• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 101
  • 98
  • 47
  • 15
  • 7
  • 7
  • 5
  • 5
  • 4
  • 1
  • Tagged with
  • 313
  • 313
  • 140
  • 125
  • 93
  • 82
  • 63
  • 59
  • 57
  • 56
  • 55
  • 40
  • 38
  • 32
  • 30
  • 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.
281

Isolation et caractérisation des cellules stromales mésenchymateuses multipotentes du tissu adipeux: Étude des sous-populations et comparaison avec la moelle osseuse. / Isolation and characterization of multipotent mesenchymal stromal cells from adipose tissue: study of sub-populations and comparison with bone marrow.

Busser, Hélène 14 December 2015 (has links)
Multipotent mesenchymal stromal cells (MSC) were first discovered in bone marrow and can be isolated from “virtually all organs”. They could participate in tissue maintenance and self-renewing process. They are able to adhere to plastic surfaces and acquire a fibroblastic shape when isolated. They are characterized by a particular phenotype and are able to differentiate into several cell types if cultivated in a specific induction medium. These characteristics were defined on MSC in culture and do not represent how they may be in situ.MSC present particular properties. They can secrete growth factors and several cytokines that give them a trophic activity on one hand and the ability to modulate the immune system on the other hand. They are also able to differentiate. These different properties make them an attractive candidate for cell therapy.MSC are already the focus of several pre-clinical and clinical studies. Nevertheless, the results of these studies are difficult to interpret due to limited understanding of their basic biology. MSC are poorly defined in situ and are heterogeneous. Their heterogeneity is dictated by their tissue of origin and cell preparation. To date, there is no standard protocol for MSC isolation and culture. This leads to numerous questions regarding patient safety, and these questions require answers.The first part of the work deals with the methods used to optimize the extraction of MSC and purification from adipose tissue, one of the main sources of autologous MSC with bone marrow. Classical methods require an enzymatic digestion step. The enzyme used and the duration of adipose tissue digestion time can induce cellular alterations and modify cell functions. Moreover, the addition of a xenobiotic increases the risk of contamination and complicates the monitoring of good manufacturing practices (GMP). We propose a method that does not require this enzymatic digestion step while being easier, safer, faster, gentler and less expensive. Compared to the classical enzymatic method, our method yields an equivalent number of MSC from adipose tissue while preserving their properties.The second part of this work focuses on the characterization of the MSC subpopulations from adipose tissue and compares them to those from bone marrow, which are the historical gold standard. The study made it possible to deepen the knowledge of MSC surface markers in situ from these 2 sources. It also evaluated the various properties of the isolated subpopulations thanks to the cell surface markers CD271, SUSD2, MSCA-1, CD44 and CD34. We showed that MSC from bone marrow express MSCA-1, CD271 and SUSD2 markers in situ. We also found that a population clearly positive for the CD34 does exist in situ with different properties compared to those of the unselected populations or the negative counterpart. 2 populations that are negative and positive for CD44 also exist with similar properties.In contrast to bone marrow MSC, only one selection was able to effectively isolate MSC from adipose tissue by a positive selection based on the expression of CD34. We also isolated a CD271+ population but only from lipoaspirate samples and not from abdominoplasty samples. Collectively, our results suggest that MSCA-1 seems to be the best marker through which to isolate MSC from bone marrow and that CD34 is the only marker able to positively isolate cells from adipose tissue. Thus, we show that the MSC from the different sources share similar properties although they have specific characteristics. The choice of the source and of the marker with which to isolate a particular subpopulation is important depending on their intended clinical use. / Les cellules stromales mésenchymateuses multipotentes (CSM) ont été mises en évidence dans la moelle osseuse et peuvent être isolées de « virtuellement tous les organes ». Elles participeraient à la maintenance et au renouvellement des tissus. Une fois isolées, elles sont capables d’adhérer à des surfaces en plastique en prenant une forme fibroblastique. Elles sont caractérisées par un phénotype particulier et peuvent se différencier en divers types cellulaires lorsque cultivées dans un milieu d’induction spécifique. Ces caractéristiques ont été définies sur les CSM en culture et ne reflètent pas forcément ce qui se passe in situ.Les CSM présentent des propriétés particulières. Elles peuvent sécréter des facteurs de croissance ainsi que de nombreuses cytokines qui leur permettent d’une part d’avoir une activité trophique et d’autre part de moduler le système immunitaire. Elles sont aussi capables de se différencier. Ces différentes propriétés les rendent particulièrement attractives pour la thérapie cellulaire.Les CSM font déjà l’objet de nombreuses études pré-cliniques et cliniques dont les résultats sont difficilement interprétables car nous n’avons à l’heure actuelle qu’une compréhension limitée de leur biologie de base. Les CSM sont encore mal définies in situ et sont hétérogènes. Cette hétérogénéité provient de leur différence d’origine et de leur préparation cellulaire :il n’existe aucune standardisation des protocoles d’isolation et de culture. Cette hétérogénéité entraine de nombreuses questions relatives à la sécurité du patient qui doivent être élucidées.La première partie de ce travail cherche à optimiser les méthodes d’extraction et de purification des CSM du tissu adipeux humain, la principale source de CSM autologues avec la moelle osseuse. Les méthodes classiques requièrent une étape de digestion enzymatique dont l’enzyme utilisée et le temps de digestion du tissu adipeux peuvent induire des altérations cellulaires et modifier leurs fonctions. De plus, l’adjonction de xénobiotiques augmente le risque de contamination et complique le suivi des bonnes pratiques de fabrication (BPF). Nous proposons une méthode qui s’affranchit de cette étape de digestion enzymatique tout en étant plus facile, plus sûre, plus rapide, moins chère et moins traumatisante pour les cellules. Elle permet d’obtenir un nombre tout aussi important de CSM du tissu adipeux que la méthode enzymatique classique en préservant leurs propriétés.La deuxième partie de ce travail vise à caractériser les sous populations de CSM du tissu adipeux humain en les comparant à celles de la moelle osseuse, source de référence historique. Cette étude a permis d’approfondir la connaissance des marqueurs de surface des CSM de ces 2 sources in situ, tout en évaluant les différentes propriétés des sous-populations isolées grâce aux marqueurs de surface CD271, SUSD2, MSCA-1, CD44 et CD34. Nous avons montré que les CSM de la moelle osseuse expriment les marqueurs MSCA-1, CD271 et SUSD2 in situ et qu’il existait une sous-population clairement positive pour le CD34 avec des propriétés différentes de celles de la population non sélectionnée ou négative pour ce marqueur. Il existe aussi 2 sous-populations positive et négative pour le CD44 avec des propriétés similaires.Contrairement aux CSM de la moelle osseuse, une seule sélection a permis d’isoler efficacement les CSM du tissu adipeux par une sélection positive sur base de l’expression du CD34. Nous avons pu aussi isoler une population CD271+ mais seulement des prélèvements de lipoaspirations et non des abdominoplasties.Au vu de nos résultats, MSCA-1 semble le meilleur marqueur pour isoler les CSM de la moelle osseuse tandis que le CD34 est le seul marqueur capable d’isoler positivement celles du tissu adipeux. Ainsi, nous montrons que les CSM issues de différentes sources partagent des propriétés similaires avec cependant des caractéristiques propres. Le choix de la source et du marqueur pour isoler une sous-population sont donc importants en fonction de leur utilité clinique envisagée. / Doctorat en Sciences biomédicales et pharmaceutiques (Médecine) / info:eu-repo/semantics/nonPublished
282

