Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 55
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Biomaterials ; 150: 150-161, 2018 Jan.
Article de Anglais | MEDLINE | ID: mdl-29040875

RÉSUMÉ

Predictive testing of anticancer drugs remains a challenge. Bioengineered systems, designed to mimic key aspects of the human tumor microenvironment, are now improving our understanding of cancer biology and facilitating clinical translation. We show that mechanical signals have major effects on cancer drug sensitivity, using a bioengineered model of human bone sarcoma. Ewing sarcoma (ES) cells were studied within a three-dimensional (3D) matrix in a bioreactor providing mechanical loadings. Mimicking bone-like mechanical signals within the 3D model, we rescued the ERK1/2-RUNX2 signaling pathways leading to drug resistance. By culturing patient-derived tumor cells in the model, we confirmed the effects of mechanical signals on cancer cell survival and drug sensitivity. Analyzing human microarray datasets, we showed that RUNX2 expression is linked to poor survival in ES patients. Mechanical loadings that activated signal transduction pathways promoted drug resistance, stressing the importance of introducing mechanobiological cues into preclinical tumor models for drug screening.


Sujet(s)
Tumeurs osseuses/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Mécanotransduction cellulaire , Sarcome d'Ewing/traitement médicamenteux , Animaux , Antinéoplasiques/pharmacologie , Matériaux biomimétiques/métabolisme , Bioréacteurs , Lignée cellulaire tumorale , Sous-unité alpha 1 du facteur CBF/métabolisme , Humains , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris , Souris SCID , Analyse de survie , Ingénierie tissulaire , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
2.
Theranostics ; 7(17): 4099-4117, 2017.
Article de Anglais | MEDLINE | ID: mdl-29158813

RÉSUMÉ

Neuroblastoma is a vascularized pediatric tumor derived from neural crest stem cells that displays vasculogenic mimicry and can express a number of stemness markers, such as SOX2 and NANOG. Tumor relapse is the major cause of succumbing to this disease, and properties attributed to cancer stem-like cells (CSLC), such as drug-resistance and cell plasticity, seem to be the key mechanisms. However, the lack of controllable models that recapitulate the features of human neuroblastoma limits our understanding of the process and impedes the development of new therapies. In response to these limitations, we engineered a perfusable, vascularized in vitro model of three-dimensional human neuroblastoma to study the effects of retinoid therapy on tumor vasculature and drug-resistance. METHODS: The in vitro model of neuroblastoma was generated using cell-sheet engineering and cultured in a perfusion bioreactor. Firstly, we stacked three cell sheets containing SKNBE(2) neuroblastoma cells and HUVEC. Then, a vascular bed made of fibrin, collagen I and HUVEC cells was placed onto a collagen-gel base with 8 microchannels. After gelling, the stacked cell sheets were placed on the vascular bed and cultured in the perfusion bioreactor (perfusion rate: 0.5 mL/min) for 4 days. Neuroblastoma models were treated with 10µM isotretionin in single daily doses for 5 days. RESULTS: The bioengineered model recapitulated vasculogenic mimicry (vessel-like structure formation and tumor-derived endothelial cells-TECs), and contained CSLC expressing SOX2 and NANOG. Treatment with Isotretinoin destabilized vascular networks but failed to target vasculogenic mimicry and augmented populations of CSLCs expressing high levels of SOX2. Our results suggest that CSLCs can transdifferentiate into drug resistant CD31+-TECs, and reveal the presence of an intermediate state STEC (stem tumor-derived endothelial cell) expressing both SOX2 and CD31. CONCLUSION: Our results reveal some roles of SOX2 in drug resistance and tumor relapse, and suggest that SOX2 could be a therapeutic target in neuroblastoma.


Sujet(s)
Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Isotrétinoïne/pharmacologie , Neuroblastome/vascularisation , Neuroblastome/traitement médicamenteux , Ingénierie tissulaire/méthodes , Tests de criblage d'agents antitumoraux/instrumentation , Tests de criblage d'agents antitumoraux/méthodes , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/anatomopathologie , Régulation de l'expression des gènes tumoraux , Humains , Protéine homéotique Nanog/génétique , Protéine homéotique Nanog/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/anatomopathologie , Neuroblastome/mortalité , Perfusion , Antigènes CD31/métabolisme , Facteurs de transcription SOX-B1/génétique , Facteurs de transcription SOX-B1/métabolisme
3.
Osteoarthritis Cartilage ; 23(2): 319-27, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25452155

