• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 13
  • 3
  • 2
  • Tagged with
  • 21
  • 21
  • 21
  • 9
  • 7
  • 6
  • 6
  • 6
  • 6
  • 5
  • 3
  • 3
  • 3
  • 3
  • 3
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
11

Engineering zonally organized articular cartilage

Nguyen, Lonnissa Hong 14 October 2011 (has links)
Cartilage regeneration is one of the most widely studied areas in tissue-engineering. Despite significant progress, most efforts to date have only focused on generating homogenous tissues whose bulk properties are similar to articular cartilage. However, anatomically and functionally, articular cartilage consists of four spatially distinct regions: the superficial, transitional, deep, and calcified zones. Each zone is characterized by unique extra-cellular matrix (ECM) compositions, mechanical properties, and cellular organization. The ECM is primarily composed of type II collagen and glycosaminoglycans (GAGs), whose relative concentrations vary between zones and therefore lead to distinctive mechanical properties. One of the major unsolved challenges in engineering cartilage has been the inability to regenerate tissue that mimics the zonal architecture of articular cartilage. Recent studies have attempted to imitate this spatial organization using zone-specific chondrocytes isolated from donor animal cartilage. Directed differentiation of a single stem population into zonally organized native-like articular cartilage has not yet been reported. This dissertation reports that hydrogels, incorporating both synthetic and natural polymers as well as cell-induced degradability, are suitable for generating zone-specific chondrogenic phenotypes from a single MSC population. Specifically, cues provided from the unique combinations of chondroitin sulfate (CS), hyaluronic acid (HA), and MMP-sensitive peptide (MMP-pep) within a PEG-based hydrogel, direct the chondrogenic differentiation of MSCs. The findings of this dissertation demonstrate the capability of creating native-like and mechanically relevant articular cartilage consisting of zone specific layers. This ability provides a new direction in cartilage tissue engineering and could be invaluable for cartilage repair if incorporated with current minimally invasive surgical techniques. / text
12

DEVELOPMENT OF HYBRID-CONSTRUCT BIOPRINTING AND SYNCHROTRON-BASED NON-INVASIVE ASSESSMENT TECHNIQUES FOR CARTILAGE TISSUE ENGINEERING

2015 December 1900 (has links)
Cartilage tissue engineering has been emerging as a promising therapeutic approach, where engineered constructs or scaffolds are used as temporary supports to promote regeneration of functional cartilage tissue. Hybrid constructs fabricated from cells, hydrogels, and solid polymeric materials show the most potential for their enhanced biological and mechanical properties. However, fabrication of customized hybrid constructs with impregnated cells is still in its infancy and many issues related to their structural integrity and the cell functions need to be addressed by research. Meanwhile, it is noticed that nowadays monitoring the success of tissue engineered constructs must rely on animal models, which have to be sacrificed for subsequent examination based on histological techniques. This becomes a critical issue as tissue engineering advances from animal to human studies, thus raising a great need for non-invasive assessments of engineered constructs in situ. To address the aforementioned issues, this research is aimed to (1) develop novel fabrication processes to fabricate hybrid constructs incorporating living cells (hereafter referred as “construct biofabrication”) for cartilage tissue regeneration and (2) develop non-invasive monitoring methods based on synchrotron X-ray imaging techniques for examining cartilage tissue constructs in situ. Based on three-dimensional (3D) printing techniques, novel biofabrication processes were developed to create constructs from synthetic polycaprolactone (PCL) polymer framework and cell-impregnated alginate hydrogel, so as to provide both structural and biological properties as desired in cartilage tissue engineering. To ensure the structural integrity of the constructs, the influence of both PCL polymer and alginate was examined, thus forming a basis to prepare materials for subsequent construct biofabrication. To ensure the biological properties, three types of cells, i.e., two primary cell populations from embryonic chick sternum and an established chondrocyte cell line of ATDC5 were chosen to be incorporated in the construct biofabrication. The biological performance of the cells in the construct were examined along with the influence of the polymer melting temperature on them. The promising results of cell viability and proliferation as well as cartilage matrix production demonstrate that the developed processes are appropriate for fabricating hybrid constructs for cartilage tissue engineering. To develop non-invasive in situ assessment methods for cartilage and other soft tissue engineering applications, synchrotron phase-based X-ray imaging techniques of diffraction enhanced imaging (DEI), analyzer based imaging (ABI), and inline phase contrast imaging (PCI) were investigated, respectively, with samples prepared from pig knees implanted with low density scaffolds. The results from the computed-tomography (CT)-DEI, CT-ABI, and extended-distance CT-PCI showed the scaffold implanted in pig knee cartilage in situ with structural properties more clearly than conventional PCI and clinical MRI, thus providing information and means for tracking the success of scaffolds in tissue repair and remodeling. To optimize the methods for live animal and eventually for human patients, strategies with the aim to reduce the radiation dose during the imaging process were developed by reducing the number of CT projections, region of imaging, and imaging resolution. The results of the developed strategies illustrate that effective dose for CT-DEI, CT-ABI, and extended-distance CT-PCI could be reduced to 0.3-10 mSv, comparable to the dose for clinical X-ray scans, without compromising the image quality. Taken together, synchrotron X-ray imaging techniques were illustrated promising for developing non-invasive monitoring methods for examining cartilage tissue constructs in live animals and eventually in human patients.
13

