• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 50
  • 41
  • 13
  • 8
  • 8
  • 6
  • 4
  • 3
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 137
  • 137
  • 137
  • 44
  • 43
  • 37
  • 36
  • 29
  • 20
  • 20
  • 19
  • 19
  • 18
  • 18
  • 18
  • 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.
71

Ensaio clínico randomizado sobre o uso de enxerto autógeno de gordura suplementado com células estromais derivadas do tecido adiposo para a reconstrução de partes moles faciais em pacientes portadores de microssomia  craniofacial / Randomized controlled clinical trial of fat grafts supplemented with adipose-derived stromal cells for facial soft tissue reconstruction in patients with craniofacial microsomia

Tanikawa, Daniela Yukie Sakai 04 March 2013 (has links)
INTRODUÇÃO: Apesar dos primeiros relatos sobre o uso clínico de células estromais derivadas do tecido adiposo (ADSC) sugerirem que esta abordagem seja factível e efetiva na reconstrução de partes moles, não existem na literatura ensaios clínicos randomizados e com inclusão de controles. Assim sendo, foi objetivo deste estudo investigar se um novo protocolo para o isolamento de ADSC e seu uso em conjunto com os enxertos de gordura é de fato capaz de aumentar a sobrevivência destes enxertos. MÉTODOS: Pacientes com microssomia craniofacial (n=18) foram selecionados e randomicamente distribuídos para o grupo EC (com suplementação de ADSC) ou o grupo EE (sem suplementação de ADSC). Para o grupo EC, ADSC imediatamente isoladas a partir de metade do volume lipoaspirado foram utilizadas para o enriquecimento de células progenitoras do enxerto. A quantidade de tecido adiposo transplantado foi determinada pela simetria com o lado não afetado, não sendo realizado qualquer tipo de supercorreção. O número total de células viáveis isoladas antes e após a suplementação dos enxertos de gordura foi calculado, e através de citometria de fluxo estas células foram examinadas quanto à presença de marcadores de superfície para células mesenquimais. Tomografia computadorizada foi realizada para avaliar ambas as hemifaces no pré-operatório e no pós-operatório de seis meses. A espessura do revestimento de partes moles em quatro diferentes pontos de referência e o volume das hemifaces foram calculados. Morbidade geral e variáveis transoperatórias foram registradas. RESULTADOS: Para ambos os grupos o volume médio injetado foi de aproximadamente 28 mL. O número médio de células viáveis isoladas antes e após a suplementação dos enxertos foi 5,6 x 105 e 9,9 x 105 células por mL de tecido adiposo (p=0,015). Análise de citometria de fluxo revelou que ADSC foram positivas para marcadores de células mesenquimais (>95% para CD 73 e CD 105). No pós-operatório de seis meses, o índice de espessura média nos pontos de 1 a 4 foi de 0,91 (p=0,002), 1,02 (p=0,01), 1,05 (p=0,001), e 1,00 (p=0,04) no grupo EC, e 0,80 (p=0,58), 0,95 (p=0,18), 0,85 (p=0,009), e 0,84 (p=0,59) no grupo EE. Para o grupo EC o volume de gordura mantido após seis meses foi de 84 % e para o grupo EE foi de 51 % (p=0,003). Nenhuma complicação foi detectada. CONCLUSÕES: Estes resultados sugerem que o isolamento e a suplementação de ADSC é efetivo, seguro e superior à lipoenxertia convencional para o aumento de partes moles da face em pacientes portadores de microssomia craniofacial / INTRODUCTION: Although first reports of the clinical use of adipose-derived stromal cells (ADSC) suggest that this approach may be feasible and effective for soft tissue reconstruction, there is a lack of randomized, controlled clinical trials in the literature. Hence, this study aimed to investigate whether a novel protocol for isolation of ADSC and their use in combination with fat tissue could really improve the long-term retention of the grafts. METHODS: Patients with craniofacial microsomia (n=18) were selected and randomly assigned to group EC (with supplementation of ADSC) or group EE (without supplementation of ADSC). For group EC, ADSC freshly isolated from half of the aspirated fat sample were used to enrich for progenitor cells in the graft. Amount of transplanted adipose tissue was determined by trying to obtain symmetry with the unaffected side without any overcorrection. Number of viable cells isolated before and after the supplementation of the grafts was calculated, and these cells were examined for mesenchymal cell surface markers using flow cytometry. Computed tomography was performed to assess both hemifaces preoperatively and at six months postoperatively. Soft tissue thicknesses at four different reference points and the hemifaces volume were calculated. Overall morbidity and surgical variables were recorded. RESULTS: For both groups mean injected volume was 28 mL. Average number of viable cells isolated before and after supplementation of the grafts was 5,6 x 105 and 9,9 x 105 cells per mL of fat tissue (p=0,015). Flow cytometry analysis revealed that the ADSC were positive for mesenchymal cell markers (>95% for CD 73 and CD 105). At six months postoperatively, average thickness ratio at points 1 to 4 was 0,91 (p=0,002), 1,02 (p=0,01), 1,05 (p=0,001), and 1,00 (p=0,04) in group EC, and 0,80 (p=0,58), 0,95 (p=0,18), 0,85 (p=0,009), and 0,84 (p=0,59) in group EE. For group EC surviving fat volume at six months was 84 % and for group EE it was 51 % (p=0,003). No complications were detected. CONCLUSIONS: These results suggest that this strategy for isolation and supplementation of ADSC is effective, safe and superior to conventional lipoinjection for facial recontouring in patients with craniofacial microsomia
72