RÉSUMÉ

OBJECTIVE: Galvanotaxis, the migratory response of cells in response to electrical stimulation, has been implicated in development and wound healing. The use of mesenchymal stem cells (MSCs) from the synovium (synovium-derived stem cells, SDSCs) has been investigated for repair strategies. Expansion of SDSCs is necessary to achieve clinically relevant cell numbers; however, the effects of culture passage on their subsequent cartilaginous extracellular matrix production are not well understood. METHODS: Over four passages of SDSCs, we measured the expression of cell surface markers (CD31, CD34, CD49c, CD73) and assessed their migratory potential in response to applied direct current (DC) electric field. Cells from each passage were also used to form micropellets to assess the degree of cartilage-like tissue formation. RESULTS: Expression of CD31, CD34, and CD49c remained constant throughout cell expansion; CD73 showed a transient increase through the first two passages. Correspondingly, we observed that early passage SDSCs exhibit anodal migration when subjected to applied DC electric field strength of 6 V/cm. By passage 3, CD73 expression significantly decreased; these cells exhibited cell migration toward the cathode, as previously observed for terminally differentiated chondrocytes. Only late passage cells (P4) were capable of developing cartilage-like tissue in micropellet culture. CONCLUSIONS: Our results show cell priming protocols carried out for four passages selectively differentiate stem cells to behave like chondrocytes, both in their motility response to applied electric field and their production of cartilaginous tissue.


Sujet(s)
Chondrogenèse/physiologie , Mobilisation de cellules souches hématopoïétiques , Cellules souches mésenchymateuses , Animaux , Bovins , Mouvement cellulaire , Cellules cultivées , Stimulation électrique , Mobilisation de cellules souches hématopoïétiques/méthodes , Ingénierie tissulaire/méthodes
4.
Annu Int Conf IEEE Eng Med Biol Soc ; 2015: 3561-4, 2015 Aug.
Article de Anglais | MEDLINE | ID: mdl-26737062

RÉSUMÉ

In vivo, cells reside in a complex environment regulating their fate and function. Most of this complexity is lacking in standard in vitro models, leading to readouts falling short of predicting the actual in vivo situation. The use of engineering tools, combined with deep biological knowledge, leads to the development and use of bioreactors providing biologically sound niches. Such bioreactors offer new tools for biological research, and are now also entering the field of cancer research. Here we present the development and validation of a modular bioreactor system providing: (i) high throughput analyses, (ii) a range of biological conditions, (iii) high degree of control, and (iv) application of physiological stimuli to the cultured samples. The bioreactor was used to engineer a three-dimensional (3D) tissue model of cancer, where the effects of mechanical stimulation on the tumor phenotype were evaluated. Mechanical stimuli applied to the engineered tumor model activated the mechanotransduction machinery and resulted in measurable changes of mRNA levels towards a more aggressive tumor phenotype.


Sujet(s)
Bioréacteurs , Tumeurs osseuses/anatomopathologie , Ingénierie tissulaire/instrumentation , Microenvironnement tumoral , Biophysique , Conception d'appareillage , Humains , Mécanotransduction cellulaire/physiologie , Ingénierie tissulaire/méthodes
5.
Eur Cell Mater ; 25: 229-47, 2013 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-23588785

RÉSUMÉ

Tissue engineering combined with gene therapy is a promising approach for promoting articular cartilage repair. Here, we tested the hypothesis that engineered cartilage with chondrocytes overexpressing a human insulin-like growth factor I (IGF-I) gene can enhance the repair of osteochondral defects, in a manner dependent on the duration of cultivation. Genetically modified chondrocytes were cultured on biodegradable polyglycolic acid scaffolds in dynamic flow rotating bioreactors for either 10 or 28 d. The resulting cartilaginous constructs were implanted into osteochondral defects in rabbit knee joints. After 28 weeks of in vivo implantation, immunoreactivity to ß-gal was detectable in the repair tissue of defects that received lacZ constructs. Engineered cartilaginous constructs based on IGF-I-overexpressing chondrocytes markedly improved osteochondral repair compared with control (lacZ) constructs. Moreover, IGF-I constructs cultivated for 28 d in vitro significantly promoted osteochondral repair vis-à-vis similar constructs cultivated for 10 d, leading to significantly decreased osteoarthritic changes in the cartilage adjacent to the defects. Hence, the combination of spatially defined overexpression of human IGF-I within a tissue-engineered construct and prolonged bioreactor cultivation resulted in most enhanced articular cartilage repair and reduction of osteoarthritic changes in the cartilage adjacent to the defect. Such genetically enhanced tissue engineering provides a versatile tool to evaluate potential therapeutic genes in vivo and to improve our comprehension of the development of the repair tissue within articular cartilage defects. Insights gained with additional exploration using this model may lead to more effective treatment options for acute cartilage defects.


