reduce friction on the scaffolds indicates that its incorporation within collagen-GAG biomaterials may enhance their lubricating ability as potential tissue-engineered cartilage replacements. To conclude, this study reports the development of an in vitro friction test capable of characterizing the coefficient of friction of ECM-derived scaffolds tested in a range of synovial fluid lubricants and demonstrates frictional properties as a potential design parameter for implants and materials for soft tissue replacement.


Assuntos
Cartilagem Articular , Líquido Sinovial , Alicerces Teciduais , Animais , Bovinos , Colágeno , Fricção , Glicosaminoglicanos , Humanos , Ácido Hialurônico , Lubrificação , Proteoglicanas
6.
Acta Biomater ; 113: 130-143, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32505800

RESUMO

Successful osteochondral defect repair requires regenerating the subchondral bone whilst simultaneously promoting the development of an overlying layer of articular cartilage that is resistant to vascularization and endochondral ossification. During skeletal development articular cartilage also functions as a surface growth plate, which postnatally is replaced by a more spatially complex bone-cartilage interface. Motivated by this developmental process, the hypothesis of this study is that bi-phasic, fibre-reinforced cartilaginous templates can regenerate both the articular cartilage and subchondral bone within osteochondral defects created in caprine joints. To engineer mechanically competent implants, we first compared a range of 3D printed fibre networks (PCL, PLA and PLGA) for their capacity to mechanically reinforce alginate hydrogels whilst simultaneously supporting mesenchymal stem cell (MSC) chondrogenesis in vitro. These mechanically reinforced, MSC-laden alginate hydrogels were then used to engineer the endochondral bone forming phase of bi-phasic osteochondral constructs, with the overlying chondral phase consisting of cartilage tissue engineered using a co-culture of infrapatellar fat pad derived stem/stromal cells (FPSCs) and chondrocytes. Following chondrogenic priming and subcutaneous implantation in nude mice, these bi-phasic cartilaginous constructs were found to support the development of vascularised endochondral bone overlaid by phenotypically stable cartilage. These fibre-reinforced, bi-phasic cartilaginous templates were then evaluated in clinically relevant, large animal (caprine) model of osteochondral defect repair. Although the quality of repair was variable from animal-to-animal, in general more hyaline-like cartilage repair was observed after 6 months in animals treated with bi-phasic constructs compared to animals treated with commercial control scaffolds. This variability in the quality of repair points to the need for further improvements in the design of 3D bioprinted implants for joint regeneration. STATEMENT OF SIGNIFICANCE: Successful osteochondral defect repair requires regenerating the subchondral bone whilst simultaneously promoting the development of an overlying layer of articular cartilage. In this study, we hypothesised that bi-phasic, fibre-reinforced cartilaginous templates could be leveraged to regenerate both the articular cartilage and subchondral bone within osteochondral defects. To this end we used 3D printed fibre networks to mechanically reinforce engineered transient cartilage, which also contained an overlying layer of phenotypically stable cartilage engineered using a co-culture of chondrocytes and stem cells. When chondrogenically primed and implanted into caprine osteochondral defects, these fibre-reinforced bi-phasic cartilaginous grafts were shown to spatially direct tissue development during joint repair. Such developmentally inspired tissue engineering strategies, enabled by advances in biofabrication and 3D printing, could form the basis of new classes of regenerative implants in orthopaedic medicine.


Assuntos
Cartilagem Articular , Cabras , Impressão Tridimensional , Animais , Regeneração Óssea , Condrogênese , Camundongos , Camundongos Nus , Engenharia Tecidual , Alicerces Teciduais
7.
Mar Drugs ; 18(2)2020 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-31979233