Comparison between therapeutic efficiency of bone marrow derived mononuclear and mesenchymal stem cells in chronic myocardial infarction

Mathieu, Myrielle 05 May 2009 (has links)
<p>Background: Stem cell therapy can facilitate cardiac repair after healed myocardial infarction but the optimal cell type remains uncertain. <p>Aims: To investigate the pathophysiology of heart failure in a canine model of healed myocardial infarction and to compare the efficacy and the safety of autologous bone marrow mononuclear cell (BMNC) transfer and mesenchymal stem cell (MSC) transfer in this model. It was a blind, randomized and placebo control study.<p>Methods: Eleven weeks after coronary ligation, 24 dogs received intramyocardial injections of BMNC, MSC or Placebo (n = 8 per groups). Echocardiography, conductance method, magnetic resonance imaging, serum neurohormones, holter monitoring, macromorphometry, histology and real time quantitative polymerase chain reaction were used to assess cardiac performance, safety and remodelling in healthy animals, before cell transplantation and up to 16 weeks’ follow-up. <p>Results: The model was characterized by decreased left ventricular end-systolic elastance and ventricular-arterial uncoupling without alteration of compliance. <p>Four months after BMNC transfer, the regional systolic function measured at echocardiographic showed a sustained improvement. This improvement was associated with an improved left ventricular end-systolic elastance and a decreased infarct size. Although the left ventricular ejection fraction stayed unchanged, the serum level of N-terminal B-type natriuretic propeptide level decreased. Mononuclear cell transfer was also associated with increased left ventricular relative wall area, increased vascular density, intramyocardial vascular remodelling and upregulation of angiogenic factors gene expression. Mesenchymal stem cell transfer only improved lately and moderately the regional systolic function, without improvement of cardiac contractility or decreased infarct size. <p>Conclusions: In a canine model of chronic myocardial infarction, BMNC transfer is superior to MSC transfer in improvement of cardiac contractility and regional systolic function, and to reduce the infarct size and plasma N-terminal B-type natriuretic propeptide level. Functional improvement is associated with a favourable angiogenic environment and neovascularization. <p> / Doctorat en Sciences biomédicales et pharmaceutiques / info:eu-repo/semantics/nonPublished
283

Modulation du potentiel angiogène des progéniteurs endothéliaux humains par des biomarqueurs plasmatiques vasculaires / Angiogenic potential modulation of human endothelial progenitor cells by vascular plasmatic biomarkers