Sujet(s)
Cartilage articulaire/anatomopathologie , Chondrocytes/métabolisme , Facteur de croissance IGF-I/métabolisme , Ingénierie tissulaire , Structures d'échafaudage tissulaires/composition chimique , Cicatrisation de plaie , Animaux , Bioréacteurs , Cartilage articulaire/métabolisme , Numération cellulaire , Chondrogenèse , Collagène de type II/métabolisme , Modèles animaux de maladie humaine , Humains , Immunohistochimie , Mâle , Implantation de prothèse , Lapins , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie , Transfection , Transgènes , beta-Galactosidase/métabolisme
6.
Osteoarthritis Cartilage ; 18(5): 714-23, 2010 May.
Article de Anglais | MEDLINE | ID: mdl-20175974

RÉSUMÉ

OBJECTIVE: The objective of the study was to investigate the combined effects of three sets of regulatory factors: cell pre-differentiation, soluble factors and medium perfusion on spatial control of human mesenchymal stem cell (hMSC) differentiation into cells forming the cartilaginous and bone regions in engineered osteochondral constructs. DESIGN: Bone-marrow derived hMSCs were expanded in their undifferentiated state (UD) or pre-differentiated (PD) in monolayer culture, seeded into biphasic constructs by interfacing agarose gels and bone scaffolds and cultured for 5 weeks either statically (S) or in a bioreactor (BR) with perfusion of medium through the bone region. Each culture system was operated with medium containing either chondrogenic supplements (C) or a cocktail (Ck) of chondrogenic and osteogenic supplements. RESULTS: The formation of engineered cartilage in the gel region was most enhanced by using undifferentiated cells and chondrogenic medium, whereas the cartilaginous properties were negatively affected by using pre-differentiated cells or the combination of perfusion and cocktail medium. The formation of engineered bone in the porous scaffold region was most enhanced by using pre-differentiated cells, perfusion and cocktail medium. Perfusion also enhanced the integration of bone and cartilage regions. CONCLUSIONS: (1) Pre-differentiation of hMSCs before seeding on scaffold was beneficial for bone but not for cartilage formation. (2) The combination of medium perfusion and cocktail medium inhibited chondrogenesis of hMSCs. (3) Perfusion improved the cell and matrix distribution in the bone region and augmented the integration at the bone-cartilage interface. (4) Osteochondral grafts can be engineered by differentially regulating the culture conditions in the two regions of the scaffold seeded with hMSCs (hydrogel for cartilage, perfused porous scaffold for bone).


Sujet(s)
Cartilage/cytologie , Cartilage/croissance et développement , Chondrogenèse/physiologie , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/physiologie , Ostéogenèse/physiologie , Ingénierie tissulaire/méthodes , Bioréacteurs , Techniques de culture cellulaire/méthodes , Différenciation cellulaire/physiologie , Cellules cultivées , Humains , Immunohistochimie , Perfusion , Contrainte mécanique , Tomodensitométrie
7.
Article de Anglais | MEDLINE | ID: mdl-19163485

RÉSUMÉ

Overcoming the limitations of diffusional transport in conventional culture systems remains an open issue for successfully generating thick, compact and functional cardiac tissues. Previously, it was shown that perfusion systems enhance the yield and uniformity of cell seeding and cell survival in thick cardiac constructs. The aim of our study was to form highly functional cardiac constructs starting from spatially uniform, high density cell seeded constructs. Disk-shaped elastomeric poly(glycerol sebacate) scaffolds were seeded with neonatal rat cardiomyocytes and cultured for eight days with direct perfusion of culture medium or statically in a six-well plate. In the perfusion experimental group, the integrity of some disks was well maintained, whereas in others a central hole was formed, resulting in ring-shaped constructs. This allowed us to also study the effects of construct geometry and of interstitial flow versus channel perfusion. The ring-shaped constructs appeared to have a denser and more uniform deposition of extracellular matrix. In response to electrical stimulation, the fractional area change of the ring-shaped constructs was 7.3 and 2.7 times higher than for disk-shaped tissues cultured in perfusion or statically, respectively. These findings suggest that a combination of many factors, including scaffold elasticity and geometry and the type of perfusion system applied, need to be considered in order to engineer a cardiac construct with high contractile activity.


Sujet(s)
Contraction musculaire , Ingénierie tissulaire/méthodes , Animaux , Animaux nouveau-nés , Bioréacteurs , Cellules cultivées , Décanoate/composition chimique , Électrothérapie/méthodes , Électrophysiologie/méthodes , Matrice extracellulaire/métabolisme , Glycérol/analogues et dérivés , Glycérol/composition chimique , Myocarde/métabolisme , Myocarde/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Perfusion , Polymères/composition chimique , Rats , Rat Sprague-Dawley
8.
Article de Anglais | MEDLINE | ID: mdl-19163486

RÉSUMÉ

Electrical stimulation has been shown to improve functional assembly of cardiomyocytes in vitro for cardiac tissue engineering. Carbon electrodes were found in past studies to have the best current injection characteristics. The goal of this study was to develop rational experimental design principles for the electrodes and stimulation regime, in particular electrode configuration, electrode ageing, and stimulation amplitude. Carbon rod electrodes were compared via electrochemical impedance spectroscopy (EIS) and we identified a safety range of 0 to 8 V/cm by comparing excitation thresholds and maximum capture rates for neonatal rat cardiomyocytes cultured with electrical stimulation. We conclude with recommendations for studies involving carbon electrodes for cardiac tissue engineering.