RESUMO

Composite biomaterial scaffolds consisting of natural polymers and bioceramics may offer an alternative to autologous grafts for applications such as bone repair. Herein, we sought to investigate the possibility of incorporating marine coral microparticles into a collagen-based scaffold, a process which we hypothesised would enhance the mechanical properties of the scaffold as well its capacity to promote osteogenesis of human mesenchymal stromal cells. Cryomilling and sieving were utilised to achieve coral microparticles of mean diameters 14 µm and 64 µm which were separately incorporated into collagen-based slurries and freeze-dried to form porous scaffolds. X-ray diffraction and Fourier transform infrared spectroscopy determined the coral microparticles to be comprised of calcium carbonate whereas collagen/coral composite scaffolds were shown to have a crystalline calcium ethanoate structure. Crosslinked collagen/coral scaffolds demonstrated enhanced compressive properties when compared to collagen only scaffolds and also promoted more robust osteogenic differentiation of mesenchymal stromal cells, as indicated by increased expression of bone morphogenetic protein 2 at the gene level, and enhanced alkaline phosphatase activity and calcium accumulation at the protein level. Only subtle differences were observed when comparing the effect of coral microparticles of different sizes, with improved osteogenesis occurring as a result of calcium ion signalling delivered from collagen/coral composite scaffolds. These scaffolds, fabricated from entirely natural sources, therefore show promise as novel biomaterials for tissue engineering applications such as bone regeneration.


Assuntos
Antozoários/química , Materiais Biocompatíveis/farmacologia , Osteogênese/efeitos dos fármacos , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/isolamento & purificação , Regeneração Óssea/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Colágeno/química , Perfilação da Expressão Gênica , Humanos , Teste de Materiais , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Cultura Primária de Células , Espectroscopia de Infravermelho com Transformada de Fourier , Difração de Raios X
8.
Biomaterials ; 188: 63-73, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30321864

RESUMO

Biological scaffolds generated from tissue-derived extracellular matrix (ECM) are commonly used clinically for soft tissue regeneration. Such biomaterials can enhance tissue-specific differentiation of adult stem cells, suggesting that structuring different ECMs into multi-layered scaffolds can form the basis of new strategies for regenerating damaged interfacial tissues such as the osteochondral unit. In this study, mass spectrometry is used to demonstrate that growth plate (GP) and articular cartilage (AC) ECMs contain a unique array of regulatory proteins that may be particularly suited to bone and cartilage repair respectively. Applying a novel iterative freeze-drying method, porous bi-phasic scaffolds composed of GP ECM overlaid by AC ECM are fabricated, which are capable of spatially directing stem cell differentiation in vitro, promoting the development of graded tissues transitioning from calcified cartilage to hyaline-like cartilage. Evaluating repair 12-months post-implantation into critically-sized caprine osteochondral defects reveals that these scaffolds promote regeneration in a manner distinct to commercial control-scaffolds. The GP layer supports endochondral bone formation, while the AC layer stimulates the formation of an overlying layer of hyaline cartilage with a collagen fiber architecture better recapitulating the native tissue. These findings support the use of a bi-layered, tissue-specific ECM derived scaffolds for regenerating spatially complex musculoskeletal tissues.


Assuntos
Condrogênese , Matriz Extracelular/química , Células-Tronco Mesenquimais/citologia , Osteogênese , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/química , Cartilagem Articular/química , Diferenciação Celular , Células Cultivadas , Cabras , Lâmina de Crescimento/química , Regeneração , Suínos , Engenharia Tecidual/métodos
9.
Biores Open Access ; 4(1): 229-41, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26309799

RESUMO

Novel strategies are urgently required to facilitate regeneration of entire bones lost due to trauma or disease. In this study, we present a novel framework for the regeneration of whole bones by tissue engineering anatomically shaped hypertrophic cartilaginous grafts in vitro that subsequently drive endochondral bone formation in vivo. To realize this, we first fabricated molds from digitized images to generate mesenchymal stem cell-laden alginate hydrogels in the shape of different bones (the temporomandibular joint [TMJ] condyle and the distal phalanx). These constructs could be stimulated in vitro to generate anatomically shaped hypertrophic cartilaginous tissues that had begun to calcify around their periphery. Constructs were then formed into the shape of the distal phalanx to create the hypertrophic precursor of the osseous component of an engineered long bone. A layer of cartilage engineered through self-assembly of chondrocytes served as the articular surface of these constructs. Following chondrogenic priming and subcutaneous implantation, the hypertrophic phase of the engineered phalanx underwent endochondral ossification, leading to the generation of a vascularized bone integrated with a covering layer of stable articular cartilage. Furthermore, spatial bone deposition within the construct could be modulated by altering the architecture of the osseous component before implantation. These findings open up new horizons to whole limb regeneration by recapitulating key aspects of normal bone development.