Combination of self-assembling peptide hydrogel and autologous chondrocytes for cartilage repair : Preclinical study in a non-human primate model / Combinaison de peptides auto-assemblants et de chondrocytes autologues pour la réparation du cartilage : étude préclinique chez le primate non-humain

Dufour, Alexandre 19 November 2018 (has links)
Le cartilage a une capacité de régénération très limitée car il n'est pas vascularisé. Laréparation de ce tissu est un défi et les techniques chirurgicales actuelles sont insatisfaisantes à longterme. Le cartilage est donc un bon candidat pour l'ingénierie tissulaire. La transplantation dechondrocytes autologues (TCA) a été la première thérapie cellulaire développée en rhumatologie maiscette procédure implique une amplification des cellules qui aboutit à une perte du phénotypechondrocytaire (perte de l'expression du collagène de type II, protéine majoritaire du cartilage), auprofit d'un phénotype fibroblastique (caractérisé par l'expression du collagène de type I, retrouvé dansles tissus fibreux). La TCA conduit donc à une greffe de chondrocytes dédifférenciés produisant unfibrocartilage, dont les propriétés mécaniques sont inférieures à celles du cartilage articulaire.Aujourd'hui, les agences de santé au niveau international s'accordent pour dire que cette procédurenécessite d'être améliorée, par un meilleur contrôle du phénotype cellulaire et l'utilisation debiomatériaux pour mieux combler les lésions articulaires. Il s'agit donc de passer de la thérapiecellulaire à l'ingénierie tissulaire du cartilage.L'objectif de nos travaux a été d'évaluer la capacité d'un gel innovant de peptides autoassemblants,l'hydrogel IEIK13, à jouer le rôle de support pour des chondrocytes humains afin qu'ilsproduisent une matrice cartilage sous l'action de facteurs chondrogéniques. L'objectif visé a été lacréation d'un gel cartilage implantable par arthroscopie. Le défi a été de surmonter la dédifférenciationdes chondrocytes inhérente à leur amplification et incontournable pour augmenter le réservoircellulaire. L'amplification de chondrocytes humains a été réalisée en présence de FGF-2 et d'insuline(cocktail FI) puis leur redifférenciation a été induite en gel IEIK13 sous l'action de BMP-2, d'insuline etd'hormone T3 (cocktail BIT). C'est la combinaison sélective des deux cocktails qui permet la séquencedédifférenciation-redifférenciation. Le phénotype des chondrocytes et la nature de la matriceextracellulaire synthétisée en gel ont été évalués dans un premier temps in vitro, par des analyses dePCR en temps réel, Western-blots et d'immunohistochimie. Dans un second temps, nous avonstransplanté le gel cartilage dans des lésions articulaires de genou d'un modèle original de primate nonhumain(singe cynomolgus), un type de gros animal dont la posture et le fonctionnement desarticulations s'apparentent à l'homme. Nos études d'imagerie non invasive (telle qu'elle est pratiquéechez l'homme) et immunohistochimiques trois mois après implantation montrent une réparationsatisfaisante des lésions, en comparaison avec les lésions laissées non comblées. L'ensemble de nosrésultats montre pour la première fois que l'hydrogel IEIK13 est un biomatériau favorable pourreconstruire le cartilage et que le primate non-humain est un modèle préclinique unique pour évaluerl'efficacité de l'ingénierie tissulaire du cartilage / Cartilage is not vascularized and presents poor capacity of self-regeneration. Repairing thistissue is a challenge and current surgical techniques are not satisfactory in the long term. Cartilage isthus a good candidate for tissue engineering. Autologous chondrocyte transplantation (ACT) was thefirst cell therapy developed for cartilage repair. This procedure implies amplification of cells whichresults in chondrocyte dedifferentiation (loss of expression of type II collagen, the major protein ofcartilage and acquisition of expression of type I collagen, the major protein found in fibrous tissues).Thus, ACT results in implantation of fibroblastic cells producing fibrocartilage with biomechanicalproperties inferior to native articular cartilage. The international health agencies agree that ACT needsto be improved with better control of the chondrocyte phenotype and use of biomaterials. Therefore,cell therapy of cartilage needs to move towards tissue engineering of cartilage.The objective of our study was to evaluate the capacity of an innovative self-assemblingpeptide (IEIK13) to support cartilage matrix production by human chondrocytes. Our goal was to createa cartilage gel that can be implanted by arthroscopy. A main challenge was to meet the problem ofchondrocyte dedifferentiation induced by cell amplification necessary to increase the cellularreservoir. Amplification of human chondrocytes was performed in the presence of FGF-2 and insulin(cocktail FI), and redifferentiation was subsequently induced in IEIK13 gel with BMP-2, insulin, andtriiodothyronine T3 (cocktail BIT). The specific combination of these two cocktails alloweddedifferentiation-redifferentiation of chondrocytes. The status of the chondrocyte phenotype and thenature of the extracellular matrix secreted in gel were first assessed in vitro by real-time PCR, Westernblottingand immunhostochemistry analyses. With a view of clinical application, we then transplantedIEIK13-engineered cartilages into defects created in knees of an original model of non-human primate(cynomolgus monkey), a type of large animal whose anatomy and biomechanics mimic human. Ournon-invasive imaging analyses and our inmmunohistochemical studies performed three months afterimplantation show correct reparation of the lesions, in comparison with the defects left untreated.Altogether, our results demonstrate for the first time that IEIK13 is a suitable biomaterial for cartilagerepair and that cynomolgus monkey represents a unique preclinical model to evaluate efficiency ofcartilage tissue engineering.
14