Sujet(s)
Bioréacteurs , Myocarde/anatomopathologie , Ingénierie tissulaire/méthodes , Animaux , Animaux nouveau-nés , Carbone/composition chimique , Stimulation électrique , Électricité , Électrochimie/méthodes , Électrodes , Conception d'appareillage , Modèles chimiques , Myocytes cardiaques/cytologie , Rats , Température
9.
Philos Trans R Soc Lond B Biol Sci ; 362(1484): 1357-68, 2007 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-17594967

RÉSUMÉ

Here, we review an approach to tissue engineering of functional myocardium that is biomimetic in nature, as it involves the use of culture systems designed to recapitulate some aspects of the actual in vivo environment. To mimic the capillary network, subpopulations of neonatal rat heart cells were cultured on a highly porous elastomer scaffold with a parallel array of channels perfused with culture medium. To mimic oxygen supply by haemoglobin, the culture medium was supplemented with a perfluorocarbon (PFC) emulsion. Constructs cultivated in the presence of PFC contained higher amounts of DNA and cardiac markers and had significantly better contractile properties than control constructs cultured without PFC. To induce synchronous contractions of cultured constructs, electrical signals mimicking those in native heart were applied. Over only 8 days of cultivation, electrical stimulation induced cell alignment and coupling, markedly increased the amplitude of synchronous construct contractions and resulted in a remarkable level of ultrastructural organization. The biomimetic approach is discussed in the overall context of cardiac tissue engineering, and the possibility to engineer functional human cardiac grafts based on human stem cells.


Sujet(s)
Matériaux biomimétiques , Biomimétique/méthodes , Coeur/physiologie , Ingénierie tissulaire/méthodes , Aérobiose , Animaux , Transport biologique , Différenciation cellulaire , Survie cellulaire , Cellules cultivées , Modèles biologiques , Myocarde/métabolisme , Myocytes cardiaques/physiologie , Oxygène , Rats , Techniques de culture de tissus
10.
Eur Spine J ; 16(11): 1848-57, 2007 Nov.
Article de Anglais | MEDLINE | ID: mdl-17447088

RÉSUMÉ

There is no optimal treatment for symptomatic degenerative disc disease which affects millions of people worldwide. One novel approach would be to form a patch or tissue replacement to repair the annulus fibrosus (AF) through which the NP herniates. As the optimal scaffold for this has not been defined the purpose of this study was to determine if porous silk scaffolds would support AF cell attachment and extracellular matrix accumulation and whether chemically decorating the scaffold with RGD peptide, which has been shown to enhance attachment for other cell types, would further improve AF cell attachment and tissue formation. Annulus fibrosus cells were isolated from bovine caudal discs and seeded into porous silk scaffolds. The percent cell attachment was quantified and the cell morphology and distribution within the scaffold was evaluated using scanning electron microscopy. The cell-seeded scaffolds were grown for up to 8 weeks and evaluated for gene expression, histological appearance and matrix accumulation. AF cells attach to porous silk scaffolds, proliferate and synthesize and accumulate extracellular matrix as demonstrated biochemically and histologically. Coupling the silk scaffold with RGD-peptides did not enhance cell attachment nor tissue formation but did affect cell morphology. As well, the cells had higher levels of type II collagen and aggrecan gene expression when compared to cells grown on the non-modified scaffold, a feature more in keeping with cells of the inner annulus. Porous silk is an appropriate scaffold on which to grow AF cells. Coupling RGD peptide to the scaffold appears to influence AF cell phenotype suggesting that it may be possible to select an appropriate scaffold that favours inner annulus versus outer annulus differentiation which will be important for tissue engineering an intervertebral disc.


Sujet(s)
Disque intervertébral/anatomie et histologie , Soie/pharmacologie , Ingénierie tissulaire/méthodes , Structures d'échafaudage tissulaires , Animaux , Bovins , Adhérence cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Collagène/biosynthèse , ADN/analyse , Électrophorèse sur gel d'agar , Matrice extracellulaire/effets des médicaments et des substances chimiques , Matrice extracellulaire/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Disque intervertébral/cytologie , Disque intervertébral/métabolisme , Disque intervertébral/ultrastructure , Microscopie électronique à balayage , Oligopeptides/pharmacologie , Porosité/effets des médicaments et des substances chimiques , Facteurs temps
11.
Bone ; 39(4): 922-31, 2006 Oct.
Article de Anglais | MEDLINE | ID: mdl-16757219