10.
Acta Biomater ; 13: 245-53, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25463500

RESUMO

Cartilaginous tissues engineered using mesenchymal stem cells (MSCs) have been shown to generate bone in vivo by executing an endochondral programme. This may hinder the use of MSCs for articular cartilage regeneration, but opens the possibility of using engineered cartilaginous tissues for large bone defect repair. Hydrogels may be an attractive tool in the scaling-up of such tissue engineered grafts for endochondral bone regeneration. In this study, we compared the capacity of different naturally derived hydrogels (alginate, chitosan and fibrin) to support chondrogenesis and hypertrophy of MSCs in vitro and endochondral ossification in vivo. In vitro, alginate and chitosan constructs accumulated the highest levels of sulfated glycosaminoglycan (sGAG), with chitosan constructs synthesizing the highest levels of collagen. Alginate and fibrin constructs supported the greatest degree of calcium accumulation, though only fibrin constructs calcified homogeneously. In vivo, chitosan constructs facilitated neither vascularization nor endochondral ossification, and also retained the greatest amount of sGAG, suggesting it to be a more suitable material for the engineering of articular cartilage. Both alginate and fibrin constructs facilitated vascularization and endochondral bone formation as well as the development of a bone marrow environment. Alginate constructs accumulated significantly more mineral and supported greater bone formation in central regions of the engineered tissue. In conclusion, this study demonstrates the capacity of chitosan hydrogels to promote and better maintain a chondrogenic phenotype in MSCs and highlights the potential of utilizing alginate hydrogels for MSC-based endochondral bone tissue engineering applications.


Assuntos
Cartilagem Articular/metabolismo , Condrogênese , Hidrogéis/química , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Engenharia Tecidual/métodos , Animais , Cartilagem Articular/citologia , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Suínos
11.
PLoS One ; 9(3): e90716, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24595316

RESUMO

Cartilaginous tissues engineered using mesenchymal stem cells (MSCs) can be leveraged to generate bone in vivo by executing an endochondral program, leading to increased interest in the use of such hypertrophic grafts for the regeneration of osseous defects. During normal skeletogenesis, canals within the developing hypertrophic cartilage play a key role in facilitating endochondral ossification. Inspired by this developmental feature, the objective of this study was to promote endochondral ossification of an engineered cartilaginous construct through modification of scaffold architecture. Our hypothesis was that the introduction of channels into MSC-seeded hydrogels would firstly facilitate the in vitro development of scaled-up hypertrophic cartilaginous tissues, and secondly would accelerate vascularisation and mineralisation of the graft in vivo. MSCs were encapsulated into hydrogels containing either an array of micro-channels, or into non-channelled 'solid' controls, and maintained in culture conditions known to promote a hypertrophic cartilaginous phenotype. Solid constructs accumulated significantly more sGAG and collagen in vitro, while channelled constructs accumulated significantly more calcium. In vivo, the channels acted as conduits for vascularisation and accelerated mineralisation of the engineered graft. Cartilaginous tissue within the channels underwent endochondral ossification, producing lamellar bone surrounding a hematopoietic marrow component. This study highlights the potential of utilising engineering methodologies, inspired by developmental skeletal processes, in order to enhance endochondral bone regeneration strategies.