d'Audigier, Clément 02 October 2013 (has links)
Rationnel : L’implication établie des progéniteurs endothéliaux circulants dans les phénomènes de néovascularisation chez l’adulte a stimulé la recherche de thérapeutiques angiogènes basées sur la greffe de ces cellules. Deux types cellulaires au phénotype endothélial sont actuellement définis entre autres par leur cinétique d’apparition en culture : les progéniteurs précoces (CFU-EC ou CAC) et tardifs (ECFC). Notre équipe a montré que l’injection thérapeutique de cellules mononucléées de moelle osseuse (BM-MNC) permettait la néovascularisation du site ischémié chez des patients atteints d’artériopathie des membres inférieurs, et que les néovaisseaux formés avaient le phénotype d’ECFC. Nous avons dans un premier temps mesuré les concentrations de différentes protéines modulant l’angiogenèse, chez des patients atteints de pathologies ischémiques et cardiovasculaires, ou impliquant des anomalies vasculaires associées à la fibrose. Ainsi, le transforming growth factor - β1 (TGF-β1) dans la fibrose pulmonaire idiopathique (FPI), la thrombospondine-1 (TSP-1) dans l’artériopathie des membres inférieurs (AMI), et le placental growth factor (PlGF) chez les patients atteints de pathologies cardiovasculaires [syndrome coronarien aigu (SCA), ou patients devant subir une chirurgie de la valve ou un pontage coronarien], se sont distingués comme potentiel biomarqueur plasmatique dans ces pathologies, et ont été étudiés dans la biologie des ECFC humaines.Résultats : Le taux plasmatique de TGF-β1 est augmenté chez les patients atteints de FPI par rapport à la population contrôle ; il a un effet pro-angiogène in vivo (vascularisation des implants de Matrigel®) et in vitro (prolifération et migration des ECFC) via les récepteurs ALK-1, ALK-5 et TGF-βRII. Le taux plasmatique de TSP-1 est augmenté chez les patients artéritiques par rapport à la population contrôle. Par ailleurs les néovaisseaux formés de patients artéritiques ayant été traités par injection locale de BM-MNC expriment la TSP-1. Dans les modèles murins de Matrigel®-plugs et d’ischémie du membre inférieur (IMI), la TSP-1 induit une diminution de la vascularisation des implants ainsi qu’une diminution de la revascularisation du membre ischémié. In vitro, la TSP-1 augmente l’adhésion via un mécanisme N-Terminal dépendant, et diminue le potentiel angiogène (prolifération et migration) des ECFC via sa liaison au récepteur CD47, ce qui active la voie de signalisation SDF-1/CXCR4. Le taux plasmatique de PlGF est augmenté chez les patients atteints de SCA par rapport à 2 populations contrôles ; il est également augmenté chez les patients ayant subit une chirurgie cardiaque. Les PlGF-1 et -2 potentialisent la tubulogenèse des ECFC in vitro via la phosphorylation du récepteur VEGFR1. Cet effet est aboli lorsque le VEGFR1 est inhibé par ARN interférence ou par le composé chimique « 4321 ». De plus ce composé « 4321 » inhibe la vascularisation des implants de Matrigel®, ainsi que la revascularisation du membre ischémié dans le modèle d’IMI.Conclusions : Le TGF-β1 joue un rôle dans le remodelage vasculaire de la FPI via les ECFC ; la TSP-1 est un potentiel biomarqueur de l’angiogenèse induite par les ECFC dans l’AMI ; l’inhibition de la voie PlGF/VEGFR1 module la tubulogenèse induite par les ECFC, cellules impliquées dans la formation de nouveaux vaisseaux. Nous avons ainsi mis en évidence 3 protéines modulant l’angiogenèse dans 3 contextes pathologiques différents, caractérisés par un remodelage vasculaire et où les ECFC sont impliquées dans leurs mécanismes physiopathologiques. Ces 3 protéines se présentent donc comme de potentiels biomarqueurs plasmatiques, modulant les propriétés angiogènes des ECFC et pouvant influencer leur efficacité en tant que produit de thérapie cellulaire. Ces protéines jouent un rôle probable dans l’équilibre homéostatique au décours des pathologies concernées. / Rationale: The pro-angiogenic capacities of endothelial progenitor cells are now well established, and their involvement in neovascularization events in adults has stimulated the research in the field of angiogenic therapy based on transplant of these cells. Current data converge towards the notion of two cell types with endothelial phenotype, defined at least by their kinetics of appearance in culture: early endothelial progenitor cells (CFU-EC or CAC) and late (ECFC). Our team has shown that the therapeutic injection of bone marrow mononuclear cells (BM-MNC) led to neovascularization of the ischemic site in patients with critical limb ischemia, and that the new vessels formed bore the phenotype of ECFC. We initially measured the concentrations of different proteins modulating angiogenesis in patients with ischemic and cardiovascular diseases, or involving vascular abnormalities associated with fibrosis. Thus, the transforming growth factor - ß1 (TGF-ß1) in idiopathic pulmonary fibrosis, the thrombospondin-1 (TSP-1) in peripheral artery disease, and the placental growth factor (PlGF) in patients with cardiovascular diseases [acute coronary syndrome (ACS), patients undergoing valve surgery or coronary artery bypass surgery], emerged as potential plasmatic biomarkers in these pathological settings, and have been studied in the biology of human ECFC.Results: TGF-ß1 plasma level is increased in patients with idiopathic pulmonary fibrosis (IPF) compared to the control population; it exerts a pro-angiogenic effect in vivo (vascularization of Matrigel ®-plugs) and in vitro (proliferation and migration of ECFC) via ALK-1, ALK-5 and TGF-ßRII receptors. TSP-1 plasma level is increased in patients with peripheral artery disease (PAD) compared to the control population. In addition, the new vessels formed in PAD patients treated by local injection of BM-MNC express TSP-1. In murine models of Matrigel ®-plugs and hindlimb ischemia, TSP-1 induces a decrease in plugs vascularization and impaired revascularization of ischemic limb. In vitro, TSP-1 increases ECFC adhesion via an N-terminal dependent mechanism and reduces their angiogenic potential (proliferation and migration) via its binding to CD47 receptor, which activates the SDF-1/CXCR4 signaling pathway. PlGF plasma level is increased in ACS patients compared with the control population and stable angina patients and is also increased in patients undergoing cardiac surgery. PlGF-1 and -2 potentiate ECFC tubulogenesis in vitro via phosphorylation of the VEGFR1 receptor. This effect was abolished when the ECFC VEGFR1 is inhibited by RNA interference or by the chemical compound "4321". In addition this compound "4321" inhibits the vascularization of Matrigel ®-plugs, and revascularization of the ischemic limb in the hindlimb ischemia model.Conclusions: TGF-ß1 is involved in the IPF vascular remodeling through ECFC; TSP-1 is a potential biomarker of angiogenesis induced by ECFC in PAD; the inhibition of the PlGF/VEGFR1 pathway modulates ECFC tubulogenesis, cells involved in the formation of new vessels. We thus identified three proteins that modulate angiogenesis in three different pathological settings characterized by a vascular remodeling and where ECFC are involved in their pathophysiology. These three proteins therefore state as potential plasmatic biomarkers, modulating ECFC angiogenic properties and are able to influence their efficacy as a cell therapy product. These plasmatic biomarkers likely play a role in the homeostasis of those pathologies progress.
284

Diferenciação de células-tronco embrionárias murinas (mESCs) em células produtoras de insulina (IPCs) e caracterização funcional do gene Purkinje cell protein 4 (Pcp4) neste processo / Differentiation of murine embryonic stem cells (mESCs) into insulin-producing cells (IPCs) and functional characterization of the Purkinje Cell Protein 4 (Pcp4) gene in this process

Patricia Mayumi Kossugue 28 May 2013 (has links)
Fontes alternativas de células &#946; têm sido estudadas para o tratamento de Diabetes mellitus tipo 1, dentre as quais a mais promissora consiste das células-tronco diferenciadas em células produtoras de insulina (IPCs). Alguns trabalhos demonstram a capacidade de células-tronco embrionárias murinas (mESCs) de formarem estruturas semelhantes a ilhotas pancreáticas, porém, os níveis de produção de insulina são insuficientes para a reversão do diabetes em camundongos diabetizados. Este trabalho visa desenvolver um protocolo adequado para geração de IPCs e contribuir para a identificação e caracterização funcional de novos genes associados à organogênese pancreática. Logo no início da diferenciação das mESCs em IPCs, foi possível verificar o surgimento de células progenitoras, evidenciado pela expressão de marcadores importantes da diferenciação beta-pancreática. Ao final do processo de diferenciação in vitro, ocorreu a formação de agrupamentos (clusters) semelhantes a ilhotas, corando positivamente por ditizona, que é específica para células &#946;-pancreáticas. Para avaliar seu potencial in vivo, estes clusters foram microencapsulados em Biodritina® e transplantados em camundongos diabetizados. Apesar dos níveis de insulina produzidos não serem suficientes para estabelecer a normoglicemia, os animais tratados com IPCs apresentaram melhores condições, quando comparados ao grupo controle, tendo melhor controle glicêmico, ganho de massa corpórea e melhor aparência da pelagem, na ausência de apatia. Além disso, análise dos clusters transplantados nestes animais indicou aumento da expressão de genes relacionados à maturação das células &#946;. Porém, quando estes clusters foram microencapsuladas em Bioprotect® e submetidos à maturação in vivo em animais normais, ocorreu um aumento drástico na expressão de todos os genes analisados, indicando sua maturação completa em células beta. O transplante destas células completamente maturadas em animais diabetizados, tornou-os normoglicêmicos e capazes de responder ao teste de tolerância à glicose (OGTT) de forma semelhante aos animais normais. A segunda parte do trabalho visou analisar genes diferencialmente expressos identificados em estudo anterior do nosso grupo, comparando, através de DNA microarray, mESCs indiferenciadas e diferenciadas em IPCs. Um dos genes diferencialmente expressos é aquele que codifica para a Purkinge cell protein 4 (Pcp4), sendo 3.700 vezes mais expresso em IPCs. Para investigar o possível papel do gene Pcp4 em células &#946; e no processo de diferenciação &#946;-pancreática, adotou-se o enfoque de genômica funcional, superexpressando e inibindo sua expressão em células MIN-6 e mESCs. Apesar da alteração na expressão de Pcp4 em células MIN-6 não ter interferido de forma expressiva na expressão dos genes analisados, quando inibido, modificou o perfil da curva de crescimento celular, aumentando seu tempo de dobramento de forma significativa e diminuindo da viabilidade celular em ensaios de indução de apoptose. Já na diferenciação de mESCs em IPCs, a superexpressão de Pcp4 interferiu de forma positiva apresentando uma tendência a aumentar a expressão dos genes relacionado à diferenciação&#946;-pancreática. Concluindo, desenvolvemos um novo protocolo de diferenciação de mESCs em IPCs as quais foram capazes de reverter o diabetes em camundongos diabetizados e descrevemos, pela primeira vez, o gene Pcp4 como sendo expresso em células &#946;-pancreáticas, podendo estar relacionado à manutenção da viabilidade celular e maturação destas células. / New cellular sources for type 1 Diabetes mellitus treatment have been previously investigated, the most promising of which seems to be the insulin producing cells (IPCs), obtained by stem cells differentiation. Some reports show that murine embryonic stem cells (mESCs) are able to form islet-like structures, however, their insulin production is insufficient to render diabetic mice normoglycemic. This work aims at developing an adequate protocol for generation of IPCs and searching for new genes which could be involved in the pancreatic organogenesis process. Early on during mESCs differentiation into IPCs, we observed the presence of progenitor cells, which were able to express pancreatic &#946;-cell markers. At the end of the differentiation process, the islet-like clusters positively stained for the insulin-specific dithizone. These clusters were microencapsulated in Biodritin® microcapsules, and then transplanted into diabetized mice. Although the levels of insulin production were insufficient for the animals to achieve normoglycemia, those which received IPCs displayed improved conditions, when compared to the control group, as judged by a better glycemic control, body weight gain and healthy fur appearance, in the absence of apathy. In addition, when these transplantated clusters were retrieved, high levels of expression of the genes related to &#946;-cell maturation were detected. IPCs were also microencapsulated in Bioprotect® and subjected to in vivo maturation in normal animals. A dramatic increase of the analyzed genes expression was observed, indicating complete maturation of the differentiated cells. When these cells were transplanted into diabetized mice, these animals achieved normoglycemia and were able to display glucose tolerance test (OGTT) response very similar to that of normal mice. In the second part of this work, we analyzed upregulated genes described in previous work from our group, comparing undifferentiated mESCs to IPCs using a microarray platform. One of these genes is that coding for the Purkinje cell protein 4 (Pcp4), which is 3,700 more expressed than in undifferentiated mESC cells. We adopted a functional genomics approach to investigate the role played by the Pcp4 gene in &#946;-cells and in &#946;-cell differentiation, by inducing overexpression and knocking down this gene in MIN-6 and mESC cells. Although the differential expression of Pcp4 in MIN-6 was not able to interfere with the expression of the genes analyzed, we observed different cell growth rates, with increased doubling time and decreased cell viability when its expression was knocked down. In addition, overexpression of Pcp4 in mESCs subjected to differentiation into IPCs apparently increases the expression of genes related to &#946;-cell differentiation. In conclusion, we developed a new protocol for ESCs differentiation into IPCs, which is able to revert diabetes in diabetized mice, and we also describe here, for the first time, the Pcp4 gene as being expressed in pancreatic &#946;-cells and possibly being related to maintenance of cell viability and &#946;-cell maturation.
285

Rôle fonctionnel des canaux potassiques activés par le calcium au sein de progéniteurs cardiaques : implication en médecine régénérative

Vigneault, Patrick 04 1900 (has links)
L'insuffisance cardiaque (IC) est un processus progressif et inexorable menant au remodelage pathologique du cœur et à la destruction du parenchyme cardiaque. Indépendamment de l'étiologie, on observe une diminution d'environ 30% du nombre de cardiomyocytes ventriculaires au stade terminal de la maladie. Reposant sur les données précliniques convergentes dans les modèles d'IC, le concept novateur de thérapie cellulaire a suscité beaucoup d’espoir en cardiologie. Bien que leur rôle dans l'homéostasie cardiaque soit controversé, les progéniteurs cardiaques endogènes (eCPCs) qui perdurent au sein du myocarde adulte possèderaient les caractéristiques optimales en vue de la régénération myocardique. Nos données électrophysiologiques montrent que le courant potassique dépendant du Ca2+ de conductance intermédiaire (IKCa3.1) est dominant et qu'il contribue à la détermination du potentiel membranaire (Vmem). L'hyperpolarisation engendrée par l'activation du canal KCa3.1 (SK4; KCNN4) maintient le gradient électrique et favorise l'entrée capacitive de Ca2+ (ECC). D'un point de vue fonctionnel, la potentialisation de la signalisation calcique intracellulaire induite par KCa3.1 semble cruciale pour la prolifération des eCPCs c-Kit+. Puisque le statut clinique est connu pour avoir des conséquences néfastes sur la fonctionnalité des cellules souches, nous avons comparé la densité du courant IKCa3.1 dans des eCPCs c-Kit+ provenant de cœurs sains et insuffisants. En accord avec les données électrophysiologiques, nos résultats démontrent que l'insuffisance cardiaque congestive (CHF) diminue significativement l'expression de KCa3.1 ainsi que des protéines régulatrices du cycle cellulaire. Les cellules souches dérivées d'explants cardiaques (EDCs) représentent un autre produit cellulaire prometteur pour la thérapie cellulaire en cardiologie. Les EDCs se composent de sous-populations complémentaires dont la proportion varie en fonction du statut clinique. Alors que la population CD90- constitue la fraction active en termes d'efficacité thérapeutique, il a été démontré qu'une proportion élevée de cellules CD90+ réduit le potentiel régénératif des EDCs. Afin de faire la lumière sur les déterminants ioniques de la thérapie cellulaire cardiaque, les propriétés électrophysiologiques des populations CD90+ et CD90- ont été comparées. Considérant l'importance de KCa3.1 pour la fonction des eCPCs c-Kit+, la présence de canaux potassiques Ca2+-dépendants (KCa) dans les EDCs a été investiguée. Nous avons identifié 2 types de canaux KCa dans les EDCs humaines. Le canal KCa1.1 (BKCa; KCNMA1) est exprimé de façon homogène alors que KCa3.1 est présent exclusivement dans les cellules CD90-. D'un point de vue fonctionnel, l'activité du canal KCa3.1 détermine le Vmem et supporte la prolifération des EDCs. Puisque ce canal est présent uniquement dans la population cardiogénique, l'expression de KCa3.1 pourrait être un facteur déterminant de la capacité régénérative des EDCs. Nous avons investigué cette hypothèse et confirmé que la transplantation de cellules génétiquement modifiées pour exprimer le canal KCa3.1 augmente la régénération cardiaque dans un modèle murin d'IC d'origine ischémique. Pour la première fois, nous avons fait la démonstration que la modulation des propriétés ioniques de cellules souches peut améliorer leur efficacité thérapeutique. / Heart failure (HF) is a progressive disease characterized by extensive pathological remodelling of the heart and myocardial damage. Regardless of the etiology, a decrease of about 30% in the number of ventricular cardiomyocytes is observed at the terminal stage of HF. Based on converging preclinical data in HF models, the innovative concept of cell therapy has generated a great deal of enthusiasm in cardiology. Although the role of cardiac stem cells in cardiac homeostasis is highly controversial, the multipotent progenitors that persist within the adult myocardium possess the ideal characteristics for cardiac regeneration, especially because of their cardiogenic committment. Plasma membrane ion channels are involved in the fundamental processes of virtually all cells that make up the human body, including stem cells. A wide range of functional ion channels was identified in ex vivo proliferated endogenous cardiac progenitor cells (eCPCs), but their function remains poorly understood. We have completed the very first characterization of the ionic profile of freshly-isolated c-Kit+ eCPCs. We found that the intermediate conductance Ca2+-activated potassium current (IKCa3.1) is the predominant conductance and contributes to the determination of membrane potential (Vmem). The hyperpolarization generated by the activation of the KCa3.1 channel (SK4; KCNN4) maintains the electrical gradient and promotes store-operated Ca2+-entry (SOCE) that activates progenitor cell proliferation. Experimental congestive heart failure (CHF) significantly decreased the expression of KCa3.1 as well as cell cycle regulatory proteins. Taken together, these findings suggest that alterations in KCa3.1 may have pathophysiological and therapeutic significance in regenerative medicine In addition to c-Kit+ eCPCs, cardiac explants-derived cells (EDCs) represent another promising cell product for myocardial repair. EDCs are obtained as a heterogeneous mixture composed of complementary subpopulations. Interestingly, it was found that a high proportion of CD90+ cells reduce the functional benefits of EDCs therapy. Consistent with this observation, it has recently been shown that the CD90- population constitutes the active fraction in terms of therapeutic efficacy. In order to gain insight into the ionic determinants of EDCs function, the electrophysiological properties of the CD90+ and CD90- populations were studied. Considering the importance of KCa3.1 in c-Kit+ CPCs, we evaluated the presence of KCa channels in human EDCs. We have identified 2 types of KCa channels in ex vivo expanded EDCs. While KCa1.1 (BKCa; KCNMA1) channel was homogeneously expressed in both subpopulations, KCa3.1 was found exclusively in the CD90- cell fraction. Similar to our previous observations in freshly isolated c-Kit+ eCPCs, KCa3.1 was responsible for the determination of Vmem under resting conditions and during SOCE. Importantly, we demonstrated that transplantation of genetically-modified EDCs to over-express KCNN4 potentiates cardiac regeneration in a murine model of ischemic cardiomyopathy. This study provides the first evidence in the literature that modulating the activity of a single plasma membrane ion channel can truly improves the therapeutic efficacy of progenitor cells.
286

Designing biomaterials for controlled cardiac stem cell differentiation and enhanced cell therapy in the treatment of congestive heart failure / Conception de biomatériaux pour le contrôle de la différenciation cardiaque à partir de cellules souches et pour l’amélioration de la thérapie cellulaire dans le traitement de l’insuffisance cardiaque sévère

Farouz, Yohan 30 September 2015 (has links)
La thérapie cellulaire se positionne comme une stratégie prometteuse pour inciter le cœur infarci à se régénérer. A cet effet, des études récentes placent des espoirs considérables dans l’utilisation des cellules souches embryonnaires et notre laboratoire a déjà démontré comment les différencier en progéniteurs cardiovasculaires, un type de précurseurs cellulaires qui ne peut aboutir qu’à la formation de cardiomyocytes, de cellules endothéliales ou de cellules de muscles lisses. Cet engagement précoce réduit leur capacité de prolifération anarchique et en même temps leur permet de rester suffisamment plastiques pour éventuellement s’intégrer plus facilement avec le tissue hôte. Cependant, les études précliniques et cliniques d’injection de ces cellules s’avérèrent décevantes. Malgré de légères améliorations de la fonction cardiaque, on observa une trop faible survie cellulaire ainsi qu’un taux de rétention des cellules dans le myocarde remarquablement bas. Afin d’étudier ce problème, mes travaux de thèse ont porté non seulement sur la conception de nouveaux biomatériaux pouvant servir de moyen de transport et d’intégration des cellules dans la zone infarcie, mais aussi sur la conception de biomatériaux permettant de contrôler précisément l’environnement cellulaire au cours du processus de différenciation de cellules souches pluripotentes humaines en cardiomyocytes. Grâce aux importantes interactions entre nos laboratoires de recherche fondamentale et de recherche clinique, nous avons tout d’abord développé de nouvelles techniques de fabrication et de caractérisation de patches de fibrine cellularisés qui sont récemment entrés dans un essai clinique de phase I. A partir de cette formulation clinique approuvée par les autorités de régulation, nous avons élaboré toute une gamme de matériaux composites uniquement à base de matières premières pertinentes dans ce cadre clinique, dans le but d’améliorer la maturation des progéniteurs cardiovasculaires une fois greffés sur le cœur défaillant. Dans cette optique, nous avons également développé un modèle in vitro permettant d’étudier précisément l’influence combinée de la rigidité du substrat et du confinement spatial sur la différenciation des cellules souches en cardiomyocytes. Grâce à des techniques de microfabrication sur substrat mou, il a été possible de positionner précisément les cellules souches pluripotentes dans des espaces restreints d’élasticité variable. Ainsi, nous avons pu observer que même en utilisant des protocoles chimiques éprouvés basés sur la modulation de cascades de signalisation impliquées dans le développement cardiaque, une très forte hétérogénéité pouvait apparaître en fonction de l’environnement physique des cellules. Nous avons ainsi pu extraire les caractéristiques principales permettant une différenciation cardiaque efficace, reproductible et standardisée et les avons appliquées à la fabrication d’une nouvelle génération de patches composés de matériaux cliniques et de couches multiples de bandes synchrones de cardiomyocytes. De fait, ces travaux ouvrent de nouvelles voies dans l’utilisation de biomatériaux pour la production industrielle de cardiomyocytes et pour la fabrication de patches cliniques, cellularisés ou non, dans le traitement de l’insuffisance cardiaque. / Cell therapy is a promising strategy to help regenerate the damaged heart. Recent studies have placed a lot of hopes in embryonic stem cells and our lab had previously found a way to differentiate them into cardiac progenitors, cells that can only differentiate into cardiomyocyte, endothelial cells or smooth muscle cells. This early commitment decreases their proliferative capabilities, yet maintains their plasticity for better integration inside the host tissue. However, clinical and pre-clinical injection studies did not really meet the expectations. Even though slight improvements in cardiac function were demonstrated, very low cell viability has been observed, as well as a very low retention of the cells inside the myocardium. To address this problem, my PhD projects not only focus on the design of new biomaterials to act as a vehicle for cell delivery and retention in the infarcted area, but also on the design of biomaterials that control the cellular environment during the differentiation of pluripotent stem cells into cardiomyocytes. Going back and forth between the labs and the clinics, we first developed new techniques for the fabrication and the characterization of a cell-laden fibrin patch that is now undergoing phase I clinical trial. From the approved clinical formulation, we then propose new blends of clinical materials that will eventually improve the maturation of the cardiac progenitors once grafted onto the failing heart. In this perspective, we developed an in vitro model to investigate the combined influence of matrix elasticity and topographical confinement on stem cell differentiation into cardiomyocytes. By using microfabrication techniques to pattern pluripotent stem cells on substrates of controlled stiffness, we demonstrate that even using a widely recognized chemical-based protocol to modulate signaling cascades during differentiation, much heterogeneity emerges depending on the cellular physical environment. We thus extracted the main features that led to controlled and reproducible cardiac differentiation and applied it to the fabrication of next generation of multi-layered anisotropic cardiac patches in compliances with clinical requirements. This work opens new routes to high-scale production of cardiomyocytes and the fabrication of cell-laden or cell-free clinical patches.
287

Der Einfluss muriner mesenchymaler Stammzellen auf murine zytokin induzierte Killerzellen in der Kokultur

Bach, Martin 19 June 2014 (has links)
Stimulating lymphocytes with Ifn-γ, anti-CD3, and interleukin-2 promotes the proliferation of a cell population coexpressing T-lymphocyte surface antigens such as CD3, CD8a, and CD25 as well as natural killer cell markers such as NK1.1, CD49, and CD69. These cells, referred to as cytokine-induced killer cells (CIKs), display cytotoxic activity against tumour cells, even without prior antigen presentation, and offer a new cell-based approach to the treatment of malignant diseases. Because CIKs are limited in vivo, strategies to optimize in vitro culture yield are required. In the last 10 years, mesenchymal stem cells (MSCs) have gathered considerable attention. Aside from their uses in tissue engineering and as support in haematopoietic stem cell transplantations, MSCs show notable immunomodulatory characteristics, providing further possibilities for therapeutic applications. In this study, we investigated the influence of murine MSCs on proliferation, phenotype, vitality, and cytotoxicity of murine CIKs in a coculture system. We found that CIKs in coculture proliferated within 7 days, with an average growth factor of 18.84, whereas controls grew with an average factor of 3.7 in the same period. Furthermore, higher vitality was noted in cocultured CIKs than in controls. Cell phenotype was unaffected by coculture with MSCs and, notably, coculture did not impact cytotoxicity against the tumour cells analysed. The findings suggest that cell–cell contact is primarily responsible for these effects. Humoral interactions play only a minor role. Furthermore, no phenotypical MSCs were detected after coculture for 4 h, suggesting the occurrence of immune reactions between CIKs and MSCs. Further investigations with DiD-labelled MSCs revealed that the observed disappearance of MSCs appears not to be due to differentiation processes.:Inhaltsverzeichnis I Abbildungsverzeichnis III Tabellenverzeichnis IV Bibliographische Beschreibung V Abkürzungsverzeichnis VII 1 Einleitung 1 1.1 CIK-Zellen (CIK) 3 1.1.1 Merkmale von CIK-Zellen 3 1.1.2 Wirkungsmechanismen von CIK-Zellen 3 1.1.3 Studienlage 4 1.1.4 Bisherige Ansätze zur Verbesserung der Kultivierungsbedingungen 6 1.2 Mesenchymale Stammzellen (MSC) 7 1.2.1 Allgemein 7 1.2.2 Differenzierung von MSC 7 1.2.3 Heterogenität und Einflussfaktoren der MSC - Identitätsproblematik 8 1.2.4 Charakterisierung von MSC 9 1.2.5 Therapeutische Einsatzmöglichkeiten von MSC 11 2 Zielformulierung 15 3 Material und Methoden 16 3.1 Tiere 16 3.2 Materialien 17 3.2.1 Materialien für Zellkultur 17 3.2.2 Materialien für FACS-Analyse 18 3.2.3 Materialien für Zytotoxizitätsassay 19 3.2.4 Materialien für CFU-F-Assay 20 3.3 Methoden 21 3.3.1 Statistische Auswertung 21 3.3.2 Zellkultur 22 3.3.3 FACS (Fluorescence Activated Cell Sorting) 26 3.3.4 Markierung der MSC mit DiD 28 3.3.5 Zytotoxizitätsassay (LDH-Freisetzungsassay) 29 3.3.6 CFU-F-Assay 32 4 Ergebnisse 34 4.1 Beeinflussung der Wachstumskurve 34 4.1.1 Der Wachstumskurvenverlauf von CIK-Zellen (Kontrollen) 34 4.1.2 Der Wachstumskurvenverlauf von CIK-Zellen in der Kokultur mit MSC 35 4.1.3 Der Wachstumskurvenverlauf in MSC-konditioniertem Medium 37 4.1.4 Der Wachstumskurvenverlauf bei Restimulierung an Tag 14 38 4.2 Beeinflussung des Oberflächenphänotyps 40 4.2.1 Der Oberflächenphänotyp von CIK-Zellen 40 4.2.2 Vergleich Oberflächenphänotyp Kontrollen mit kokultivierten CIK 43 4.3 Beeinflussung der Vitalität 46 4.4 Beeinflussung der Zytotoxizität 48 4.5 Identifizierung der MSC 49 4.5.1 Adhärenz an Plastikoberflächen 50 4.5.2 Fibroblastenähnliche Wachstumsmorphologie 50 4.5.3 Wachstum in Colony-Forming-Units 51 4.5.4 Der Oberflächenphänotyp von MSC 53 4.6 Schicksal der MSC in der Kokultur 54 4.6.1 Der Oberflächenphänotyp der adhärenten Zellen nach Kokultur 54 4.6.2 Kokultur mit DiD gelabelten MSC 57 5 Diskussion 59 5.1 Beeinflussung der Wachstumskurve 60 5.1.1 Mechanismen der Beeinflussung des Wachstumskurvenverlaufs 60 5.1.2 Fehlerbetrachtung 68 5.2 Identifizierung der CIK sowie Beeinflussung von Phänotyp und Vitalität 69 5.3 Beeinflussung der Zytotoxizität 70 5.3.1 Vergleich Zytotoxizität Kontrollen mit Kokulturen 70 5.3.2 Fehlerbetrachtung 71 5.4 Identifizierung der MSC 72 6 Schlussfolgerung 75 7 Ausblick 77 8 Zusammenfassung 79 Literaturverzeichnis 83 Danksagung I
288

Léčba poranění míchy pomocí transplantace různých typů kmenových buněk / Treatment of spinal cord injury by transplantation different types of stem cells