RÉSUMÉ

Bone auto- and allografts have inherent drawbacks, therefore the treatment of non-unions and critical size defects in load bearing long bones would benefit from the use of osteopromotive biodegradable, biocompatible and mechanically durable matrices to enhance migration or delivery of cell populations and/or morphogens/cytokines. Silk fibroin biomaterial scaffolds were evaluated as osteopromotive matrices in critical sized mid-femoral segmental defects in nude rats. Four treatment groups were assessed over 8 weeks in vivo: silk scaffolds (SS) with human mesenchymal stem cells (hMSCs) that had previously been differentiated along an osteoblastic lineage in vitro (group I; pdHMSC/SS); SS with undifferentiated hMSCs (group II; udHMSC/SS); SS alone (group III; SS); and empty defects (group IV). When hMSCs were cultured in vitro in osteogenic medium for 5 weeks, bone formation was characterized with bimodal peak activities for alkaline phosphatase at 2 and 4 weeks. Calcium deposition started after 1 week and progressively increased to peak at 4 weeks, reaching cumulative levels of deposited calcium at 16 mug per mg scaffold wet weight. In vivo osteogenesis was characterized by almost bridged defects with newly formed bone after 8 weeks in group I. Significantly (P < 0.01) greater bone volumes were observed with the pdHMSC/SS (group I) implants than with groups II, III or IV. These three groups failed to induce substantial new bone formation and resulted in the ingrowth of cells with fibroblast-like morphology into the defect zone. The implantation of pdHMSC/SS resulted in significantly (P < 0.05) greater maximal load and torque when compared to the other treatment regimens. The pdHMSC/SS implants demonstrated osteogenic ability in vitro and capacity to thrive towards the healing of critical size femoral segmental defects in vivo. Thus, these new constructs provide an alternative protein-based biomaterial for load bearing applications.


Sujet(s)
Matériaux biocompatibles/usage thérapeutique , Fémur/effets des médicaments et des substances chimiques , Soie/métabolisme , Phosphatase alcaline/métabolisme , Animaux , Matériaux biocompatibles/métabolisme , Calcium/métabolisme , Cellules cultivées , Fémur/anatomopathologie , Fémur/chirurgie , Fibroïne/métabolisme , Humains , Immunohistochimie , Mâle , Transplantation de cellules souches mésenchymateuses/méthodes , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Répartition aléatoire , Rats , Rat nude , Facteurs temps , Ingénierie tissulaire/méthodes , Tomodensitométrie/méthodes , Transplantation hétérologue , Résultat thérapeutique
12.
J Control Release ; 111(1-2): 219-27, 2006 Mar 10.
Article de Anglais | MEDLINE | ID: mdl-16458987

RÉSUMÉ

The pharmaceutical utility of silk fibroin (SF) materials for drug delivery was investigated. SF films were prepared from aqueous solutions of the fibroin protein polymer and crystallinity was induced and controlled by methanol treatment. Dextrans of different molecular weights, as well as proteins, were physically entrapped into the drug delivery device during processing into films. Drug release kinetics were evaluated as a function of dextran molecular weight, and film crystallinity. Treatment with methanol resulted in an increase in beta-sheet structure, an increase in crystallinity and an increase in film surface hydrophobicity determined by FTIR, X-ray and contact angle techniques, respectively. The increase in crystallinity resulted in the sustained release of dextrans of molecular weights ranging from 4 to 40 kDa, whereas for less crystalline films sustained release was confined to the 40 kDa dextran. Protein release from the films was studied with horseradish peroxidase (HRP) and lysozyme (Lys) as model compounds. Enzyme release from the less crystalline films resulted in a biphasic release pattern, characterized by an initial release within the first 36 h, followed by a lag phase and continuous release between days 3 and 11. No initial burst was observed for films with higher crystallinity and subsequent release patterns followed linear kinetics for HRP, or no substantial release for Lys. In conclusion, SF is an interesting polymer for drug delivery of polysaccharides and bioactive proteins due to the controllable level of crystallinity and the ability to process the biomaterial in biocompatible fashion under ambient conditions to avoid damage to labile compounds to be delivered.


Sujet(s)
Préparations à action retardée/composition chimique , Fibroïne/composition chimique , Polymères/composition chimique , Adsorption , Animaux , Bombyx/composition chimique , Chromatographie en phase liquide à haute performance , Cristallisation , Préparations à action retardée/pharmacocinétique , Dextrane/composition chimique , Dextrane/pharmacocinétique , Fibroïne/isolement et purification , Fluorescence , Horseradish peroxidase/composition chimique , Horseradish peroxidase/pharmacocinétique , Méthanol/composition chimique , Microscopie à force atomique , Masse moléculaire , Lysozyme/composition chimique , Lysozyme/pharmacocinétique , Spectroscopie infrarouge à transformée de Fourier , Propriétés de surface , Technologie pharmaceutique/méthodes , Facteurs temps
13.
Conf Proc IEEE Eng Med Biol Soc ; 2006: 3656-8, 2006.
Article de Anglais | MEDLINE | ID: mdl-17947047