Assuntos
Cartilagem/citologia , Células-Tronco Mesenquimais/citologia , Osteogênese , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Animais , Calcificação Fisiológica , Cálcio/metabolismo , Cartilagem/irrigação sanguínea , Cartilagem/fisiologia , Células Cultivadas , Condrócitos/citologia , Condrogênese , Hidrogéis/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Suínos
12.
Acta Biomater ; 9(3): 5484-92, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23159563

RESUMO

Chondrogenically primed bone marrow-derived mesenchymal stem cells (MSCs) have been shown to become hypertrophic and undergo endochondral ossification when implanted in vivo. Modulating this endochondral phenotype may be an attractive approach to engineering the osseous phase of an osteochondral implant. The objective of this study was to engineer an osteochondral tissue by promoting endochondral ossification in one layer of a bilayered construct and stable cartilage in the other. The top half of bilayered agarose hydrogels were seeded with culture expanded chondrocytes (termed the chondral layer) and the bottom half of the bilayered agarose hydrogels with MSCs (termed the osseous layer). Constructs were cultured in chondrogenic medium for 21days and thereafter were either maintained in chondrogenic medium, transferred to hypertrophic medium, or implanted subcutaneously into nude mice. This structured chondrogenic bilayered co-culture was found to enhance chondrogenesis in the chondral layer, appearing to help re-establish the chondrogenic phenotype that is lost in chondrocytes during monolayer expansion. Furthermore, the bilayered co-culture appeared to suppress hypertrophy and mineralization in the osseous layer. The addition of hypertrophic factors to the media was found to induce mineralization of the osseous layer in vitro. A similar result was observed in vivo where endochondral ossification was restricted to the osseous layer of the construct, leading to the development of an osteochondral tissue. This novel approach represents a potential new treatment strategy for the repair of osteochondral defects.


Assuntos
Osso e Ossos/fisiopatologia , Osteogênese , Engenharia Tecidual , Alicerces Teciduais/química , Animais , Osso e Ossos/patologia , Calcificação Fisiológica , Condrogênese , Técnicas de Cocultura , Colágeno/metabolismo , DNA/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Hipertrofia , Imuno-Histoquímica , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Sus scrofa
13.
Tissue Eng Part A ; 18(11-12): 1161-70, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22429262

RESUMO

Joint-derived stem cells are a promising alternative cell source for cartilage repair therapies that may overcome many of the problems associated with the use of primary chondrocytes (CCs). The objective of this study was to compare the in vitro functionality and in vivo phenotypic stability of cartilaginous tissues engineered using bone marrow-derived stem cells (BMSCs) and joint tissue-derived stem cells following encapsulation in agarose hydrogels. Culture-expanded BMSCs, fat pad-derived stem cells (FPSCs), and synovial membrane-derived stem cells (SDSCs) were encapsulated in agarose and maintained in a chondrogenic medium supplemented with transforming growth factor-ß3. After 21 days of culture, constructs were either implanted subcutaneously into the back of nude mice for an additional 28 days or maintained for a similar period in vitro in either chondrogenic or hypertrophic media formulations. After 49 days of in vitro culture in chondrogenic media, SDSC constructs accumulated the highest levels of sulfated glycosaminoglycan (sGAG) (∼2.8% w/w) and collagen (∼1.8% w/w) and were mechanically stiffer than constructs engineered using other cell types. After subcutaneous implantation in nude mice, sGAG content significantly decreased for all stem cell-seeded constructs, while no significant change was observed in the control constructs engineered using primary CCs, indicating that the in vitro chondrocyte-like phenotype generated in all stem cell-seeded agarose constructs was transient. FPSCs and SDSCs appeared to undergo fibrous dedifferentiation or resorption, as evident from increased collagen type I staining and a dramatic loss in sGAG content. BMSCs followed a more endochondral pathway with increased type X collagen expression and mineralization of the engineered tissue. In conclusion, while joint tissue-derived stem cells possess a strong intrinsic chondrogenic capacity, further studies are needed to identify the factors that will lead to the generation of a more stable chondrogenic phenotype.


Assuntos
Cartilagem/fisiologia , Células-Tronco/citologia , Engenharia Tecidual/métodos , Tecido Adiposo/citologia , Animais , Células da Medula Óssea/citologia , Cartilagem/efeitos dos fármacos , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Colágeno/metabolismo , Meios de Cultura/farmacologia , Módulo de Elasticidade/efeitos dos fármacos , Glicosaminoglicanos/metabolismo , Hipertrofia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fenótipo , Sefarose/farmacologia , Coloração e Rotulagem , Sus scrofa , Membrana Sinovial/citologia
14.
Biochem Biophys Res Commun ; 417(1): 305-10, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22155244

RESUMO

The local oxygen tension is a key regulator of the fate of mesenchymal stem cells (MSCs). The objective of this study was to investigate the effect of a low oxygen tension during expansion and differentiation on the proliferation kinetics as well as the subsequent osteogenic and chondrogenic potential of MSCs. We first hypothesised that expansion in a low oxygen tension (5% pO(2)) would improve both the subsequent osteogenic and chondrogenic potential of MSCs compared to expansion in a normoxic environment (20% pO(2)). Furthermore, we hypothesised that chondrogenic differentiation in a low oxygen environment would suppress hypertrophy of MSCs cultured in both pellets and hydrogels used in tissue engineering strategies. MSCs expanded at 5% pO(2) proliferated faster forming larger colonies, resulting in higher cell yields. Expansion at 5% pO(2) also enhanced subsequent osteogenesis of MSCs, whereas differentiation at 5% pO(2) was found to be a more potent promoter of chondrogenesis than expansion at 5% pO(2). Greater collagen accumulation, and more intense staining for collagen types I and X, was observed in pellets maintained at 20% pO(2) compared to 5% pO(2). Both pellets and hydrogels stained more intensely for type II collagen when undergoing chondrogenesis in a low oxygen environment. Differentiation at 5% pO(2) also appeared to inhibit hypertrophy in both pellets and hydrogels, as demonstrated by reduced collagen type X and Alizarin Red staining and alkaline phosphatase activity. This study demonstrates that the local oxygen environment can be manipulated in vitro to either stabilise a chondrogenic phenotype for use in cartilage repair therapies or to promote hypertrophy of cartilaginous grafts for endochondral bone repair strategies.


Assuntos
Condrogênese , Células-Tronco Mesenquimais/fisiologia , Osteogênese , Oxigênio , Engenharia Tecidual/métodos , Animais , Células da Medula Óssea/citologia , Técnicas de Cultura de Células , Diferenciação Celular , Proliferação de Células , Glicosaminoglicanos , Pressão Parcial , Suínos
15.
J Tissue Eng Regen Med ; 5(9): 747-58, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21953872

RESUMO

The objective of this study was to investigate how a combination of different scaffold architectures and rotational culture would influence the functional properties of thick cartilaginous tissues engineered using either chondrocytes or bone marrow-derived mesenchymal stem cells (BM-MSCs). Expanded porcine chondrocytes and BM-MSCs were suspended in 2% agarose and cast in custom-designed moulds to produce either regular solid or channelled construct cylinders. The study consisted of three seperate experimental arms. First, chondrocyte and BM-MSC constructs were cultured in free swelling conditions for 9 weeks. Second, constructs were subjected to rotational culture for a period of 3 weeks. Finally, BM-MSC-seeded constructs were subjected to delayed rotational culture, in which constructs were first cultured for 3 weeks in free swelling conditions, followed by an additional 3 weeks in rotating culture conditions. Constructs were supplemented with TGFß3 during the first 3 weeks of all experiments. The introduction of channels alone had little effect on the spatial patterns of tissue accumulation in either chondrocyte- or BM-MSC-seeded constructs. The two cell types responded differentially to rotational culture, resulting in the formation of a more homogeneous tissue in chondrocyte-seeded constructs, but significantly inhibiting chondrogenesis of BM-MSCs. This inhibition of chondrogenesis in response to dynamic culture conditions was not observed if BM-MSC-seeded constructs were first maintained in free swelling conditions for 3 weeks prior to rotation. The results of this study demonstrate that bioreactor culture conditions that are beneficial for chondrocyte-based cartilage tissue engineering may be suboptimal for BM-MSCs.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Cultura de Células/métodos , Condrócitos/citologia , Condrogênese/efeitos dos fármacos , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/citologia , Sefarose/farmacologia , Azul Alciano/metabolismo , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Colágeno Tipo II/metabolismo , Glicosaminoglicanos/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Rotação , Coloração e Rotulagem , Sus scrofa , Fatores de Tempo , Alicerces Teciduais/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...