Dubišová, Jana January 2015 (has links)
Spinal cord injury (SCI) is complicated injury with serious socioeconomic consequences for the patient and his whole family. Big difficulty cause also extremely high living expenses for the patient with this type of injury. That's why there is a need for therapeutic methods which would help patients after SCI to recover the lost functions and be able at least partially to return to their normal life. Different therapeutic methods are being used for SCI treatment. In this study we used four various types of stem cells: human bone marrow stem cells (hBM-MSCs), human umbilical cord mesenchymal stem cells (hUC-MSCs), neural precursors derived from induced pluripotent stem cells (iPS-NPs) and neural stem cell line derived from human fetal spinal cord tissue (SPC-01). These cells have been transplanted intrathecally or intraspinally 7 days after induction of the experimental model of SCI in the rat. We studied expressions of genes related to neurogenesis, growth factors and inflammation 10 and 28 days after SCI. Our analysis showed significant changes in gene expression 10 days after SCI. Significant up-regulation in expression of vascular endothelial growth factor (Vegf), ciliary neurotrophic factor (Cntf) and interferon regulatory factor 5 (Irf5) were found after transplantation of hBM-MSCs and hUC-...
289

Cell transplantation and gene therapy approaches for the treatment of retinal degenerative disorders

Eberle, Dominic 21 December 2012 (has links)
Photoreceptors are of prime importance for humans, since vision is one of the most important senses for us. In our daily life, where nearly every action is dependent on visual input, an impairment or a loss of eyesight leads to severe disability. With a non-syndromic prevalence of 1:4000, retinitis pigmentosa, a collective term for a group of inherited retinal eye diseases, represents, together with age-related macula degeneration, one of the main causes for visual impairment and blindness in industrialized countries. The dominant reason for vision loss is, in both cases, the irreversible loss of photoreceptor cells located in the outer nuclear layer of the retina. To date, no effective treatment is available to preserve or regain visual function in affected patients. Recent promising strategies for new retinal therapeutical approaches focus on one hand on the development of gene therapies, where an introduced wild-type allele compensates a mutated gene, and on the other hand on cell therapies, where stem or photoreceptor precursor cells (PPCs) are transplanted to the sub-retinal space to replace degenerated host photoreceptors. The current study is subdivided into three parts, addressing the issue of non-reversible photoreceptor cell loss due to retinal degenerative diseases by investigating in the first two parts new qualitative as well as quantitative approaches in the field of retinal cell therapy, while in the third part an ocular gene therapeutical approach targeting prominin-1, a gene involved in retinal degenerative disorders, was investigated. Briefly, this study shows in the first part, a significant enhancement of the integration rate of PPCs in wild-type host retinas, achieved by pre-transplantational sorting, using the recently discovered PPC - specific cell surface marker CD73. This sets another step further towards retinal cell therapy by increasing the effectiveness of such treatment. Next to this quantitative approach, it is also shown that the quality of transplanted photoreceptor precursor cells is comparable to native photoreceptors by demonstrating, that an indispensable prerequisite of every photoreceptor cell, the outer segment, is developed by transplanted PPCs after proper integration. Importantly, transplanted PPCs develop native outer segments even when not integrated in the host tissue but located in the sub-retinal space, as it is predominantly observed after transplantation into severely degenerated retinas. These results substantiate the feasibility of cell therapeutical treatment of severely degenerated retinas. At the end of this part, it is demonstrated, that outer segments are not formed properly by PPCs transplanted to the vitreal side of the retina. This suggests an influence of signaling molecules, presumably secreted by retinal pigment epithelial cells into the sub-retinal space, on transplanted PPC final differentiation. Since intensive research is done to differentiate stem cells into PPCs for cell therapeutical transplantation, these results may contribute significantly to this research by demonstrating, that factors secreted by the retinal pigment epithelium might play a crucial role for successful stem cell to PPC differentiation. The last part of my work investigates a gene therapeutical approach to cure inherited retinal degenerative diseases. One gene, where reported mutations cause retinal degeneration in humans is prominin-1, a protein expressed at cell membrane evaginations in a variety of cell types. Interestingly, the prominin-1 knock-out mouse is characterized exclusively by disorganized photoreceptor outer segment formation and progressive retinal degeneration. Successful delivery of a wild-type form of mouse prominin-1 using adeno-associated viral vector transfer, into the photoreceptors of prominin-1 - deficient mice is demonstrated. The divergent results show on one hand a rescue of the thickness of the photoreceptor outer nuclear layer on a short time period (3 weeks post treatment), and on the other hand long-term data (8-10 weeks post treatment) suggests histologically as well as functionally a negative effect on treated photoreceptors. This might be due to effects caused by an over-expression of prominin-1 and will be investigated in future studies. In conclusion, distinct and important investigations were made which contribute significant puzzle pieces to new cell- as well as gene therapeutical approaches for the treatment of retinal degenerative disorders.
290

Tolerogenní dendritické buňky jako nová buněčná terapie v diabetu I. typu / Tolerogenic dendritic cells as a novel cell-based therapy in type 1 diabetes

Kroulíková, Zuzana January 2019 (has links)
Utilization of tolerogenic dendritic cells (tolDCs) as a cell-based therapy represents a promising strategy in treatment of autoimmune diseases including type 1 diabetes (T1D). Numerous protocols have been established to generate tolDCs ex vivo and their therapeutic effect has been demonstrated in animal models of autoimmune diseases. In this thesis we compared three different variants of such protocols which are based on the combined treatment of bone marrow- derived DCs with vitamin D and dexamethasone applied at different time points of their maturation towards tolDCs. We assessed the efficiency of these protocols in regards of their effect on the expression of co-stimulatory molecules CD40, CD80, CD86, and MHC II and the chemokine receptor CCR7 on the surface of tolDCs. Then, we evaluated the migration pattern of antigen unloaded tolDCs in vivo as well as their effect on the induction of immune responses and cell proliferation of lymph node cells. This was achieved by labelling of tolDCs with membrane dye PKH26 and by following their migration path by flow cytometry after intraperitoneal (i.p) or subcutaneous (s.c.) injection into either left or right side of the body. On day 1, 3, 5, 7, and 9, the presence of PKH26+ tolDCs was examined in spleen, pancreatic, mesenteric, inguinal and axillary...

Page generated in 0.069 seconds