RÉSUMÉ

With the ultimate goal to engineer a meniscus substitute based on autologous cells, we aimed this work at identifying (i) a human cell source capable of generating fibrocartilaginous tissues and (ii) a culture environment promoting the development of bi-zonal constructs, resembling the complex structure and function of a meniscus. The post-expansion differentiation capacity of different chondrogenic cells readily available by knee arthroscopy, namely inner meniscus, fat pad, synovial membrane cells and articular chondrocytes (AC), was assessed within hyaluronan based non-woven meshes. Under our experimental conditions, only expanded AC generated tissues containing relevant amounts of glycosaminoglycans (GAG) and with cell phenotypes compatible with those of the inner and outer meniscus regions. Physical conditioning of constructs generated by expanded AC was applied using mixed flasks. The hydrodynamic environment of mixed flasks was instrumental to promote the formation of bi-zonal tissues, with an inner region rich in GAG and stiffer in compression and an outer rim rich in collagen and stiffer in tension. Therefore, the use of AC cultured within porous scaffolds in mixed flasks allowed engineering of constructs resembling some aspects of the phenotype and function of meniscus tissue.


Sujet(s)
Chondrocytes/cytologie , Ménisques de l'articulation du genou/cytologie , Tendons/cytologie , Ingénierie tissulaire/méthodes , Résistance à la compression , Élasticité , Fibrocartilage/cytologie , Humains , Ingénierie tissulaire/tendances , Transplantation autologue
14.
Orthod Craniofac Res ; 8(3): 209-18, 2005 Aug.
Article de Anglais | MEDLINE | ID: mdl-16022723

RÉSUMÉ

The clinical utility of tissue engineering depends upon our ability to direct cells to form tissues with characteristic structural and mechanical properties across different hierarchical scales. Ideally, an engineered graft should be tailored to (re)establish the structure and function of the native tissue being replaced. Engineered grafts of such high fidelity would also foster fundamental research by serving as physiologically relevant models for quantitative in vitro studies. The approach discussed here involves the use of human mesenchymal stem cells (hMSC) cultured on custom-designed scaffolds (providing a structural and logistic template for tissue development) in bioreactors (providing environmental control, biochemical and mechanical cues). Cartilage, bone and ligaments have been engineered by using hMSC, highly porous protein scaffolds (collagen; silk) and bioreactors (perfused cartridges with or without mechanical loading). In each case, the scaffold and bioreactor were designed to recapitulate some aspects of the environment present in native tissues. Medium flow facilitated mass transport to the cells and thereby enhanced the formation of all three tissues. In the case of cartilage, dynamic laminar flow patterns were advantageous as compared to either turbulent steady flow or static (no flow) cultures. In the case of bone, medium flow affected the geometry, distribution and orientation of the forming bone-like trabeculae. In the case of ligament, applied mechanical loading (a combination of dynamic stretch and torsion) markedly enhanced cell differentiation, alignment and functional assembly. Taken together, these studies provide a basis for the ongoing work on engineering osreochondral grafts for a variety of potential applications, including those in the craniofacial complex.


Sujet(s)
Bioréacteurs , Transplantation osseuse , Cartilage/transplantation , Ingénierie tissulaire/instrumentation , Matériaux biocompatibles/composition chimique , Os et tissu osseux/cytologie , Os et tissu osseux/physiologie , Cartilage/cytologie , Cartilage/physiologie , Techniques de culture cellulaire , Différenciation cellulaire/physiologie , Collagène/composition chimique , Humains , Ligaments/cytologie , Ligaments/physiologie , Cellules souches mésenchymateuses/physiologie , Rhéologie , Soie/composition chimique , Contrainte mécanique
15.
Osteoarthritis Cartilage ; 13(2): 129-38, 2005 Feb.
Article de Anglais | MEDLINE | ID: mdl-15694574

RÉSUMÉ

OBJECTIVE: An in vitro model was used to test the hypothesis that culture time and adjacent tissue structure and composition affected chondrogenesis and integrative repair in engineered cartilage. METHOD: Engineered constructs made of bovine calf chondrocytes and hyaluronan benzyl ester non-woven mesh were press-fitted into adjacent tissue rings made of articular cartilage (AC), devitalized bone (DB), or vital bone (VB) and cultured in rotating bioreactors for up to 8 weeks. Structure (light and electron microscopy), biomechanical properties (interfacial adhesive strength, construct compressive modulus), biochemical composition (construct glycosaminoglycans (GAG), collagen, and cells), and adjacent tissue diffusivity were assessed. RESULTS: Engineered constructs were comprised predominately of hyaline cartilage, and appeared either closely apposed to adjacent cartilage or functionally interdigitated with adjacent bone due to interfacial deposition of extracellular matrix. An increase in culture time significantly improved construct adhesive strength (P<0.001), modulus (P=0.02), GAG (P=0.04) and cellularity (P<0.001). The type of adjacent tissue significantly affected construct adhesion (P<0.001), modulus (P<0.001), GAG (P<0.001) and collagen (P<0.001). For constructs cultured in rings of cartilage, negative correlations were observed between ring GAG content (log transformed) and construct adhesion (R2=0.66, P<0.005), modulus (R2=0.49, P<0.05) and GAG (R2=0.44, P<0.05). Integrative repair was better for constructs cultured adjacent to bone than cartilage, in association with its solid architectural structure and high GAG content, and best for constructs cultured adjacent to DB, in association with its high diffusivity. CONCLUSIONS: Chondrogenesis and integrative repair in engineered cartilage improved with time and depended on adjacent tissue architecture, composition, and transport properties.