Exploring New Therapeutic Strategies for Osteoarthritis: From Genetic Manipulation of Skeletal Tissues to Chemically-modified Synthetic Hydrogels

Huang, Henry 31 March 2017 (has links)
Osteoarthritis (OA), a degenerative disease of articular joints, is the leading cause of chronic disability in the US and affects more than a third of adults over 65 years old. Due to the obesity epidemic and an aging population, the prevalence of OA is expected to rise in both young and old adults. There are no disease modifying OA drugs. Therefore, providing any treatment options that delay the onset or progression of OA is highly desirable. The scope of this dissertation examines two different strategies to promote translational therapies for OA. The first approach investigated whether Smad ubiquitin regulatory factor 2 (Smurf2), an E3 ubiquitin ligase, could be a potential therapeutic target for OA. The second approach examined the incorporation of small chemical residues to enhance the physical and bioactivity of a bioinert scaffold for cartilage tissue repair. Overexpression of Smurf2 in chondrocytes was shown to accelerate spontaneous OA development in mice. We hypothesized that reduced Smurf2 expression could slow the progression of OA and enhance the performance of cells for cartilage repair. By performing surgical destabilization of the medial meniscus (DMM) on Smurf2-deficient mice, loss of Smurf2 was shown to mitigate OA changes in young mice but this protection diminished in older mice. Assessment of Smurf2-deficient chondrocytes in vitro revealed an upregulation of chondrogenic genes compared to wild-type; however, these differences were not seen at the protein level, deterring its potential use for cell-based therapies. During the course of this study, new insights about how age and sex affects different joint compartments in response to DMM surgery were also uncovered. These results broadened existing understanding of DMM-induced OA in mice but also questioned the validity of such a model to identify disease modifying targets that are translatable to OA in humans with advanced age. Due to a lack of innate repair mechanisms in cartilage, damage to cartilage increases the risk of developing OA early. Tissue engineering provides a unique strategy for repairing damaged cartilage by delivering cells in a well-controlled environment that can promote the formation of neotissue. We hypothesized that synthetic chemical residues could enhance the mechanical properties of a bioinert scaffold and promote matrix production of encapsulated chondrocytes. Covalent incorporation of small anionic or zwitterionic chemical residues in a polyethylene glycol-based hydrogel improved its stiffness and resistance to fluid flow, however, the resulting physical environment can also exert a dominant negative effect on matrix production of encapsulated chondrocytes. These results suggest that modulating the biosynthesis of chondrocytes with biochemical signals requires a concurrent reduction in any conflicting mechanotransduction signaling, emphasizing the importance of a degradable system to promote new cartilage formation. In summary, this dissertation establishes Smurf2 as a modulator of OA progression but implies that other factors such as age or protein(s) with redundant Smurf2 functions may play a role in limiting its effect as a therapeutic target. This work also reveals fundamental biology about how chondrocytes behave in response to physical and chemical cues in their microenvironment, which will aid in the design of better scaffolds for cartilage tissue engineering.
15

Multifactorial Media Analysis via Design of Experiment for Type II Collagen in Primary Rabbit Chondrocytes

Velez Toro, Javier A 01 January 2021 (has links)
Osteoarthritis is a prevalent disease that affects the articular cartilage of the joints. Millions of people suffer worldwide and it is a major cause of disability in the United States. Current research for treatments of osteoarthritis are studying tissue-engineered cartilage in vitro generated by articular chondrocytes. A challenge faced in vitro for cartilage tissue engineering is the failure of chondrocytes to produce adequate expression of type II collagen. Surprisingly, the media commonly used in vitro lacks 14 vitamins and minerals present in the physiological environment of chondrocytes. Therefore, studying the interactions between micronutrients and chondrocytes may help in potentially increasing the amount of type II collagen expressed by these cells. This project studied the combinatorial effects of vitamins and minerals in defined chondrogenic media on type II collagen expression. Linolenic acid was determined to have predominantly negative effects on chondrogenesis and Vitamin B7 to have beneficial effects. Multiple vitamins and minerals displayed significant interactions, both positive and negative.
16

Potentialité des cellules stromales de la gelée de Wharton en ingénierie du cartillage / Potentiality of stromal cells from wharton’s jelly in cartilage engineering

Reppel, Loïc 24 October 2014 (has links)
Les cellules stromales/souches mésenchymateuses de la gelée de Wharton humaines (CSM-WJ) représentent une source abondante et intéressante de cellules souches pour des applications en ingénierie cellulaire et tissulaire. Leur origine fœtale leur confère des caractéristiques spécifiques par rapport aux cellules stromales/souches mésenchymateuses isolées à partir de moelle osseuse humaine (CSM-MO). Tout d'abord, le but de ce travail est d'optimiser les conditions de culture des CSM-GW pour leur utilisation clinique ultérieure. Nous nous concentrons sur l'influence de la concentration en oxygène lors de l'expansion en monocouche de P1 à P7 sur plusieurs paramètres permettant de caractériser les CSM. Les résultats obtenus sont comparés à ceux obtenus avec les CSM-MO. Notre travail a montré des différences entre les deux sources cellulaires en termes de prolifération et de différenciation adipocytaire. D’après nos résultats, l'hypoxie, au cours de l'expansion, est un paramètre important à prendre en compte en ce qui concerne la prolifération et le potentiel de différenciation chondrocytaire. L'influence des facteurs obstétricaux sur les caractéristiques des CSM-GW est également explorée. Cette étude se situant également dans le cadre de l’ingénierie tissulaire du cartilage, la seconde phase du projet consiste à induire la différenciation des cellules en chondrocytes en ensemençant ces dernières dans un biomatériau à base d’alginate et d’acide hyaluronique, et sur une cinétique de 28 jours. Les résultats obtenus sont comparés à ceux obtenus avec les CSM-MO. Après 4 semaines de culture, les CSM-GW sont capables de s'adapter à leur environnement et d’exprimer des gènes et des protéines matriciels spécifiques du cartilage tels que le collagène de type 2, qui se trouve plus exprimé après différenciation à partir des CSM-GW qu’à partir de CSM-MO / Mesenchymal Stromal/Stem Cells from human Wharton’s jelly (WJ-MSC) are an abundant and interesting source of stem cells for applications in cell and tissue engineering. Their fetal origin confers specific characteristics compared to Mesenchymal Stromal/Stem Cells isolated from human bone marrow (BM-MSC). First, the aim of this work is to optimize WJ-MSC culture conditions for their subsequent clinical use. We focus on the influence of oxygen concentration during monolayer expansion on several parameters to characterize MSC. The results are compared to those obtained with BM-MSC. Our work distinguishes WJ-MSC from BM-MSC in terms of proliferation and adipogenic differentiation. Considering our results, hypoxia during cell expansion is an important parameter to take into account regarding proliferation potential but also chondrogenic differentiation potential. The influence of obstetric factors on WJ-MSC characteristics is also explored. In cartilage tissue engineering context, the second phase of the project is to induce cell differentiation into chondrocytes by seeding them in Alginate/Hyaluronic Acid hydrogel scaffold, and during 28 days. The results obtained are compared to those obtained with BM-MSC. After 4 weeks of culture, WJ-MSC are able to adapt to their environment and express specific cartilage-Related genes and matrix proteins such as type 2 collagen, which is found more expressed after differentiation fromWJ-MSC, than from BM-MSC
17

Development of hydrodynamically engineered cartilage in response to insulin-like growth factor-1 and transforming growth factor-beta1: formation and role of a type I collagen-based fibrous capsule

Yang, Yueh-Hsun 20 September 2013 (has links)
Articular cartilage which covers the surfaces of synovial joints is designed to allow smooth contact between long bones and to absorb shock induced during joint movement. Tissue engineering, a means of combining cells, biomaterials, bioreactors and bioactive agents to produce functional tissue replacements suitable for implantation, represents a potential long-term strategy for cartilage repair. The interplay between environmental factors, however, gives rise to complex culture conditions that influence the development of tissue-engineered constructs. A fibrous capsule that is composed of abundant type I collagen molecules and resembles fibrocartilage usually forms at the outer edge of neocartilage, yet the understanding of its modulation by environmental cues is still limited. Therefore, this dissertation was aimed to characterize the capsule formation, development and function through manipulation of biochemical parameters present in a hydrodynamic environment while a chemically reliable media preparation protocol for hydrodynamic cultivation of tissue-engineered cartilage was established. To this end, a novel wavy-wall bioreactor (WWB) that imparts turbulent flow-induced shear stress was employed as the model system and polyglycolic acid scaffolds seeded with bovine primary chondrocytes were cultivated under varied biochemical conditions. The results demonstrated that tissue morphology, biochemical composition and mechanical strength of hydrodynamically engineered cartilage were maintained as the serum content decreased by 80% (from 10% to 2%). Transient exposure of the low-serum constructs to exogenous insulin-like growth factor-1 (IGF-1) or transforming growth factor-β1 (TGF-β1) further accelerated their development in comparison with continuous treatment with the same bioactive molecules. The process of the capsule formation was found to be activated and modulated by the concentration of serum which contains soluble factors that are able to induce fibrotic processes and the capsule development was further promoted by fluid shear stress. Moreover, the capsule formation in hydrodynamic cultures was identified as a potential biphasic process in response to concentrations of fibrosis-promoting molecules such as TGF-β. Comparison between the capsule-containing and the capsule-free constructs, both of which had comparable tissue properties and were produced by utilizing the WWB system in combination with IGF-1 and TGF-β1, respectively, showed that the presence of the fibrous capsule at the construct periphery effectively improved the ability of engineered cartilage to integrate with native cartilage tissues, but evidently compromised its tissue homogeneity. Characterization of the fibrous capsule and elucidation of the conditions under which it is formed provide important insights for the development of tissue engineering strategies to fabricate clinically relevant cartilage tissue replacements that possess optimized tissue homogeneity and properties while retaining a minimal capsule thickness required to enhance tissue integration.
18

Optimisation de dispositifs médicaux thérapeutiques implantables pour l'ingénierie tissulaire osseuse et cartilagineuse / Implantable therapeutic medical device optimisation for bone and cartilage tissue engineering

Wagner, Quentin 15 December 2017 (has links)
Notre équipe a optimisé la formulation de dispositifs médicaux implantables pour l’ingénierie tissulaire osseuse et cartilagineuse. A ces fins, nous nous sommes basés sur des implants nanostructurés d’origine naturelle ou synthétique conçus au sein du laboratoire par la méthode d’électrospinning, pour imiter la matrice extracellulaire du compartiment osseux, et un hydrogel composé d’alginate et d’acide hyaluronique imitant la composition du compartiment cartilagineux. Dans une première partie de mon travail, pour la régénération osseuse, nous avons optimisé la formulation d’un implant nanostructuré à base de chitosane pour une accélération de cette régénération. Ceci a été possible en rendant actif ce dispositif médical implantable par incorporation de nanoparticules de silice, conférant à la construction nanocomposite des propriétés mécaniques accrues, et une excellente biocompatibilité avec le tissu hôte. Une autre étude pour la même visée a permis d’élaborer une nouvelle stratégie d’ensemencement de dispositif implantable synthétique et nanostructuré par des microtissus cellulaires, remplaçant un ensemencement de cellules isolées et permettant des performances de minéralisation accrues à l’intérieur de l’implant. Dans un deuxième temps, pour la régénération de l’unité ostéoarticulaire, nous avons proposé deux implants bi-compartimentés et hybrides comportant des microtissus de cellules souches mésenchymateuses. Ces implants sont composés d’un hydrogel contenant les cellules souches permettant la régénération du cartilage, et d’une membrane collagénique naturelle (Bio-Gide®) ou synthétique (membrane de polycaprolactone), dotée de nanoréservoirs (technologie brevetée par le laboratoire) de facteur de croissance ostéogénique (BMP-7) pour une régénération du socle osseux (os sous-chondral) de l’unité os-cartilage. La troisième partie de mon travail a concerné la vascularisation des implants osseux et particulièrement l’accélération du recrutement vasculaire. Dans ce cadre plus vasculaire, nous avons proposé une stratégie qui vise à doter un implant synthétique nanostructuré de facteur de croissance angiogénique (VEGF), puis à lui appliquer un ensemencement séquentiel de cellules mésenchymateuses adultes « ostéoblastes humains» et de cellules endothéliales humaines (HUVECs). Cette stratégie a permis un recrutement et une hiérarchisation accrue des cellules endothéliales dans l’implant. En conclusion, l’optimisation des implants développés au laboratoire permettra sans nul doute de proposer dans un futur proche de nouveaux dispositifs médicaux implantables (DMI) thérapeutique combinés de type DMI-MTI (Médicaments de Thérapie Innovante) pour l’ingénierie tissulaire osseuse et cartilagineuse en particulier en médecine régénérative ostéo-articulaire. / Our team optimized the formulation of implantable medical devices for bone and cartilage tissue engineering. To that end, we based our work on nanostructured implants, either natural or synthetic, made in the laboratory by electrospinning process, to mimic bone extracellular matrix, and hydrogel of alginate/hyaluronic acid to mimic cartilage extracellular matrix. First, concerning bone regeneration, we optimized the formulation of a nanostructured scaffold composed of natural chitosan to enhance bone regeneration. This was made possible by doping this implantable medical device with silica nanoparticles, offering this nanocomposite better mechanical properties, and excellent biocompatibility with host tissue. Another study with the same aim allowed elaborating a new cell seeding strategy, to seed these implantable medical devices with cell microtissues instead of single cells, offering higher mineralisation efficiencies within the implant. Consequently, for the regeneration of the osteochondral unit, we proposed two compartmented and hybrid implants comprising mesenchymal stem cells microtissues. Those implants are made of a hydrogel containing the stem cells, allowing the regeneration of cartilage, and a membrane, either natural (collagenic Bio-Gide®) or synthetic (electrospun polycaprolactone) equipped with nanoreservoirs (technology patented by the laboratory) of osteogenic growth factor (BMP-7) for the regeneration of osseous stand (the subchondral bone) of the bone-cartilage unit. Finally, to study the improvement in vascular recruitment, we proposed a new strategy combining the modification of an implantable device with angiogenic growth factor (VEGF), prior to its sequential seeding with mesenchymal cells “human osteoblasts” and human endothelial cells (HUVECs). This strategy allowed higher recruitment and structuration of endothelial cells within the implant. To conclude, the implant optimisation strategies developed in the laboratory will certainly allow proposing in the near future new combined Advanced Therapy Medicinal Products (ATMPs) and Implantable Medical Device for bone and cartilage regeneration, in particular in the field of osteoarticular regenerative nanomedicine.
19

Évaluation des caractéristiques des hydrogels d’alginate supplémentés en acide hyaluronique ou en hydroxyapatite lors de la différenciation des cellules souches mésenchymateuses issues de la gelée de Wharton / Evaluation of characteristics of alginate/hyaluronic acid and alginate/hydroxyapatite hydrogels during differentiation of Wharton's Jelly mesenchymal stem cells

Yu, Hao 18 July 2017 (has links)
Dans le domaine de l'ingénierie du cartilage, les hydrogels à base d'alginate (Alg) et de cellules souches mésenchymateuses (CSM) sont utilisés comme biomatériaux pouvant être utilisés pour combler des lésions cartilagineuses plus ou moins profondes. Cependant, pour reproduire l’organisation zonale du cartilage, des biomatériaux multiphasiques sont nécessaires. Afin de guider la différenciation des CSM dans les différentes strates du biomatériau, sans apports de facteurs de croissance, des composants naturels du cartilage (acide hyaluronique, HA) ou de la matrice osseuse (hydroxyapatite, Hap) peuvent être ajoutés à l’alginate. L’objectif de ce travail de thèse consiste à analyser l’impact de la composition de biomatériaux à base d’alginate enrichi soit en HA soit en Hap sur le comportement des CSM. La première partie de notre travail à consister à évaluer le comportement des CSM issues de la gelée de Wharton dans ces hydrogels. Nos résultats mettent en évidence que les hydrogels d’Alg/Hap possèdent non seulement de meilleures propriétés mécaniques que les hydrogels Alg/HA et favorisent la viabilité des CSM ainsi que leur différenciation par rapport aux CSM ensemencées dans un hydrogel d’Alg/HA. La méthode de stérilisation du biomatériau représente une étape incontournable, dont on doit impérativement évaluer les multiples effets, en particulier pour ce qui touche au comportement des cellules, mais aussi au maintien de l’intégrité des propriétés physicochimiques de l'hydrogel. Ainsi, dans une seconde partie du travail, nous avons montré que le traitement de stérilisation par autoclave induisait un effet négatif sur les caractéristiques initiales de l'hydrogel à base d'alginate. Il ressort également de cette investigation sur les modes de stérilisation, que la stérilisation des hydrogels avec des UV est plus efficace et permet de préserver au mieux les propriétés spécifiques de l'hydrogel, notamment de l’Alg/HA. Enfin, dans une troisième partie de notre travail, nous avons évalué l’évolution des propriétés mécaniques au cours de la différenciation et l’impact de celles-ci sur la différenciation des CSM ainsi que sur leurs propriétés immunomodulatrices. À partir de ces résultats, nous avons montré que les caractéristiques physico-chimiques des hydrogels d’Alg/ha et Alg/hap influençaient non seulement le potentiel de différenciation des CSM-GW mais également la sécrétion des facteurs solubles impliqués dans l’immunomodulation. Ces propriétés physico-chimiques étant influencées dès le procédé de stérilisation, il est alors conseillé de les prendre en compte dans toutes les étapes de l’ingénierie tissulaire / In the field of cartilage engineering, alginate (Alg)-based hydrogels and mesenchymal stem cells (MSC) are widely used as raw biomaterials and stem cells which can be used to fill cartilage lesions of varying depth. However, to reproduce the zonal organization of articular cartilage, a graft multilayer is necessary. In order to guide the differentiation of MSCs in different strata of the biomaterials, without input of growth factors, natural cartilage components (hyaluronic acid, HA) or bone matrix (hydroxyapatite, Hap) can be added into the alginate. The aim of this work is to analyze the impact of the composition of alginate enriched either in HA or in Hap on the behavior of MSCs. The first part of our work is to evaluate the behavior of WJ-MSCs into these hydrogels. Our results have shown that Alg/ Hap hydrogels not only possess better mechanical properties than Alg/HA hydrogels, but also promote the viability of MSCs and their differentiation from MSC seeded into the Alg/HA hydrogel. The sterilization method of biomaterial is an essential step, the multiple effects of which must be evaluated, in particular as regards the behavior of the cells, but also to maintain the integrity of the physicochemical properties of hydrogel. Thus, in a second part of this work, we showed that the autoclave sterilization treatment induced a negative effect on the initial characteristics of alginate hydrogel. It is also apparent from this investigation of the sterilization modes that the sterilization of hydrogels with UV is more efficient and makes it possible to preserve the specific properties of the hydrogel as best as possible, in particular Alg/HA. Finally, in a third part of our work, we also evaluated the evolution of the mechanical properties during the differentiation and the impact of these on the differentiation of MSCs and their immunomodulatory properties. From these results, we have shown that the physico-chemical characteristics of Alg / ha and Alg/hap hydrogels influence not only the differentiation potential of WJ-MSC but also the secretion of soluble factors involved in immunomodulation. Since these physicochemical properties are influenced by the sterilization process, it is advisable to take them into account in all stages of tissue engineering
20

Nouvelles stratégies thérapeutiques des affections articulaires du cheval : évaluation du potentiel thérapeutique des chondrocytes autologues et des cellules souches de cordon ombilical (sang et gelée de Wharton) : vers l'industrialisation de cellules médicaments. / New therapeutic strategies for articular disorders in the equine model : therapeutic potential evaluation of autologous chondrocytes and umbilical cord stem cells (from umbilical cord blood and Wharton jelly) : toward industrialization of drug cells

Rakic, Rodolphe 05 September 2017 (has links)
Les affections articulaires touchant le cartilage, telles que les lésions focales et l’arthrose, correspondent aux principales causes de baisse de performance et d’arrêt prématuré de la carrière sportive du cheval. Ainsi, le traitement des affections du cartilage représente un enjeu vétérinaire majeur dans le monde équin, du fait des importantes pertes financières qu’elles occasionnent à la filière. Les faibles capacités de réparation intrinsèque du cartilage, ainsi que l’absence de thérapie à long terme des dommages cartilagineux, nécessitent le recours à des thérapies de nouvelles générations telle que l’ingénierie tissulaire du cartilage. Dans ce cadre, notre étude s’est attachée à comparer différents types cellulaires pour la génération de cartilage in vitro, afin d’envisager une implantation pour traiter les atteintes cartilagineuses chez le cheval. Une technique initialement développée chez l’Homme, la transplantation de chondrocytes autologues, représente toujours un « gold standard » en ingénierie tissulaire du cartilage. Dans ce travail de thèse, après avoir développé une nouvelle génération de substitut cartilagineux de haute qualité biologique, à partir de chondrocytes articulaires équins, des limites techniques et biologiques inhérentes au type cellulaire persistent. Ainsi, nos travaux se sont tournés vers la recherche de types cellulaires alternatifs. Les cellules souches/stromales mésenchymateuses (CSM) néonatales issues de cordon ombilical telles que les CSM de sang placentaire (CSM-SPL) et les CSM de gelée de Wharton (CSM-GW) pourraient représenter un avantage thérapeutique du fait de leur isolement non-invasif, de leur forte prolifération cellulaire et de leur capacité de différenciation en chondrocyte. Il est néanmoins indispensable de définir le meilleur candidat thérapeutique, parmi ces deux sources cellulaires, pour l’obtention d’un substitut cartilagineux de qualité biologique optimale. Ces résultats de thèse ont montré d’importantes différences dans le processus de chondrogenèse de ces deux sources de CSM néonatales et plaident en faveur de l’utilisation des CSM-SPL dans le cadre d’une stratégie thérapeutique d’ingénierie tissulaire du cartilage équin. Ces travaux ont permis une meilleure compréhension de la biologie du chondrocyte et des CSM. De surcroît, ces travaux permettent d’envisager de futurs essais cliniques chez le cheval, afin de traiter les affections articulaires de ce modèle gros animal. / Articular cartilage disorders, such as focal defects and osteoarthritis, are the main causes of decreased performance or early retirement of sport- and racehorses. Thus, cartilage disorders represent a major veterinary issue in the equine industry, due to significant financial losses. Poor intrinsic cartilage repair properties and the absence of long- term therapy for cartilage defects lead to the development and use of new generation therapies such as autologous chondrocytes implantation. In this context, our study aimed to compare different cell types for the in vitro cartilage generation, in order to implant the biological substitute to treat cartilage defects in the horse. A therapeutic strategy initially developed in human medicine, the autologous chondrocytes transplantation, always represents a "gold standard" in cartilage tissue engineering. In the present study, after developing a new generation of cartilaginous substitute of high biological quality, composed of equine articular chondrocytes, technical and biological limits inherent to the cell type persist. Thus, we have used alternative cell types such as neonatal mesenchymal stem/stromal cells (MSCs) from umbilical cord, such as umbilical cord blood MSC (UCB-MSCs) and umbilical cord matrix or Wharton jelly MSCs (UCM- MSCs). These MSCs sources could represent a therapeutic advantage due to their non-invasive isolation, their high cell proliferation and their ability to differentiate into chondrocytes. Nevertheless, it is essential to define the best therapeutic candidate between these two MSCs sources, to obtain an optimal quality for the neocartilaginous substitute. Our data highlighted important differences in the chondrogenesis process of these two neonatal MSCs sources, allowing us to consider UCB-MSCs as the best therapeutic candidate for equine cartilage tissue engineering. This work allows a better understanding of the chondrocyte and MSCs biology. Moreover, this work leads the way to setting-up future clinical trials in the horse, in order to treat articular defects of this large animal model.

Page generated in 0.108 seconds