Devenir des propriétés immunomodulatrices des cellules souches mésenchymateuses de la gelée de Wharton au cours de la différenciation chondrocytaire / Become immunomodulatory properties of mesenchymal stem cells of Wharton's jelly during chondrocyte differentiation

Avercenc-Léger, Léonore 27 November 2017 (has links)
Ce travail a pour objet de déterminer les conditions optimales de production de substituts allogéniques capables de combler les lésions cartilagineuses dans le cadre du traitement de l’arthrose. Il s’oriente particulièrement sur la composante cellulaire de ces substituts. L’usage de cellules souches mésenchymateuses issues de cordons ombilicaux (CSM-GW) implique de déterminer quels facteurs obstétricaux, liés à l’environnement direct et indirect des CSM-GW, peuvent influencer leur prolifération ainsi que leur différenciation chondrocytaire. Dans une première partie de ce travail, trois types de facteurs ont été étudiés : les facteurs liés à l’enfant donneur, au déroulement de l’accouchement et de la délivrance, à la grossesse et à la mère. Nos données montrent que les CSM-GW ont des capacités prolifératives améliorées lorsque l’accouchement s’est déroulé à terme et sans complication, avec utilisation de Syntocinon® pendant le travail. Sur la base de ces résultats, nous avons utilisé les CSM-GW les plus efficaces dans le cadre de l’ingénierie du cartilage. Il a ensuite été essentiel d’élucider le profil d’action des CSM-GW dans un contexte allogénique. Le deuxième temps de ce travail a donc consisté à chercher le profil de stimulation le plus performant, au regard de la viabilité des cellules et de l’évolution de la sécrétion des facteurs solubles responsables des propriétés immunomodulatrices des CSM-GW au cours de la différenciation chondrocytaire. Nous avons alors mimé, in vitro et en biomatériaux d’Alginate/Acide hyaluronique (Alg/HA) une telle situation en stimulant les CSM-GW avec différentes doses d’IFN-γ et de TNF-α. Selon nos résultats, la stimulation par IFN-γ et TNF-α sur les CSM-GW en biomatériaux d’Alg/HA est plus efficace lorsque ces deux cytokines sont utilisées conjointement et n’est pas délétère pour la viabilité cellulaire aux concentrations respectives de 20 et 30 ng/mL. Cette double stimulation induit une augmentation de la sécrétion d’IL-6 et de PGE-2 par les CSM-GW, ne modifie pas leur sécrétion de TGF-β, et diminue la sécrétion de VEGF. Nous avons confirmé ces données lors d’une mise en situation fonctionnelle : des cocultures avec des cellules mononucléées de sang périphérique (PBMC) de donneurs sains nous ont permis d’évaluer la réponse des CSM-GW lors d’une situation allogénique. Ces mises en situations allogéniques ont été étudiées à différents temps afin d’évaluer les propriétés immunologiques des CSM-GW au cours du temps passé en biomatériaux. Nos résultats montrent que les CSM-GW peuvent exprimer des molécules HLA-G ainsi qu’IDO, mais ces expressions sont limitées en biomatériaux d’Alg//HA. Les CSM-GW en biomatériaux d’Alg/HA en situation allogénique ne sont pas immunogènes, quel que soit le temps de différenciation. En revanche, leurs capacités immunomodulatrices décroissent au cours du temps et sont plus fortes à J0 et J3 de la différenciation chondrocytaire, ce qui oriente vers une utilisation précoce de ces cellules. Les conclusions de ce travail permettent de (i) sélectionner les cordons idoines à l’ingénierie cellulaire et l’ingénierie du cartilage, (ii) définir les conditions permettant de mimer une situation allogénique in vitro, (iii) connaitre les propriétés immunomodulatrices des CSM-GW au cours de la culture en biomatériaux d’Alg/HA, y compris en situation allogénique / The purpose of this work is to determine the optimal conditions for allogeneic substitutes production, adapted to filling the cartilaginous lesions in osteoarthritis treatment. It focuses on the cellular component of these substitutes. The use of mesenchymal stem cells from umbilical cords (WJ-MSC) involves determining which factors, related to direct and indirect environment of the WJ-MSC, can influence their proliferation and chondrogenic differentiation. In a first part of our work, three types of factors were studied: related to the donor child, the course of labor and delivery, pregnancy and the mother. Our results show that WJ-MSC have enhanced proliferative capacities when coming from full-term birth and without complications, with the use of Syntocinon® during labor. On this basis, we used the most effective WJ-MSC for cartilage engineering. It was then essential to elucidate their action profile in allogeneic context. We stimulated WJ-MSC embedded in Alginate/Hyaluronic Acid (Alg/HA) scaffolds with different concentrations of IFN-γ and TNF-α in order to determine the most effective stimulation profile, with regard to viability of the cells and evolution of immunomodulatory soluble factors secretion. According to our results, the stimulation by IFN-γ and TNF-α on WJ-MSC in Alg/HA scaffolds is more effective when these two cytokines are used together and is not deleterious for cell viability at the concentrations of 20 and 30 ng/mL, respectively. This double stimulation induces an increase in the secretion of IL-6 and PGE-2 by the WJ-MSC, a decrease in the secretion of VEGF and does not modify the secretion of TGF-β. We confirmed these data during a functional study: cocultures with peripheral blood mononuclear cells (PBMC) from healthy donors allowed us to evaluate the response of WJ-MSC in an allogeneic situation. These allogeneic situations have been studied at different times to evaluate the immunological properties of WJ-MSC during the time of chondrogenic differentiation. Our results show that WJ-MSC can express HLA-G molecules as well as IDO, but these expressions are limited in Alg/HA biomaterials. Finally, the WJ-MSC in Alg/HA biomaterials in allogeneic conditions are not immunogenic, regardless of the time of differentiation. On the other hand, their immunomodulatory capacities decrease over time and are stronger at day 0 and day 3 of chondrogenic differentiation, which leads to an early use of these cells. Finally, this work allows us to (i) select the umbilical cords suitable for cellular and cartilage engineering, (ii) define the conditions mimicking in vitro an allogeneic situation, (iii) elucidate the immunomodulatory properties of WJ-MSC during Alg/HA biomaterials chondrogenic differentiation, including allogeneic situations
73

Análise das características biológicas das células estromais mesenquimais multipotentes obtidas de diferentes regiões anatômicas de pacientes com Pseudoartrose Congênita da Tíbia / Analysis of the biologic characteristics of multipotent mesenchymal stromal cells obtained from different anatomic regions of patients with Congenital Pseudoarthrosis of the Tibia

Romero, Jenny Manzano 09 November 2018 (has links)
A Pseudoartrose Congênita da Tíbia (PCT) é uma das doenças mais desafiantes da ortopedia pediátrica pela dificuldade em obter a união óssea e, quando esta ocorre, em mantê-la. É uma doença muito rara, difícil de tratar devido à sua falta de conhecimento sobre a patogênese. As Células estromais mesenquimais multipotentes (CMM) podem desempenhar um papel na patogênese do PCT, possivelmente devido à falha da diferenciação osteogênica. O estudo das CMM pode ajudar a compreender a patogênese da doença e desenvolver novas estratégias terapêuticas baseados no uso desta célula no futuro próximo. Frente ao exposto, este trabalho teve como objetivo a análise das características biológicas das CMM isoladas de diferentes regiões anatômicas de medula óssea de pacientes com PCT. Para isto, amostras de medula óssea foram coletas a partir de locais afetados e não afetadas pela doença: Crista ilíaca do membro não afetada (CINA), crista ilíaca do membro afetada (CIA), tíbia não afetada (TNA), e tíbia afetada (TA). O numero de pacientes incluídos no estudo foi três: PCT1, PCT2 e PCT3. Os resultados mostraram que todas as células isoladas de pacientes com PCT apresentavam características compatíveis com as CMM. A taxa de formação de unidades formadoras de colônias das células da TA tanto no PCT2 quanto no PCT3 foi significativamente menor em relação às células da TNA e CINA respectivamente (p<0.05). A quantidade de células positivas para o marcador CD146 foi menor nas células da TA do PCT1 e PCT2, A análise estatística mostrou que não há uma diferença significativa. Este marcador esta relacionado com a capacidade multipotente e formação óssea in vivo. No PCT1 observou-se que formação de matriz mineralizada das CMM isoladas da CIA foi significativamente maior em relação a TA. Além disso, as células da TA do PCT1 observou-se um uma secreção significativa de alguns citocinas envolvidas no processo de formação óssea, como CCL2, CCL3, CCL4, TNA-alfa, PDGF-BB, e GM-CSF. A alteração destas citocinas pode levar a situações complicadas como o caso de não consolidação óssea. Com os resultados obtidos, se há demonstrado que as CMM da tíbia afetada tenta formar osso, mas no local da lesão é insuficiente, por tal motivo é preciso realizar estudos focados no mecanismo molecular. / Congenital pseudoarthrosis of the tibia (CPT) is one of the most challenging orthopedic diseases because of the difficulty in obtaining bone union and, when it happens, in maintaining it. It is a rare disease, difficult-to-treat due to the lack of knowledge about to pathogenesis. Multipotent mesenchymal stromal cells (MSC) may play a role in the pathogenesis of PCT, possibly due to a failure in the osteogenic differentiation. Studying these cells can help to better understand the pathogenesis of the disease and develop new therapeutic strategies based on the use of MSC in the near future. In view of the above, this work had the objective of analyzing the biological characteristics of CMM isolated from different anatomic regions of bone marrow of patients with PCT. For this, bone marrow samples were collected from sites affected and unaffected by the disease: unaffected limb iliac crest (CINA), affected limb iliac crest (CIA), unaffected tibia (TNA), and affected tibia (TA). The number of patients included in the study was three: PCT1, PCT2 and PCT3. The results showed that all cells isolated from PCT patients had characteristics compatible with CMM. The rate of formation of colonyforming units of TA cells in both PCT2 and PCT3 was significantly lower in TNA and CINA cells respectively (p <0.05). The amount of cells positive for the CD146 marker was lower in the TA cells of PCT1 and PCT2. Statistical analysis showed no significant difference. This marker is related to the multipotent capacity and bone formation in vivo. In PCT1 it was observed that the formation of mineralized matrix of CMCs isolated from CIA was significantly higher in relation to AT. In addition, PCT1 TA cells showed a significant secretion of some cytokines involved in the bone formation process, such as CCL2, CCL3, CCL4, TNA-alpha, PDGF-BB, and GM-CSF. The alteration of these cytokines can lead to complicated situations such as the case of non-consolidation of bone. With the results obtained, if the CMM of the affected tibia has been shown to try to form bone, but at the site of the lesion is insufficient, it is necessary to carry out studies focused on the molecular mechanism.
74

Células estromais mesenquimais multipotentes promovem a metástase de melanoma pela ativação da transição epitélio-mesenquimal / Multipotent mesenchymal stromal cells promote melanoma metastasis through activation of the epithelial-to-mesenchymal transition

Souza, Lucas Eduardo Botelho de 11 June 2012 (has links)
A interação entre células tumorais e células estromais tem um papel central na progressão neoplásica. As células estromais mesenquimais multipotentes (MSCs) podem se integrar ao microambiente tumoral onde modulam o crescimento dos tumores por meio de distintos mecanismos. Entretanto, pouco se sabe sobre o papel das MSCs na metástase, a principal causa de morte em pacientes com câncer. Utilizando um modelo de melanoma murino ortotópico, nós demonstramos que MSCs obtidas da medula óssea de camundongos (MO-MSCs) ocupam o nicho perivascular nos tumores primários e aumentam 2,5 vezes a incidência de micrometástases pulmonares quando co-infundidas com células de melanoma B16. Observamos ainda que o meio condicionado das MO-MSCs não altera o potencial de colonização pulmonar das células B16 infundidas sistemicamente. Isto indica que as MO-MSCs modulam as fases iniciais da cascata metastática, durante a qual ocorrem os processos de invasão e intravasão nos vasos sangüíneos. Em correlação com estes efeitos pró-metastáticos, o secretoma das MO-MSCs induziu a transição epitélio-mesenquimal (EMT) nas células de melanoma in vitro. Após cultivo em meio condicionado das MO-MSCs, as células B16 adquiriram uma morfologia evidentemente fibroblástica. Ao mesmo tempo, houve o rearranjo dos filamentos de actina e o aumento da expressão de marcadores mesenquimais como fibronectina, vimentina, FSP1, N-caderina e ZEB2, acompanhado da repressão transcricional de E-caderina. A ativação da EMT pelo secretoma das MO-MSCs resultou na aquisição de propriedades metastáticas nas células de melanoma. Após cultivo em meio condicionado de MO-MSCs, as células B16 tiveram seu potencial de ancoragem à fibronectina reduzido, ao passo que houve o aumento na mobilidade e no potencial de invasão em matrizes tridimensionais. Utilizando inibidores competitivos de ATP contra o receptor tirosina-cinase Met, demonstramos que a aquisição de todas as propriedades metastáticas avaliadas e a ativação da EMT nas células de melanoma é mediada pela ativação da via HGF/Met. Estes dados destacam o papel das MOMSCs no microambiente tumoral como fonte perivascular de moléculas indutoras da EMT, cuja ativação leva a aquisição de traços metastáticos nas células de melanoma. Além disso, a inibição da via HGF/Met pode neutralizar os efeitos das MO-MSCs sobre as células tumorais, contribuindo para a repressão de propriedades fundamentais que sustentam a progressão e a disseminação neoplásica. Estas informações são importantes para o desenvolvimento seguro das MO-MSCs como ferramenta terapêutica e demonstram a importância da sinalização entre MSCs e células tumorais na disseminação metastática. Mais especificamente, estas observações reforçam a inibição da via HGF/Met como uma abordagem promissora para o tratamento da metástase. / The crosstalk between tumor cells and stromal cells can profoundly impact tumor progression. Multipotent mesenchymal stromal cells (MSCs) have been reported to integrate the tumor microenvironment where they are described to modulate tumor growth by distinct mechanisms. However, little is known about the impact of MSCs on metastasis, the main cause of death in patients with cancer. Using an orthotopic mouse melanoma model, we showed that mouse bone marrow-derived MSCs (BMMSCs) occupy the perivascular niche within primary tumors and increased by 2.5-fold the incidence of lung micrometastases after co-infusion with B16 melanoma cells. Also, MO-MSCs conditioned medium did not affect the lung colonization ability of systemically infused B16 cells. This indicates that MO-MSCs induces the initial steps of the metastatic cascade, during which the invasion and intravasion occurs. Correlating with these metastatic effects, the BM-MSCs\' secretome activated the epithelial-to-mesenchymal transition (EMT) in B16 cells in vitro. After culture in BMMSCs\' conditioned medium, B16 cells acquired an evident fibroblastic morphology. Simultaneously, we observed the rearrangement of actin filaments and the upregulation of mesenchymal markers such as fibronectin, vimentin, FSP1, Ncadherin and ZEB2. In agreement with the loss of epithelial phenotype, BM-MSCs\' secretome also suppressed E-cadherin expression in B16 cells. The activation of EMT by BM-MSCs leaded to the acquisition of metastatic traits in melanoma cells. After culture in BM-MSCs\' conditioned medium, B16 cells displayed reduced anchorage to fibronectin and increased motility and invasiveness in threedimensional matrix plugs. Inhibition of Met receptor with competitive ATP inhibitors demonstrated that the induction of EMT and the resultant acquisition of metastatic traits are driven by activation of HGF/Met signaling pathway. Taken together, these evidences highlight the role of BM-MSCs as a perivascular source of EMT-inductive signals, whose activation leads to acquisition of metastatic traits in melanoma cells. Furthermore, inhibition of HGF/Met signaling pathway can neutralize the effects of BM-MSCs on tumor cells, thereby allowing the repression of fundamental properties which support tumor progression and metastasis. This information is useful to safely develop BM-MSCs as therapeutic tool and demonstrate the relevance of the signaling between MSCs and tumor cells during metastasis. More specifically, it reinforces that inhibition of Met signaling can be a promissory approach for the treatment of metastasis.
75

Análise das características biológicas das células estromais mesenquimais multipotentes obtidas de diferentes regiões anatômicas de pacientes com Pseudoartrose Congênita da Tíbia / Analysis of the biologic characteristics of multipotent mesenchymal stromal cells obtained from different anatomic regions of patients with Congenital Pseudoarthrosis of the Tibia

Jenny Manzano Romero 09 November 2018 (has links)
A Pseudoartrose Congênita da Tíbia (PCT) é uma das doenças mais desafiantes da ortopedia pediátrica pela dificuldade em obter a união óssea e, quando esta ocorre, em mantê-la. É uma doença muito rara, difícil de tratar devido à sua falta de conhecimento sobre a patogênese. As Células estromais mesenquimais multipotentes (CMM) podem desempenhar um papel na patogênese do PCT, possivelmente devido à falha da diferenciação osteogênica. O estudo das CMM pode ajudar a compreender a patogênese da doença e desenvolver novas estratégias terapêuticas baseados no uso desta célula no futuro próximo. Frente ao exposto, este trabalho teve como objetivo a análise das características biológicas das CMM isoladas de diferentes regiões anatômicas de medula óssea de pacientes com PCT. Para isto, amostras de medula óssea foram coletas a partir de locais afetados e não afetadas pela doença: Crista ilíaca do membro não afetada (CINA), crista ilíaca do membro afetada (CIA), tíbia não afetada (TNA), e tíbia afetada (TA). O numero de pacientes incluídos no estudo foi três: PCT1, PCT2 e PCT3. Os resultados mostraram que todas as células isoladas de pacientes com PCT apresentavam características compatíveis com as CMM. A taxa de formação de unidades formadoras de colônias das células da TA tanto no PCT2 quanto no PCT3 foi significativamente menor em relação às células da TNA e CINA respectivamente (p<0.05). A quantidade de células positivas para o marcador CD146 foi menor nas células da TA do PCT1 e PCT2, A análise estatística mostrou que não há uma diferença significativa. Este marcador esta relacionado com a capacidade multipotente e formação óssea in vivo. No PCT1 observou-se que formação de matriz mineralizada das CMM isoladas da CIA foi significativamente maior em relação a TA. Além disso, as células da TA do PCT1 observou-se um uma secreção significativa de alguns citocinas envolvidas no processo de formação óssea, como CCL2, CCL3, CCL4, TNA-alfa, PDGF-BB, e GM-CSF. A alteração destas citocinas pode levar a situações complicadas como o caso de não consolidação óssea. Com os resultados obtidos, se há demonstrado que as CMM da tíbia afetada tenta formar osso, mas no local da lesão é insuficiente, por tal motivo é preciso realizar estudos focados no mecanismo molecular. / Congenital pseudoarthrosis of the tibia (CPT) is one of the most challenging orthopedic diseases because of the difficulty in obtaining bone union and, when it happens, in maintaining it. It is a rare disease, difficult-to-treat due to the lack of knowledge about to pathogenesis. Multipotent mesenchymal stromal cells (MSC) may play a role in the pathogenesis of PCT, possibly due to a failure in the osteogenic differentiation. Studying these cells can help to better understand the pathogenesis of the disease and develop new therapeutic strategies based on the use of MSC in the near future. In view of the above, this work had the objective of analyzing the biological characteristics of CMM isolated from different anatomic regions of bone marrow of patients with PCT. For this, bone marrow samples were collected from sites affected and unaffected by the disease: unaffected limb iliac crest (CINA), affected limb iliac crest (CIA), unaffected tibia (TNA), and affected tibia (TA). The number of patients included in the study was three: PCT1, PCT2 and PCT3. The results showed that all cells isolated from PCT patients had characteristics compatible with CMM. The rate of formation of colonyforming units of TA cells in both PCT2 and PCT3 was significantly lower in TNA and CINA cells respectively (p <0.05). The amount of cells positive for the CD146 marker was lower in the TA cells of PCT1 and PCT2. Statistical analysis showed no significant difference. This marker is related to the multipotent capacity and bone formation in vivo. In PCT1 it was observed that the formation of mineralized matrix of CMCs isolated from CIA was significantly higher in relation to AT. In addition, PCT1 TA cells showed a significant secretion of some cytokines involved in the bone formation process, such as CCL2, CCL3, CCL4, TNA-alpha, PDGF-BB, and GM-CSF. The alteration of these cytokines can lead to complicated situations such as the case of non-consolidation of bone. With the results obtained, if the CMM of the affected tibia has been shown to try to form bone, but at the site of the lesion is insufficient, it is necessary to carry out studies focused on the molecular mechanism.
76

Effets de l’hypoxie respiratoire sur les progéniteurs médullaires dans un modèle murin d’hypodynamie : intérêt pour la réparation osseuse / In Vivo Hypobaric Hypoxia, Hypodynamia and Bone Healing in Mice

Durand, Marjorie 18 December 2013 (has links)
La réparation osseuse est assurée par le recrutement constant de cellules souches/progéniteurs ostéo-compétents de nature hématopoïétique (CSH/PH), et mésenchymateuse (CSM). Une approche prometteuse pour le traitement des défauts osseux graves consisterait à favoriser le recrutement et la mobilisation des CSH/PH et des CSM à partir de la moelle osseuse vers le site de lésion. Plusieurs facteurs environnementaux sont connus pour moduler la prolifération, la mobilisation et la différenciation des progéniteurs ostéocompétents, dont l’hypoxie et l’hypodynamie (absence de contraintes mécaniques). Le but de ce travail de thèse a été d’investiguer in vivo l’impact de l’hypoxie respiratoire et de l’absence de contraintes mécaniques, appliquées séparément ou ensemble sur i) la mobilisation des progéniteurs ostéocompétents et sur ii) la réparation d’un défaut osseux cavitaire fémoral chez la souris. Sur un modèle murin dépourvu de défaut osseux, nos données montrent que l’hypoxie respiratoire est un agent mobilisateur des progéniteurs ostéocompétents, et qu’elle pourrait donc potentiellement exercer des effets bénéfiques sur la réparation osseuse. Toutefois, les effets de l’hypoxie sont modulés selon le statut hypodynamique ou non de l’animal. L’absence de contraintes mécaniques limite la mobilisation des progéniteurs érythrocytaires et mésenchymateux initiée par l’hypoxie, suggérant un effet potentiellement délétère de l’hypodynamie en condition hypoxique dans le contexte de la réparation osseuse. Chez les souris opérées, nous confirmons que l’hypoxie respiratoire déclenchée lors des phases de remodelage améliore la réparation du défaut osseux cavitaire. Une mobilisation des progéniteurs mésenchymateux et hématopoïétiques du fémur contra-latéral vers le fémur opéré est noté, mais le mode d’action de l’hypoxie passerait plutôt par une accélération du mécanisme de réparation dans la zone lésée. De façon intéressante, nous montrons que l’hypodynamie ne diminue pas le bénéfice apporté par l’hypoxie respiratoire à la réparation osseuse. En conclusion, ce travail de thèse identifie l’hypoxie respiratoire comme un candidat thérapeutique pertinent pour l’amélioration de la réparation osseuse. Bien que la perte des contraintes mécaniques module la biologie des cellules ostéoprogénitrices en absence de lésion, l’hypodynamie ne semble pas influencer la consolidation osseuse dans le cadre d’une amélioration de la réparation par un épisode hypoxique tardif. / Many environmental factors are known to influence bone cell fate, including proliferation, mobilization and differentiation of osteoprogenitor cells deriving from both hematopoietic and mesenchymal lineages. Among these factors, hypoxia and unloading (lack of mechanical loading / hypodynamia) are of particular interest. This study aims at investigating the impact of short-term hypobaric hypoxia and hindlimb unloading applied alone or in combination i) on the mobilization of osteocompetent progenitor cells on mice and ii) on the healing in a mouse model of surgical metaphyseal bone defect.In mice free of bone defect, our data indicate that respiratory hypobaric hypoxia acts as a mobilizing stimulus for osteoprogenitor cells. However, the effects of hypoxia in the bone marrow depend on whether mice are subjected to hindlimb unloading or not: hypodynamia tends to restrain the mobilization of both mesenchymal and erythroid progenitors under hypoxia. This suggests a potential detrimental influence of hypodynamia in the course of bone healing in hypoxic condition.In mice with surgery, we showed that hypobaric hypoxia during the remodelling process strongly enhances bone healing. A mobilization of both mesenchymal and hematopoietic progenitors is detected from the contralateral femur to the operated femur. In the lesion area, an acceleration of the repair process is evidenced. Interestingly, hindlimb unloading does not exert any negative influence on bone repair in our animal model. In conclusion, this study identifies delayed hypobaric hypoxia as a potent candidate to enhance bone healing. Even if unloading exerts significant effects on the biology of osteoprogenitor cells on mice free of bone defect, its influence is not detrimental for bone repair.
77

Desenvolvimento de metodologia para produção de soro AB humano para suplementação de meio de cultura destinado ao cultivo de células mesenquimais / Development of methodology for human AB serum production for culture medium supplementation for the cultivation of mesenchymal cells

Santos, Vanessa Tieko Marques dos 02 December 2015 (has links)
O crescente número de aplicações clínicas envolvendo células mesenquimais estromais multipotentes (CMMs) gera a necessidade da produção em larga escala destas células com adequada qualidade terapêutica. As CMMs são geradas atualmente através de culturas aderentes na presença de soro fetal bovino (SFB). Entretanto, apesar da eficiência na promoção do crescimento celular, o uso de SFB não é isento de desvantagens e riscos. Além do alto custo, sua utilização pode gerar risco de contaminação do produto final com agentes adventícios como vírus e príons. Por outro lado, a sua variabilidade em diferentes lotes e fornecedores dificulta a padronização do meio e reprodutibilidade do cultivo. Uma alternativa promissora para a cultura de células destinadas a terapia celular é a substituição de SFB por soro AB humano obtido a partir de plasma humano. Neste cenário, o objetivo deste trabalho foi produzir soro AB humano e avaliar a sua qualidade e eficácia como substituto do SFB na produção de células mesenquimais destinadas à terapia celular. Para a produção do soro AB humano foram avaliados tanto Plasma comum (PC>24h) (n=3) quanto Plasma isento de crioprecipitado (PCIC) (n=3). Após a produção, foi realizado o controle de qualidade dos lotes produzidos sendo verificado que os mesmos atenderam as exigências necessárias para a utilização na terapia celular. As duas fontes de plasma utilizadas para a produção do soro AB humano apresentaram características semelhantes quanto aos constituintes bioquímicos e demais parâmetros analíticos e foram eficazes na suplementação do cultivo e expansão das CMMs. A suplementação do meio com 10% de soro AB humano foi eficaz tanto no cultivo em culturas estáticas quanto em microcarregadores. Esta característica alinhase com a possibilidade de produção em larga escala, uma vez que, permitiu a expansão das CMMs de maneira similar à condição controle (suplementada com 10% de SFB) e preservou as características imunofenotípicas, funcionais e perfil citogenético pós cultivo. Além disso, o soro AB humano produzido pode ser utilizado por no mínimo doze meses após produção, quando armazenado em temperatura inferior a 20ºC negativos / The growing number of clinical applications involving multipotent mesenchymal stromal cells (MSCs) generates the need for large-scale production of these cells with appropriate treatment quality. The MSCs are now generated through adherent cultures in the presence of fetal bovine serum (FBS). However, despite the efficiency of this method, the use of FBS is not exempt of drawbacks and hazards. Besides the high cost, their use can lead to the risk of final product contamination with adventitious agents such as viruses and prions. In addition, due to its high variability of reproducibility, this methodology requires high level of standardization. A promising alternative would be to replace FBS by human AB serum from human plasma. In this scenario, our objective was to produce human AB serum and evaluate their quality and effectiveness as a FBS substitute on production of mesenchymal cells for therapy. For the production of human AB serum were evaluated both common plasma (CP > 24h; n=3) and plasma cryoprecipitate depleted (PCD; n=3). Quality controls were carried out to stablish minimal requirements of quality to be used in a cell therapy scenario. Both plasma sources of human AB serum presented similar biochemical characteristics and other analytical parameters, being effective in supplementation of the culture and expansion of MSCs. Supplementing the culture medium with 10% human AB serum was effective both in cultivation of static cultures as for microcarriers. This condition might indicate that this method would be useful for large scale production, allowing the expansion of MSCs similarly to the control condition and maintaining immunophenotypic, functional and cytogenetic characteristics. Furthermore, the human AB serum produced can be used for at least twelve months after production when stored at temperature below 20ºC negative
78

An investigation into the potential of mesenchymal stromal cells to attenuate graft-versus-host disease

Melinda Elise Christensen Unknown Date (has links)
Survival of patients with poor prognosis or relapsed haematopoietic malignancies can be markedly improved by allogeneic haematopoietic stem cell transplantation (HSCT). HSCT reconstitutes the immune and haematopoietic systems after myeloablative conditioning and inhibits the recurrence of the malignancy by a graft-versus-leukaemia (GVL) response mediated by donor T cells. However, significant post-transplant complications such as graft-versus-host disease (GVHD) continue to plague the event-free survival of this curative procedure. GVHD is facilitated by donor T cells that recognise histocompatibility antigens on host antigen presenting cells (APC), such as dendritic cells (DC). Current treatment options for GVHD are focused on these T cells. However, these treatments result in an increased incidence of infection, graft rejection and relapse. A novel means of immunosuppression in GVHD is the use of multi-potent, mesenchymal stromal cells (MSC). MSC are non-immunogenic cells that actively suppress T cell function in vitro, and can resolve steroid-refractory GVHD in the clinic. Despite their use in the clinic, there is a paucity of pre-clinical data. Our aim was to investigate the in vivo efficacy of MSC to control GVHD while maintaining the beneficial GVL effect, and to begin to understand the mechanism by which MSC exert their immunosuppressive effects. We isolated and characterised MSC from murine bone/bone marrow and demonstrated that they suppressed T cell proliferation in vitro, even at low ratios of 1 MSC per 100 T cells. This was true of both donor-derived MSC, and MSC derived from unrelated donors (third party). Importantly, we observed that MSC significantly reduced T cell production of the pro-inflammatory cytokines TNFα and IFNγ in culture supernatants and that IFNγ plays a key role in the ability of MSC to suppress T cell proliferation. In vivo, we examined the effects of donor-derived MSC on GVHD severity and onset in two myeloablative murine models of HSCT. A major histocompatibility complex (MHC)-mismatched donor-recipient pair combination was used as a proof–of-principle model [UBI-GFP/BL6 (H-2b)àBALB/c (H-2d)], and an MHC-matched, minor histocompatibility antigen (miHA) mismatched donor-recipient pair combination was used to mimic MHC-matched sibling transplantation [UBI-GFP/BL6 (H-2b)àBALB.B (H-2b)]. We examined a number of variables related to MSC infusion including timing, dose and route of injection. We found that early post transplant infusion of MSC by the intraperitoneal injection was most effective at delaying death from GVHD, compared to pre-transplant infusion or intravenous injection. Furthermore, we found that the dose of MSC was critical, as infusion of too few MSC was ineffective and infusion of too many MSC exacerbated the development of GVHD. Taken together, these results suggest that timing, dose and route of injection are all important factors to be considered to ensure successful therapeutic outcome. To investigate the in vivo mechanism of action, we conducted timed sacrifice experiments in the MHC-mismatched model to determine if MSC altered cytokine secretion and cellular effectors, such as DC, known to play a key role in GVHD. Despite the fact that MSC given post-HSCT enter an environment full of activated DC and IFNγ levels, by day 3 and 6 post infusion, these activated DC and IFNγ levels are decreased compared to controls or mice infused with MSC pre-transplant (p<0.05). This confirmed our in vitro data that IFNγ played an important role in MSC-mediated immunosuppression. In addition, when we removed a major source of IFNγ production in vivo by administering the T cell depleting antibody KT3 to mice with or without MSC, we found that although T cell depletion prolonged survival, MSC were unable to further enhance this effect. This was also true when MSC were used in combination with the conventional immunosuppressant cyclosporine. Finally, we examined whether the infusion of MSC would compromise the GVL effect. We found that whilst MSC could delay the onset of GVHD, in our model they did not alter the anti-tumour effects of the donor T cells. Overall, we have shown that MSC can delay but not prevent death from GVHD when administered at an appropriate time and dose and that IFNγ is required for MSC-mediated immunosuppression in our model. These data suggest that patients undergoing HSCT should be monitored for IFNγ, and administered MSC when high levels are reached. Whilst MSC may be a promising therapy for patients with severe GVHD, we highlight that further investigation is warranted before MSC are accepted for widespread use in the clinic. The risks and benefits for transplant recipients should be carefully considered before utilising MSC to treat or prevent GVHD.
79

Characterization and Modeling of the Remodeling Process that Occurs in Modular Tissue Engineered Constructs Assembled Within Microfluidic Perfusion Chambers

Khan, Omar 31 August 2011 (has links)
Using a modular approach, a vascularized tissue construct is created by embedding functional cells within submillimeter-sized collagen cylinders (modules) while the outside surfaces are seeded with endothelial cells (EC). The void spaces created by randomly packing modules into a container form EC-lined perfusion channels. Upon implantation, the tissues are remodeled by and integrated into the host and experience, to some degree, immune and inflammatory responses. This work utilized microfluidic techniques to study and model the tissue remodeling in vitro in the absence of the host response. When the construct’s tortuous perfusion channels were reproduced in poly(dimethylsiloxane) microfluidic devices and lined with EC, perfusion at higher flow rates reduced EC activation and maintained the desired quiescent EC phenotype. When applying these results to collagen constructs, higher flow rates were not achievable due to the weak mechanical properties of collagen. To increase the collagen’s mechanical strength, a semi-synthetic collagen/poloxamine-methacrylate hydrogel was examined but due to its heterogeneous surface composition, there was inadequate EC attachment and the material was deemed unsuitable for this application. Proceeding with lower flow rates, tissues assembled within microfluidic perfusion chambers from EC-seeded collagen modules showed that over the course of 24 hours, perfusion did not significantly increase activation but instead increased KLF2 expression, a transcription factor involved in the establishment of EC quiescence, and disrupted VE-cadherin bonds between adjacent EC. However, after 1 week of perfusion, the majority of EC were lost. To ameliorate this loss, mesenchymal stromal cells (MSC) were embedded within the modules in order to take advantage of their anti-apoptotic and immunomodulation effects. The MSC temporarily mitigated the loss of the EC but did not prevent it. They did, however, take on a phenotype similar to smooth muscle cells and migrated towards the EC. Perhaps this indicates that the combination of EC, MSC and perfusion drives the creation and assembly of pseudo vessels. Together, the microfluidic techniques used in this study to assemble and perfuse modular tissues revealed new insights into the remodeling process and exposed critical issues surrounding the adaptation of the EC to the combination of perfusion, remodeling and changing flow fields.
80

Characterization and Modeling of the Remodeling Process that Occurs in Modular Tissue Engineered Constructs Assembled Within Microfluidic Perfusion Chambers

Khan, Omar 31 August 2011 (has links)
Using a modular approach, a vascularized tissue construct is created by embedding functional cells within submillimeter-sized collagen cylinders (modules) while the outside surfaces are seeded with endothelial cells (EC). The void spaces created by randomly packing modules into a container form EC-lined perfusion channels. Upon implantation, the tissues are remodeled by and integrated into the host and experience, to some degree, immune and inflammatory responses. This work utilized microfluidic techniques to study and model the tissue remodeling in vitro in the absence of the host response. When the construct’s tortuous perfusion channels were reproduced in poly(dimethylsiloxane) microfluidic devices and lined with EC, perfusion at higher flow rates reduced EC activation and maintained the desired quiescent EC phenotype. When applying these results to collagen constructs, higher flow rates were not achievable due to the weak mechanical properties of collagen. To increase the collagen’s mechanical strength, a semi-synthetic collagen/poloxamine-methacrylate hydrogel was examined but due to its heterogeneous surface composition, there was inadequate EC attachment and the material was deemed unsuitable for this application. Proceeding with lower flow rates, tissues assembled within microfluidic perfusion chambers from EC-seeded collagen modules showed that over the course of 24 hours, perfusion did not significantly increase activation but instead increased KLF2 expression, a transcription factor involved in the establishment of EC quiescence, and disrupted VE-cadherin bonds between adjacent EC. However, after 1 week of perfusion, the majority of EC were lost. To ameliorate this loss, mesenchymal stromal cells (MSC) were embedded within the modules in order to take advantage of their anti-apoptotic and immunomodulation effects. The MSC temporarily mitigated the loss of the EC but did not prevent it. They did, however, take on a phenotype similar to smooth muscle cells and migrated towards the EC. Perhaps this indicates that the combination of EC, MSC and perfusion drives the creation and assembly of pseudo vessels. Together, the microfluidic techniques used in this study to assemble and perfuse modular tissues revealed new insights into the remodeling process and exposed critical issues surrounding the adaptation of the EC to the combination of perfusion, remodeling and changing flow fields.

Page generated in 0.1362 seconds