Sujet(s)
Os et tissu osseux/physiologie , Cartilage articulaire/physiologie , Ingénierie tissulaire/méthodes , Animaux , Phénomènes biomécaniques , Os et tissu osseux/anatomie et histologie , Os et tissu osseux/ultrastructure , Cartilage articulaire/anatomie et histologie , Cartilage articulaire/ultrastructure , Bovins , Cellules cultivées , Chondrocytes/physiologie , Chondrogenèse/physiologie , Diffusion , Collagènes fibrillaires/métabolisme , Acide hyaluronique/métabolisme , Microscopie électronique/méthodes , Facteurs temps
16.
Biorheology ; 41(3-4): 445-58, 2004.
Article de Anglais | MEDLINE | ID: mdl-15299276

RÉSUMÉ

One approach to functional tissue engineering of cartilage is to utilize bioreactors to provide environmental conditions that stimulate chondrogenesis in cells cultured on biomaterial scaffolds. We report the combined use of a three-dimensional in vitro model and a novel bioreactor with perfusion of culture medium and mechanical stimulation in long-term studies of cartilage development and function. To engineer cartilage, scaffolds made of a non-woven mesh of polyglycolic acid (PGA) were seeded with bovine calf articular chondrocytes, cultured for an initial 30-day period under free swelling conditions, and cultured for an additional 37 day period in one of the three groups: (1) free-swelling, (2) static compression (on 24 h/day, strain control, static offset 10%), and (3) dynamic compression (on 1 h/day; off 23 h/day; strain control, static offset 2%, dynamic strain amplitude 5%; frequency 0.3 Hz). Constructs were sampled at timed intervals and assessed with respect to structure, biochemical composition, and mechanical function. Mechanical simulation had little effect on the compositions, morphologies and on mechanical properties of construct interiors discs, but it resulted in distincly different properties of the peripheral rings and face sides. Contructs cultured with mechanical loading maintained their cylindrical shape with flat and parallel top and bottom surfaces, and retained larger amounts of GAG. The modular bioreactor system with medium perfusion and mechanical loading can be utilized to define the conditions of cultivation for functional tissue engineering of cartilage.


Sujet(s)
Cartilage articulaire , Ingénierie tissulaire/méthodes , Animaux , Matériaux biocompatibles , Bioréacteurs , Cartilage articulaire/composition chimique , Cartilage articulaire/cytologie , Bovins , Chondrogenèse , Collagène/analyse , Matrice extracellulaire/composition chimique , Glycosaminoglycanes/analyse , Perfusion , Contrainte mécanique
17.
Annu Rev Biomed Eng ; 6: 131-56, 2004.
Article de Anglais | MEDLINE | ID: mdl-15255765

RÉSUMÉ

Tissue engineering is emerging as a significant clinical option to address tissue and organ failure by implanting biological substitutes for the compromised tissues. As compared to the transplantation of cells alone, engineered tissues offer the potential advantage of immediate functionality. Engineered tissues can also serve as physiologically relevant models for controlled studies of cells and tissues designed to distinguish the effects of specific signals from the complex milieu of factors present in vivo. A high number of ligament failures and the lack of adequate options to fully restore joint functions have prompted the need to develop new tissue engineering strategies. We discuss the requirements for ligament reconstruction, the available treatment options and their limitations, and then focus on the tissue engineering of ligaments. One representative tissue engineering system involving the integrated use of adult human stem cells, custom-designed scaffolds, and advanced bioreactors with dynamic loading is described.


Sujet(s)
Ligaments/anatomopathologie , Mésoderme/cytologie , Cellules souches/cytologie , Ingénierie tissulaire/méthodes , Animaux , Matériaux biocompatibles , Bioréacteurs , Collagène/composition chimique , Humains , Phénotype
18.
Orthopade ; 33(6): 721-6, 2004 Jun.
Article de Allemand | MEDLINE | ID: mdl-15004669

RÉSUMÉ

Extensive osteochondral lesions require repair of the cartilage and underlying bone. We generated osteochondral repair tissue by tissue engineering. Standardized defects, 7 x 5 x 5 mm, were created in femoropatellar grooves of adult rabbits. Engineered cartilage, generated in vitro starting from chondrocytes and a biodegradable scaffold, was implanted using Collagraft as subchondral support. Cell-free implants, defects left empty, and unoperated knee joints served as controls. Explants were characterized morphologically and mechanically. Engineered cartilage implants were superior to cell-free implants and to natural healing of empty defects with respect to the histologic score and Young's modulus of the 6-month repair tissue. These data suggest that engineered cartilage can provide primary stability for the treatment of critical osteochondral defects.


Sujet(s)
Cartilage/croissance et développement , Cartilage/transplantation , Chondrocytes/anatomopathologie , Réaction à corps étranger/anatomopathologie , Réaction à corps étranger/physiopathologie , Articulation du genou/chirurgie , Ingénierie tissulaire/méthodes , Remodelage osseux , Cartilage/anatomopathologie , Cartilage/physiopathologie , Cellules cultivées , Élasticité , Articulation du genou/anatomopathologie , Articulation du genou/physiopathologie , Résistance à la traction , Résultat thérapeutique , Mise en charge
19.
Calcif Tissue Int ; 74(5): 458-68, 2004 May.
Article de Anglais | MEDLINE | ID: mdl-14961210

RÉSUMÉ

Bone is a dynamic tissue that is able to sense and adapt to mechanical stimuli by modulating its mass, geometry, and structure. Bone marrow stromal cells (BMSCs) are known to play an integral part in bone formation by providing an osteoprogenitor cell source capable of differentiating into mature osteoblasts in response to mechanical stresses. Characteristics of the in vivo bone environment including the three dimensional (3-D) lacunocanalicular structure and extracellular matrix composition have previously been shown to play major roles in influencing mechanotransduction processes within bone cells. To more accurately model this phenomenon in vitro, we cultured human BMSCs on 3-D, partially demineralized bone scaffolds in the presence of four-point bending loads within a novel bioreactor. The effect of mechanical loading and dexamethasone concentration on BMSC osteogenic differentiation and mineralized matrix production was studied for 8 and 16 days of culture. Mechanical stimulation after 16 days with 10 nM dexamethasone promoted osteogenic differentiation of BMSCs by significantly elevating alkaline phosphatase activity as well as alkaline phosphatase and osteopontin transcript levels over static controls. Mineralized matrix production also increased under these culture conditions. Dexamethasone concentration had a dramatic effect on the ability of mechanical stimulation to modulate these phenotypic and genotypic responses. These results provide increased insight into the role of mechanical stimulation on osteogenic differentiation of human BMSCs in vitro and may lead to improved strategies in bone tissue engineering.


Sujet(s)
Cellules de la moelle osseuse/cytologie , Différenciation cellulaire/physiologie , Ostéogenèse/physiologie , Cellules stromales/cytologie , Ingénierie tissulaire/méthodes , Adulte , Phosphatase alcaline/métabolisme , Technique de déminéralisation de l'os , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Trame osseuse/effets des médicaments et des substances chimiques , Trame osseuse/physiologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Résistance à la compression/effets des médicaments et des substances chimiques , Dexaméthasone/pharmacologie , Relation dose-effet des médicaments , Glucocorticoïdes/pharmacologie , Humains , Ostéogenèse/effets des médicaments et des substances chimiques , RT-PCR , Contrainte mécanique , Cellules stromales/effets des médicaments et des substances chimiques , Résistance à la traction/effets des médicaments et des substances chimiques , Ingénierie tissulaire/instrumentation
20.
Tissue Eng ; 8(4): 591-601, 2002 Aug.
Article de Anglais | MEDLINE | ID: mdl-12201999

RÉSUMÉ

Bovine calf articular chondrocytes were seeded onto biodegradable polyglycolic acid (PGA) scaffolds and cultured in either control medium or medium supplemented with 1, 10, or 100 ng/mL of bone morphogenetic proteins (BMPs) BMP-2, BMP-12, or BMP-13. Under all conditions investigated, cell-polymer constructs cultivated for 4 weeks in vitro macroscopically and histologically resembled native cartilage. Addition of 100 ng/mL of BMP-2, BMP-12, or BMP-13 increased the total mass of the constructs relative to the controls by 121%, 80%, and 62%, respectively, which was accompanied by increases in the absolute amounts of collagen, glycosaminoglycans (GAG), and cells. The addition of 100 ng/mL of BMP-2, BMP-12, or BMP-13 increased the weight percentage of GAG in the constructs by 27%, 18%, and 15%, and decreased the weight percent of total collagen to 63%, 89%, and 83% of controls, respectively. BMP-2, but not BMP-12 or BMP-13 promoted chondrocyte hypertrophy. Taken together, these data suggest that BMP-2, BMP-12, and BMP-13 increase growth rate and modulate the composition of engineered cartilage and that 100 ng/mL of BMP-2 has the greatest effect. In addition, in vitro engineered cartilage provides a system for studying the effects of BMPs on chondrogenesis in a well-defined environment.


Sujet(s)
Protéines morphogénétiques osseuses/métabolisme , Cartilage/cytologie , Ingénierie tissulaire/méthodes , Animaux , Bovins , Chondrocytes/métabolisme , Matrice extracellulaire/métabolisme , Glycosaminoglycanes/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE