• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 101
  • 96
  • 47
  • 15
  • 7
  • 7
  • 5
  • 5
  • 4
  • 1
  • Tagged with
  • 311
  • 311
  • 139
  • 123
  • 92
  • 81
  • 62
  • 59
  • 57
  • 56
  • 55
  • 39
  • 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.
171

Thérapie cellulaire de la maladie de Parkinson : transplantation intranigrale vs intrastriatale / Cell therapy in Parkinson’s disease : intranigral versus intrastriatal transplantation

Droguerre, Marine 17 December 2015 (has links)
La maladie de Parkinson (MP) est une pathologie neurodégénérative associée principalement à une perte progressive de neurones dopaminergiques de la substance noire (SN) conduisant à une diminution de dopamine au niveau du striatum. Une des approches thérapeutiques expérimentales de la MP est la greffe de neurones dopaminergiques, non pas au niveau de la SN, mais directement dans la région cible, le striatum, et ceux avec des résultats variables. Dans cette étude, nous avons comparé en détail la récupération fonctionnelle suite à la transplantation de mésencéphale ventral (MV) fœtal provenant de souris exprimant la GFP sous le contrôle du promoteur de la tyrosine hydroxylase soit au niveau de la SN soit dans le striatum de souris adultes lésées unilatéralement à la 6-hydroxydopamine. Les conséquences anatomiques et fonctionnelles ont été analysées par des approches comportementales, électrophysiologiques et immunohistochimiques. Nos résultats montrent que les neurones greffés dans les deux emplacements envoient des projections vers le striatum. De plus, les deux types de greffes induisent une amélioration significative de la motricité ainsi que de l'activité des neurones du striatum. Toutefois, seule la greffe intranigrale a permis la restauration de la motricité fine des membres antérieurs et un retour à une excitabilité des neurones striataux à l’état basal. / Parkinson’s disease (PD) is a neurodegenerative disorder associated with a progressive loss of dopaminergic (DA) neurons in the substantia nigra (SN), leading to a loss of dopamine in the striatum. One of the experimental therapeutic approaches in PD is the graft of DA neurons not in their ontogenic site, the SN, but directly into the target region, the striatum and those leads to variable results. In this study, we have analyzed in detail the functional outcome of fetal VM tissue expressing GFP under the control of a tyrosine hydroxylase promoter grafts placed either into the SN or striatum in unilaterally 6-hydroxydopamine lesioned adult mice. Anatomical and functional outcome were analyzed using behavioral, electrophysiological and immunohistochemical approaches. Our results show that transplanted neurons in both locations can survive and re-innervate the striatum. Furthermore, both grafts locations significantly restored motor performance and induced the recovery of striatal firing properties. However, only intranigral transplantation allows recovery of fine motor skills of previous members and efficiently normalized cortico-striatal responses.
172

Estudo comparativo entre células estromais mesenquimais derivadas de pacientes com diabetes mellitus tipo 1 e de indivíduos saudáveis em relação ao potencial terapêutico no diabetes experimental / Comparative analysis of mesenchymal stromal cells derived from patients with type 1 diabetes mellitus and healthy individuals regarding the therapeutic potential in experimental diabetes

Yaochite, Juliana Navarro Ueda 23 May 2014 (has links)
O diabetes mellitus do tipo 1 (DM-1) é uma doença autoimune caracterizada pela destruição seletiva de células pancreáticas produtoras de insulina. O tratamento convencional é feito com insulina e existe atualmente a necessidade de desenvolver alternativas terapêuticas para o DM-1, como por exemplo o tratamento com células-tronco. As células estromais mesenquimais multipotentes (multipotent mesenchymal stromal cells-MSCs) representam uma fonte de células ideal para terapias celulares em virtude de seu fácil isolamento, expansão e capacidades imunomoduladora e regenerativa. Ainda não está esclarecido se MSCs isoladas de indivíduos com doenças autoimunes possuem alterações funcionais que poderiam limitar seu uso no contexto do transplante autólogo. Já foi descrito que MSCs de pacientes com esclerose múltipla, artrite reumatoide e lúpus eritematoso sistêmico possuem alterações fenotípicas e/ou funcionais. No entanto, pouco se sabe acerca das características das MSCs de pacientes com DM-1. Desse modo, o objetivo principal deste trabalho foi avaliar a eficácia da infusão de MSCs derivadas de pacientes com DM-1 no tratamento do diabetes experimental e comparar os resultados obtidos com o tratamento feito com MSCs isoladas de indivíduos saudáveis. Além disso, investigamos os efeitos da hiperglicemia in vitro sobre as MSCs, estabelecemos a melhor via de administração das MSCs em camundongos diabéticos e caracterizamos fenotipicamente e funcionalmente as MSCs de pacientes com DM-1. O diabetes experimental foi induzido em camundongos C57BL/6 por meio da administração de estreptozotocina. MSCs isoladas de tecido adiposo murino (ADMSCs) foram administradas em camundongos diabéticos pelas vias intravenosa, intraperitoneal, intrapancreática ou intraesplênica. MSCs foram isoladas da medula óssea de pacientes com DM-1 recém-diagnosticado (DM1-MSCs) e de indivíduos saudáveis (C-MSCs). A morfologia, tamanho celular, perfil imunofenotípico, diferenciação em adipócitos, migração e capacidade imunossupressora foram avaliados. 1x106 DM1-MSCs ou C-MSCs foram injetadas pela via intraesplênica em camundongos diabéticos 20 dias após indução do diabetes. A glicemia foi monitorada periodicamente. Após sacrifício dos camundongos, avaliamos a histologia do tecido pancreático, níveis de insulina circulante, a população de células T reguladoras no baço e linfonodos pancreáticos, o perfil de citocinas no soro e homogeneizado pancreático. A migração das ADMSCs Luc+ injetadas foi avaliada por meio de processamento de imagem baseado em bioluminescênia. Sete dias após cultivo com diferentes concentrações de glicose, as MSCs não apresentaram alterações nos parâmetros avaliados. A injeção de MSCs em camundongos diabéticos por meio da via intraesplênica foi a mais eficiente, promovendo reversão da hiperglicemia em cerca de 70-100% dos camundongos tratados. As DM1-MSCs apresentaram morfologia, tamanho celular, perfil imunofenotípico, diferenciação in vitro em adipócitos e capacidade imunossupressora in vitro semelhante às MSCs de indivíduos saudáveis. No entanto, as DM1-MSCs apresentaram maior migração in vitro em relação às C-MSCs. Não houve diferenças significantes do tratamento de camundongos diabéticos feito com DM1-MSCs ou C-MSCs, uma vez que ambas MSCs foram capazes de reverter a hiperglicemia, promover aumento da massa de células pancreáticas, bem como diminuir os níveis de IL-2 e IFN- no pâncreas dos camundongos tratados. Considerando-se que as MSCs de pacientes com DM-1 recém-diagnosticados não apresentaram alterações fenotípicas ou funcionais, elas poderiam ser transplantadas de forma autóloga nesses pacientes, representando uma nova alternativa terapêutica para o DM-1. / Type 1 diabetes mellitus (DM-1) is an autoimmune disease characterized by a selective destruction of insulin-producing pancreatic cells. The conventional treatment for DM-1 patients is the administration of insulin and new therapeutic approaches are needed, such as stem cell therapies. Mesenchymal stromal cells (MSCs) represent an important stem cell source for cell therapies because they are easy to isolate, present good capacity of expansion and they exhibit immunomodulatory and regenerative properties. However, it is not fully understood if MSCs from autoimmune patients are functionally defective or not, limiting their use in the autologous transplantation setting. There are some reports in the literature showing that MSCs from patients with multiple sclerosis, rheumatoid arthritis or systemic lupus erythematosus exhibit phenotypical and/or functional alterations. However, little is known about MSCs isolated from DM-1 patients (DM1-MSCs). Taking this into account, the aim of this work was to evaluate the efficacy of DM1-MSCs transplantation in diabetic mice and to compare this treatment with the treatment using MSCs isolated from healthy individuals. We also evaluated the influence of in vitro hyperglycemia on MSCs and we established the best route of MSCs administration in diabetic mice. The phenotypical and functional characteristics of DM1-MSCs were analyzed. The experimental diabetes model was induced in C57BL/6 male mice by the administration of streptozotocin. MSCs were isolated from mouse adipose tissue (ADMSCs) and injected in diabetic mice by intravenous, intraperitoneal, intrapancreatic or intrasplenic routes. DM1-MSCs were isolated from bone marrow aspirates of newly-diagnosed type 1 diabetes patients and C-MSCs were obtained from healthy individuals. The morphology, immunophenotypic profile, cell size, adipocyte differentiation (in vitro), migration (in vitro) and immunosuppressive capacity (in vitro) were evaluated. 1x106 DM1-MSCs or C-MSCs were injected by intrasplenic route in diabetic mice 20 days after diabetes induction. Glycemia was frequently monitored. The pancreatic tissue (histology and immunohistochemistry), serum insulin levels, the regulatory T cells population in spleen and pancreatic lymph nodes, the serum and pancreatic homogenate cytokine profiles were evaluated after MSCs administration. The homing of ADMSCs Luc+ was analyzed by bioluminescence based image processing. MSCs cultured for seven days with different glucose concentrations did not exhibit alterations in all evaluated parameters. The intravenous, intraperitoneal, intrapancreatic or intrasplenic routes of MSCs administration were tested. The intrasplenic route was the most efficient and promoted diabetes reversion in about 70-100% of MSCs-treated mice. No differences in morphology, cell size, immunophenotypic profile, adipocyte differentiation and immunosuppressive capacity were found for DM1-MSCs when compared with C-MSCs. However, the migration capacity of DM1-MSCs was higher than C-MSCs. The intrasplenic administration of DM1-MSCs or C-MSCs in diabetic mice similarly promoted a decrease in blood glucose levels, improved insulin-producing cell mass and modulated the production of IL-2 e IFN- in pancreatic tissue. Taking into account that MSCs from newly-diagnosed DM-1 patients did not show phenotypical and functional alterations, these cells could be isolated from diabetic patients representing a new therapeutic approach for DM-1 patients (autologous transplantation).
173

Reconstituição imunológica após transplante autólogo de células-tronco hematopoéticas em pacientes com diabetes mellitus tipo 1 e esclerose múltipla / Immune reconstitution after autologous hematopoietic stem cell transplantation in type 1 diabetes and multiple sclerosis patients.

Arruda, Lucas Coelho Marliére 16 August 2013 (has links)
Ensaios clinicos tem demonstrado que a imunossupressao em altas doses (IAD) seguida de transplante autologo de celulas tronco hematopoeticas (TACTH) e capaz de suprimir a atividade inflamatoria em pacientes com doencas autoimunes (DAIs) e induzir remissoes clinicas prolongadas nesses pacientes, porem os mecanismos de acao do TACTH ainda nao estao bem esclarecidos. O racional dessa terapia baseia-se na eliminacao das celulas autorreativas pela IAD e na reconstituicao de um sistema imunologico novo e tolerante apos o transplante a partir dos precursores hematopoeticos. O objetivo deste trabalho foi avaliar a reconstituicao imunologica em pacientes com diabetes mellitus tipo 1 (DM1, N=21) e pacientes com esclerose multipla (EM, N=37) sequencialmente apos o TACTH, e correlacionar os dados imunologicos com a resposta clinica dos pacientes ao transplante. Os pacientes com EM e DM1 foram divididos em dois grupos com base na resposta clinica apos o transplante: respondedores (EM-R; N=22) e nao-respondedores (EM-NR; N=15); livres de insulina por periodo maior ou igual a 3 anos (DM13 anos; N=11) e livres de insulina por periodo menor que 3 anos (DM1<3 anos; n=10); e acompanhados clinica e imunofenotipicamente por seis anos. Em relacao ao periodo pre-transplante, todos os grupos de pacientes com DM1 e EM apresentaram: 1, diminuicao do numero absoluto de celulas T CD3+ praticamente em todos os periodos pos-transplante avaliados, indicando uma intensa linfopenia decorrente da IAD; 2, aumento acentuado do numero de linfocitos T CD8+ e a diminuicao dos linfocitos T CD4+, resultando na inversao da razao CD4:CD8 durante todo o seguimento pos-transplante avaliado; 3, aumento significativo no primeiro ano apos o transplante das subpopulacoes de celulas T CD8+ de memoria central CD27+CD45RO+ e memoria efetora CD27- CD45RO+; 4, normalizacao dos numeros de linfocitos T CD4+ e CD8+ naive CD27+CD45RO- somente cinco anos apos o transplante, enquanto o numero de celulas T CD4+CD45RA+CD31+ recem- emigrantes do timo manteve-se abaixo dos valores pre-transplante durante todo o periodo avaliado, demonstrando que durante os seis anos de seguimento apos a IAD/TACTH predominaram mecanismos timo-independentes de reconstituicao imunologica; 5, normalizacao dos numeros de linfocitos B CD19+ entre dois a tres meses pos-transplante. O grupo de pacientes com DM1 que obteve melhor resposta clinica apos o tratamento com IAD/TACTH (DM13anos) apresentou, em comparacao ao periodo pre-transplante, numero diminuido de celulas T CD3+ (linfopenia) em varios periodos pos-transplante, numero aumentado de celulas T CD8+CD28- supressoras no primeiro ano pos-transplante principalmente, numero diminuido de celulas T CD4+ de memoria efetora nos periodos 2 a 9 meses pos-transplante e numero aumentado de celulas T reguladoras CD4+CD25hiFOXP3+ pos-transplante. O grupo de pacientes com EM com melhor resposta clinica apos o tratamento com IAD/TACTH (EM-R) apresentou, em comparacao ao periodo pre-transplante, numero diminuido de celulas T CD3+ (linfopenia) em varios periodos pos-transplante e numeros aumentados de celulas T reguladoras CD4+CD25hiFOXP3+ e CD8+CD28- supressoras nos primeiros tres anos pos-transplante. Vale ressaltar que os pacientes com DM1 e EM que apresentaram melhor resposta clinica permaneceram linfopenicos por maiores periodos de tempo apos o transplante. Desse modo, esse estudo revelou que a resposta terapeutica dos pacientes com DM1 e EM ao TACTH depende de uma linfopenia persistente, alem do aumento de celulas T reguladoras e supressoras e diminuicao de celulas T CD4+ de memoria apos o transplante. / Clinical trials have shown that high-dose immunosuppression (HDI) followed by autologous hematopoietic stem cell transplantation (AHSCT) is able to suppress the inflammatory activity in patients with autoimmune diseases (AID) and induce prolonged clinical remissions in these patients, but the mechanisms of action of AHSCT are still not well understood. The rationale of this therapy is based on the elimination of autoreactive cells by HDI and on the reconstitution of a new tolerant immune system after transplantation from hematopoietic precursors. The aim of this study was to evaluate the immune reconstitution in patients with type 1 diabetes mellitus (T1D, N=21) and patients with multiple sclerosis (MS, N=37) sequentially after the AHSCT, and correlate the immunological data with the clinical response of these patients to the transplant. Patients with MS and T1D were divided into two groups based on clinical response following transplantation: response (MS-R, N=22) and non-response (MS-NR, N=15); insulin-free for a period longer or equal to 3 years (T1D3 years, n=11) and insulin-free for less than 3 years (T1D<3 years, n=10); and accompanied clinical and immunophenotypically by six years. Regarding the pre-transplant period, all groups of patients with T1D and MS showed: 1, decreased absolute number of CD3+ T cells in virtually all post-transplant periods evaluated, indicating an intense lymphopenia resulting from HDI; 2, sharp increase in the number of CD8+ T lymphocytes and decreased CD4+ T lymphocytes, resulting in inversion of CD4:CD8 ratio throughout the follow-up post-transplant evaluation; 3, a significant increase, in the first year after transplantation, of CD8+ T central memory CD27+CD45RO+ and effector memory CD27-CD45O+ cell subpopulations; 4, normalization of CD4+ and CD8+ T naive CD27+CD45RO- lymphocytes numbers only five years after transplantation, whereas the number of CD4+CD45RA+CD31+ T cells newly emigrants from the thymus remained below the values pre- transplant during the study period, showing that during the six years of follow-up after the HDI/AHSCT mechanisms thymus-independent immune reconstitution were predominant; 5, normalization of CD19+ B lymphocytes numbers in two to three months post-transplant. The group of patients with DM1 that had the best clinical response after treatment with IAD/AHSCT (T1D 3years) showed, in comparison with the pre-transplant period, decreased number of CD3+ T cells (lymphopenia) at various times after transplantation, increased CD8+CD28- suppressor T cells numbers in the first year post-transplant, decreased number of CD4+ effector memory in periods of 2 and 9 months post-transplant and increased number of CD4+CD25hiFOXP3+ regulatory T cells after transplantation. The group of MS patients with better clinical response after treatment with IAD/AHSCT (MS-R) showed, in comparison to the pre-transplant period, decreased number of CD3+ T cells (lymphopenia) at various times after transplantation and increased numbers of CD4+CD25hiFOXP3+ regulatory Tcell and CD8+CD28- suppressor in the first three years post- transplant. It is noteworthy that patients with T1D and MS which showed better clinical response remained lymphopenic for longer periods of time after transplantation. Thus, this study revealed that the therapeutic response of patients with T1D and MS depend on a AHSCT to persistent lymphopenia, and increased regulatory and suppressor T cells and decreased number of CD4+ effector memory after transplantation.
174

Diferenciação de células-tronco em hepatócitos e desenvolvimento de modelo pré-clínico de fibrose hepática para ensaios de terapia celular / Mesenchymal stem cell differentiation in hepatocytes and development of pre-clinic model of hepatic fibrosis for cellular therapy assays

Oliveira, Érica Moreira de 09 December 2013 (has links)
Este trabalho teve como objetivo desenvolver um protocolo para a diferenciação in vitro de células-tronco mesenquimais (CTM) em hepatócitos e a padronização de um modelo animal de fibrose hepática induzida por dimetilnitrosamina (DMN) para ensaios pré-clínicos de transplante de CTM. CTM isoladas de fontes variadas apresentaram morfologia fibroblastóide e aderência ao plástico e o padrão de marcadores de superfície celular esperado na análise por citometria de fluxo. A capacidade de diferenciação osteogênica e adipogênica dessas células foi comprovada pelas colorações de vermelho de alizarina, oil red e azul de toluidina, respectivamente, confirmando, que as células isoladas para este estudo se comportaram como CTM conforme proposto pela Sociedade Internacional de Pesquisa em Células-tronco. A diferenciação hepática foi avaliada quanto à morfologia e capacidade das células diferenciadas de estocar glicogênio confirmada por PAS (ácido periódico-Schiff), de sintetizar albumina confirmada por imunofluorescência, além da capacidade de expressar genes hepato-específicos verificada por ensaios de PCR em tempo real. Com base na literatura para diferenciação hepática, diferentes protocolos de um, dois e três passos foram testados. CTM humanas mostraram capacidade de produzir e estocar glicogênio e de sintetizar albumina, apenas quando diferenciadas com protocolos de três etapas, porém sem uma expressão aumentada dos genes hepato-específicos albumina, &#945;-fetoproteína e c-Met. Uma etapa de diferenciação endodérmica, previamente aplicada à diferenciação hepática, aumentou a capacidade de produzir e estocar glicogênio das CTM diferenciadas. Para a padronização do modelo de fibrose hepática induzida por DMN, foram realizados experimentos de dose-resposta e foi verificado o efeito da hepatectomia em modelos mistos DMN/hepatectomia. A injúria hepática e o efeito do transplante de CTM foram avaliados por análise macroscópica dos fígados, histologia das biópsias de fígados corados com HE e tricromo de Masson e parâmetros bioquímicos séricos. Alterações macroscópicas, histológicas e nos níveis séricos de fosfatase alcalina indicam a indução da fibrose hepática nos ratos Wistar tratados com DMN na dose de 10 &#181;g/g de peso animal por três dias consecutivos durante quatro semanas, mas não observamos nenhum efeito induzido pela hepatectomia. Porém, este modelo com DMN se mostra semelhante a estágios iniciais de uma fibrose hepática. O transplante de 1 x 107 CTM de veia de cordão umbilical humano (VCUH) no modelo de injúria hepática induzida por DMN não resultou em melhora da fibrose, diminuição dos níveis séricos de fosfatase alcalina e nem em ganho de peso dos animais quando comparados aos animais tratados com PBSA após a injúria hepática (grupo placebo). Em conjunto, esses resultados sugerem que CTM humanas se diferenciam após tratamentos mais complexos, onde os indutores hepatogênicos são sequencialmente adicionados ao meio de modo a mimetizar a sinalização durante o desenvolvimento embrionário. O transplante de CTM de VCUH parece não ter efeito positivo em um modelo pré-clínico de injúria hepática similar a estágios iniciais de fibrose. Financiado por CNPq (573578/2008-7) e FAPESP (2007/54260-2). / This study aimed to develop an in vitro differentiation protocol of mesenchymal (MSC) stem cells to hepatocytes and to standardize an animal model for hepatic fibrosis induced by dimethylnitrosamine (DMN) for preclinical transplant assays of MSC. MSC isolated from various sources presented fibroblastoid morphology, plastic adherence, and the expected pattern of cell surface markers by flow cytometry analysis. The capacity of osteogenic, adipogenic and chondrogenic differentiation of these cells was confirmed by alizarin red, oil red and toluidine blue staining, respectively, confirming that the cells isolated for this study behave as MSC, as proposed by the International Society for Stem Cell Research. Hepatogenic differentiation was evaluated by analysis of cell morphology, capacity to store glycogen confirmed by PAS (periodic acid-Schiff), albumin synthesis confirmed by immunofluorescence, as well as hepatic-specific gene expression verified by real time PCR assays. Based on the published literature on hepatic differentiation, several protocols of one, two, and three steps were tested. Human MSC differentiated solely when treated in a three step-protocol, showing the ability to produce and store glycogen and synthesize albumin; however the expression of hepatic-specific genes such as albumin, &#945;-fetoprotein and c-Met was not increased. An endoderm differentiation stage, added to the hepatic differentiation protocol, increased the capacity to produce and store glycogen of differentiated MSC. In order to standardize the model of liver fibrosis induced by DMN, dose-response experiments were performed and the effect of hepatectomy in mixed models DMN/hepatectomy was observed. Severity of liver injury and the effect of cell transplantation were evaluated by macroscopic analysis of the livers, histology of liver biopsies stained with HE and Masson\'s trichrome, and evaluation of serum biochemical parameters. The macroscopic and histological observations, and altered alkaline phosphatase serum levels indicated the success in inducing liver fibrosis in DMN-treated rats at a dose of 10 &#181;g/g of animal weight for three consecutive days, during four weeks, without any additional effect upon hepatectomy. Transplanting 1 x 107 umbilical cord MSC in the model of liver injury induced by DMN did not result in improvement of the fibrosis, decrease of alkaline phosphatase serum levels, or in weight gain of the treated animals compared to animals treated with PBSA after liver injury (placebo group). Together, these results suggest that human MSC are capable of differentiating to hepatocyte-like cells after more complex protocols, where hepatogenic inducers are sequentially added to the medium in order to mimic signaling that occurs during fetal development. Transplantation of undifferentiated umbilical cord MSC did not have any positive effect in a preclinical liver injury model characterized by an early stage of fibrosis. Supported by CNPq (573578/2008-7) and FAPESP (2007/54260-2).
175

Avaliação de células-tronco mesenquimais do cordão umbilical humano em lesão de órgãos e disfunção endotelial na sepse / Evaluation of human umbilical cord mesenchymal stem cells in organ damage and endothelial dysfunction in sepsis

Cóndor Capcha, José Manuel 23 June 2015 (has links)
A sepse é uma doença relacionada como a presença de infeção junto a uma resposta inflamatória sistêmica; sua fisiopatologia envolve uma rede complexa de citocinas e mediadores inflamatórios que causam a injúria de diversos tecidos. Na atualidade são muitas as tentativas para diminuir a mortalidade, porém até agora, não existe uma estratégia específica para tratar a doença. As células-tronco mesenquimais da geleia de Wharton do cordão umbilical (CTM-GW) são conhecidas por expressar genes e fatores envolvidos na angiogênese e imunomodulação. Nós usamos o modelo de ligadura e punção do ceco (LPC) para analisar o papel da CTM-GW em disfunção orgânica relacionada à sepse. Foi utilizada a citometria de fluxo para avaliar o fenótipo das células isoladas. Dividimos ratos Wistar em grupos: sham (operação simulada); LPC; e LPC + CTM (106 CTM-GW i.p., 6 horas após LPC). Às 24 h pós-LPC, foram avaliadas a função renal, hepática e outras variáveis do estudo. As CTM-GW foram negativas para CD3, CD34, CD45 e HLA-DR, enquanto eles foram positivos para CD73, CD90 e CD105. O tratamento com CTM na sepse reduziu a mortalidade, melhorou a filtração glomerular (aferido pelo clearance de inulina), função tubular, reduziu a lesão hepática e demostrou uma ação anti-inflamatória. O tratamento também apresentou um efeito anti-apoptótico e protetor do tecido renal e do endotélio, mediante a regulação da expressão de VEGF, AQP2 e eNOS. Em conclusão as CTM-GW diminuem a injúria renal e hepática, portanto, pode desempenhar um papel protetor na sepse / Sepsis is a disease related to the presence of infection with a systemic inflammatory response. The pathophysiology involves complex cytokine and inflammatory mediator networks that cause injury to various tissues. Currently, there are many attempts to reduce mortality, but so far, there is no specific strategy for treating the disease. Human umbilical cord Wharton\'s jelly-derived mesenchymal stem cells (hWJ-MSCs) are known to express genes and factors involved in angiogenesis and immunomodulation. We used a cecal ligation and puncture (CLP) model to analyze the role of hWJ-MSCs in sepsis-related organ dysfunction. We used flow cytometry to evaluate hWJ-MSC phenotypes. We divided Wistar rats into groups: sham (sham-operated); CLP; and CLP+MSC (106 WJ-MSCs, i.p., 6 h after CLP). At 24 h post-CLP, we evaluated renal function, liver and other variables. hWJ-MSCs were negative for CD3, CD34, CD45 and HLA-DR, whereas they were positive for CD73, CD90 and CD105. In sepsis, treatment with MSC reduced mortality, improved glomerular filtration rate (measured by inulin clearance), tubular function, reduced liver damage and decreased the inflammatory markers. The treatment also showed an anti-apoptotic effect and protected the renal tissue and endothelium by up-regulation the expression of VEGF, AQP2 and eNOS. In conclusion, hWJ-MSCs decrease renal and hepatic injury, therefore, may play a protective role in sepsis
176

Efeitos da terapia celular com a associação de células-tronco mesenquimais e osteoblastos no reparo do tecido ósseo / Effects of cell therapy with association of mesenchymal stem cells and osteoblasts in bone tissue repair

Santos, Thiago de Santana 27 June 2014 (has links)
A regeneração de defeitos ósseos continua sendo um grande desafio na área de Odontologia e Medicina. É bem estabelecido que células-tronco mesenquimais (CTMs) e osteoblastos (OBs) desempenham um papel crítico na osteogênese, tornando-se candidatos a utilização em procedimentos de terapia celular que visam otimizar o processo de reparação óssea. Porém, pouco se sabe sobre a interação entre CTMs e OBs, e a maioria dos estudos enfatiza o efeito dos OBs sobre CTMs, fazendo com que a influência das CTMs na atividade osteogênica dos OBs continue sendo uma questão desafiadora. Baseados em nossos estudos anteriores, formulamos a hipótese de que a terapia celular que fizesse uso de uma associação de CTMs e OBs poderia ser mais eficaz para o reparo do tecido ósseo do que essas células isoladamente, principalmente como resultado da estimulação de OBs por CTMs. Para tal, foi realizado estudo in vitro para avaliar os efeitos das CTMs sobre os OBs e in vivo para avaliar os efeitos dessas células, isoladamente e combinadas, sobre a reparação óssea. CTMs da medula óssea de rato foram cultivadas em meio de crescimento para manterem-se como CTMs ou em meio osteogênico para diferenciarem-se em OBs. Após alcançar a subconfluência, as células foram cultivadas in vitro em três diferentes condições: (1) co-cultura direta de CTMs e OBs usando três proporções celulares (1:1, 1:2 e 2:1), (2) co-cultura indireta de CTMs e OBs usando insertos e (3) OBs cultivados em meio condicionado por CTMs. Para avaliação das respostas celulares foram realizados ensaios de proliferação celular, atividade de fosfatase alcalina (ALP), formação de matriz mineralizada, expressão gênica de marcadores osteoblásticos, imunolocalização de sialoproteína óssea (BSP) e osteopontina (OPN) e migração celular. Para os experimentos in vivo, as células foram carreadas em esponja de colágeno através de vários ciclos de centrifugação. Após, defeitos ósseos em calvária de rato foram preenchidos com (1) esponja de colágeno sem células, (2) esponja de colágeno com CTMs, (3) esponja de colágeno com OBs e (4) esponja de colágeno com associação de CTMs e OBs. Para avaliação da reparação óssea in vivo após 4 semanas, foram realizadas análises histomorfométricas através de cortes histológicos e microtomografia computadorizada. Os dados foram comparados pelo teste de Kruskal-Wallis e, se necessário pelo teste de Mann-Whitney (p&le;0,05). Foi observado que CTMs têm efeito repressivo sobre a proliferação e as expressões fenotípicas e genotípicas de OBs (P&le;0,05). Em relação ao reparo dos defeitos ósseos, somente naqueles tratados com células observou-se formação óssea predominantemente como ilhotas isoladas e diferenças, principalmente qualitativas, entre os tipos celulares utilizados, com tendência de maior formação óssea em defeitos tratados com OBs em comparação ao uso de CTMs. Com base nos resultados obtidos, pôde-se concluir que as CTMs apresentam efeito inibitório sobre OBs e que a terapia celular com OBs parece ser mais eficaz no reparo do tecido ósseo. / The regeneration of bone defects remains a major challenge in the field of Dentistry and Medicine. It is well known that mesenchymal stem cells (MSCs) and osteoblasts (OBs) play critical roles in osteogenesis, making them promising alternatives to be employed in cell therapy procedures to enhance the process of bone regeneration. Studies about the crosstalk between MSCs and OBs are mainly focused on the effect of OBs on MSCs, thus how MSCs may affect OBs phenotype expression remains a challenging question. Based on our previous studies, we have hypothesized that cell therapy using a combination of MSCs and OBs could be more effective for the bone repair than these cells separately, mainly due to the stimulation of OBs by MSCs. For this, we carried out in vitro experiments to evaluate the effects of MSCs on the OBs and in vivo experiments to assess the effects of these cells either isolated or combined on bone repair. Rat bone marrow MSCs were cultured either in growth medium to keep MSCs features or in osteogenic medium to differentiate into OBs. After reaching subconfluence, cells were grown in vitro in three different conditions: (1) direct coculture of MSCs and OBs using three cell proportions (1:1, 1:2 and 2:1), (2) indirect coculture of MSCs and OBs using transwell porous filters and (3) OBs cultured in MSCs conditioned medium. Cell responses were evaluated by assaying cell proliferation, alkaline phosphatase activity (ALP), mineralized matrix formation, gene expression of osteoblast markers, immunolocalization of bone sialoprotein (BSP) and osteopontin (OPN), and cell migration. For in vivo experiments, cells were seeded into collagen sponge by a centrifugation method. After, calvarial defects were implanted with (1) collagen sponge without cells, (2) collagen sponge with MSCs, (3) collagen sponge with OBs, and (4) collagen sponge and association of MSCs with OBs. To evaluate bone repair at the end of 4 weeks, histomorphometric analyzes were carried out using histological slides and micro-computed tomography. Data were compared by Kruskal-Wallis test and, if appropriated, by Mann-Whtiney test (p&le;0.05). It was observed that MSCs repressed proliferation, phenotypic and genotypic expressions of OBs (P&le;0.05). Bone formation was observed only in cell treated defects as isolated islets and qualitative differences were noticed among cell types, with a tendency of more bone formation in OBs treated defects compared with MSCs ones. Based on these results, we can conclude that MSCs exhibited inhibitory effect on OBs and that cell therapy with OBs seemed to be more effective for bone repair.
177

Étude des propriétés physiques et mécaniques de microsphères d'alginate au cours d'un cycle de congélation-décongélation et application pour la cryoconservation de cellules souches mésenchymateuses encapsulées / Study of physical and mechanical properties of alginate microspheres during a freeze-thaw cycle and its application for the cryopreservation of encapsulated mesenchymal stem cells

Hayer, Benoît d' 22 May 2018 (has links)
La thérapie cellulaire et les médicaments de thérapie innovante sont des solutions prometteuses pour la régénération des tissus ou organes présentant des défauts fonctionnels ou organiques. Avant le stade de l'insuffisance cardiaque terminale (stade IV NYHA) suite à un infarctus du myocarde, l'implantation d'un patch de fibrine cellularisé avec des progéniteurs myocardiques sur le site de nécrose de l'infarctus, est l'une des perspectives qui permettrait de régénérer un muscle cardiaque fonctionnel et apparait comme étant une alternative nouvelle avec notamment un essai clinique de phase I en cours (ESCORT : NCT02057900). Cependant, cette thérapie innovante présente de réelles contraintes, parmi lesquelles, un protocole nécessitant, i) une utilisation pour la production de cellules progénitrices myocardiques CD15+, de DMSO, de sérum foetal bovin, de trypsine porcine, de fibroblastes murins pouvant être la source d'une contamination chimique ou microbiologique, ii) une caractérisation importante des cellules produites, pour déterminer leur viabilité, leur pureté, leur état de différenciation, iii) d'implanter le patch de fibrine cellularisé dans un délai limité avant l'obtention des résultats de stérilité et d'endotoxines, iv) d'inciser le péricarde et de former une poche, geste chirurgical très invasif, afin d'implanter le patch cellularisé. Avec l'objectif de limiter ces contraintes et de renforcer la sécurisation pharmaceutique de ce médicament de thérapie innovante, les différents axes de ce travail ont porté sur i) l'ajout, juste avant l'implantation, d'une étape de cryoconservation des cellules dans un milieu sans sérum et sans DMSO, mais avec des agents cryoprotectants de qualité pharmaceutique. L'avantage apporté par la cryoconservation étant de rendre possible une production par lot, et la réalisation des contrôles sans contrainte de temps avant l'implantation, ii) la vectorisation des cellules par une encapsulation dans des microsphères formant une suspension injectable et permettant une implantation directement au travers du péricarde et immédiatement après la décongélation, iii) l'utilisation de polymères bioadhésifs afin de maintenir les microsphères au site d'implantation. Dans un premier temps, ce travail a permis d'identifier l'alginate de sodium de faible viscosité à 1,2% comme polymère pour réaliser l'encapsulation à l'aide d'une buse vibrante de 120 µm de diamètre. La nature et la concentration d'agents cryoprotectants ont également été définies. Les agents cryoprotectants ont été sélectionnés parmi les oses (glucose, saccharose, tréhalose), les polyols (glycérol, mannitol, sorbitol) et l'urée, à une concentration permettant d'atteindre une osmolarité totale de 500 mOsm/L pour abaisser le point de congélation de l'eau. Enfin le chitosane de faible viscosité à 0,5% a été utilisé comme polymère bioadhésif de surface pour maintenir les propriétés mécaniques et la forme des microsphères après la congélation. Dans un second temps, une évaluation biologique a permis de mesurer l'impact des étapes du procédé d'encapsulation et de cryoconservation, sur des cellules souches mésenchymateuses humaines utilisées comme modèle. Il a ainsi été possible d'optimiser le protocole ce qui a eu pour effet d'augmenter la viabilité, évaluée après encapsulation et congélation par une analyse en cytométrie de flux avec le 7AAD, de moins de 5% à environ 35%. / Cell therapy and advanced therapy medicinal products are promising solutions for the regeneration of tissues or organs with functional or organic defects. Before the terminal heart failure stage (stage IV NYHA) following a myocardial infarction, the implantation of a cellularized fibrin patch with myocardial progenitors at the location of the infarct necrosis, is one of the perspectives that would allow a functional heart muscle to regenerate and appears to be a new alternative, in particular, with an ongoing Phase I clinical trial (ESCORT : NCT02057900). However, this innovative therapy presents real constraints, among which, a protocol requiring, i) the use for the production of CD15+ myocardial progenitor cells of, DMSO, bovine fetal serum, porcine trypsin, and murine fibroblasts which may be the source of chemical or microbiological contamination, ii) an important characterization of the produced cells, to determine their viability, purity, and state of differentiation, iii) to implant the cellularized fibrin patch within a limited time frame before getting the results of sterility and endotoxins, iv) to incise the pericardium and to form a pouch, a very invasive surgical gesture, in order to implant the cellularized patch inside. With the objective of limiting these constraints and strengthening the pharmaceutical safety of this innovative therapy medication, different axes of this work have focused on i) the addition, just before the implantation, of a step of cryopreservation of the cells in a medium without serum and without DMSO, but with pharmaceutical-grade cryoprotectants. The advantages of cryopreservation is to allow production in batches, and controls to be carried out without time constraints before the implantation, ii) the vectorization of the cells by encapsulation in microspheres forming an injectable suspension and allowing direct implantation through the pericardium immediately after thawing, iii) the use of bioadhesive polymers to maintain the microspheres at the location of the implantation. This study initially enabled to identify a low-viscosity sodium alginate at 1.2% as a polymer being used for the encapsulation with the use of a vibrating nozzle which diameter is of 120 µm. The nature and the concentration of the cryoprotectants have also been defined. The cryoprotectants were selected from oses (glucose, sucrose, trehalose), polyols (glycerol, mannitol, sorbitol) and urea, at a concentration which achieves a total osmolarity of 500 mOsm/L in order to lower the freezing point of water. Finally, low viscosity chitosan at 0.5% was used as a bioadhesive polymer at the surface of the microspheres to maintain their shapes and mechanical properties after freezing. In a second step, a biological evaluation allowed to measure the impact of the encapsulation and the cryopreservation processes, on human mesenchymal stem cells used as a model. It was thus possible to optimize the protocol, which in return increased the viability ; evaluation made after encapsulation and freezing by a flow cytometry analysis with 7AAD ; from less than 5% to about 35%.
178

Human cytomegalovirus-specific regulatory and effctor T cells are clonally identical

Schwele, Sandra 28 September 2009 (has links)
Die Mehrzahl der im Thymus generierten CD4+CD25high regulatorischen T-Zellen (Treg) besitzt hohe Affinität gegenüber körpereigenen Antigenen. Es ist bekannt, dass T-Zell Rezeptoren (TCR) auf Treg Zellen in der Peripherie zusätzlich auch fremde Antigene verschiedener Pathogene wie Parasiten, Bakterien und Viren erkennen. Wenig ist bekannt über das klonale T-Zell Rezeptor Repertoire dieser Treg Populationen und ihre Beziehung zu CD4+CD25low effektor T-Zellen (Teff) im Menschen. In dieser Studie analysieren wir humane TCR auf expandierten Treg and Teff Zellen mit definierter Antigen Spezifität für Haupthistokompatibilitätskomplex (MHC) Klasse II restringierte „fremde“ Epitope des Cytomegalovirus (CMV). Bemerkenswerterweise fanden wir, dass der gleiche TCR Vb-CDR3 Klon in beiden funktionell unterschiedlichen Subpopulationen in vitro dominant expandiert ist. Im Unterschied zu ihren klonal-identischen Teff Gegenspielern, exprimieren die suppressiven Treg Zellen kaum CD127 und IL-2, aber hohe Mengen an IFNg und IL-10. Zusammen mit der signifikant erhöhten FOXP3 Expression, trotz unvollständiger foxp3-DNA Demethylierung, lassen sich die CMV-spezifischen CD4+CD25high Treg Zellen einem induzierten Treg (iTreg) Phänotyp zuordnen mit Ähnlichkeit zum beschriebenen Tr-1 Phänotyp. Darüber hinaus konnten wir die klonale TCR Identität auch in frisch isolierten CD4+CD25low und CD4+CD25high Subpopulationen bestätigen, was die Entstehung von CMV-spezifischen Treg Zellen bereits in vivo nahe legt. Periphere CD25high Treg Zellen supprimieren die anti-virale Immunantwort in Patienten mit häufigen CMV-Reaktivierungen, was auf ihre Bildung als Reaktion chronischer Antigenexposition interpretiert werden kann. Unsere Ergebnisse beweisen erstmals direkt, dass aus dem gleichen humanen T-Zell Klon Teff und Treg Zellen mit identischer Spezifität entstehen können und lassen vermuten, dass die Treg Induktion in der Peripherie durch häufige Antigenexposition vorangetrieben wird. / The majority of thymically arised regulatory CD4+CD25high T cells (Treg) show high affinity to self-antigens. It has been proposed that T-cell receptors (TCR) on Treg cells in the periphery also recognize foreign-antigens from pathogens, such as bacteria and viruses. Studies in mice have shown that peripheral Treg cells can be generated not only from naïve T cells but also from effector T cells (Teff). However, in humans the clonal TCR-repertoire of these Treg populations and their relation to effector CD4+CD25low Teff is not sufficiently known up to date. Here, we analyzed human TCRs derived from expanded Treg and Teff cells with defined specificity to MHC class-II restricted “foreign” epitopes of Cytomegalovirus (CMV). Remarkably, we found that both functionally distinct subsets share the same dominant TCR-CDR3 clones in vitro. In contrast to their Teff counterparts, the Treg cells express low CD127 and IL-2, but high IL-10 upon antigen stimulation. Therefore, together with increased FOXP3 expression, but incomplete foxp3 DNA-demethylation, human CMV-antigen specific Treg cells exhibit an induced phenotype (iTreg) in vitro with similarity to recently described Tr-1 phenotype. Moreover, the clonal identity was confirmed in freshly isolated CD4+CD25low and CD4+CD25high subsets, suggesting their generation occurred already in vivo. Peripheral CD25high Treg cells suppress the anti-viral immune response in patients with frequent CMV-reactivations, implying their development as reaction on chronic antigen-exposure. Our results demonstrate directly for the first time, that the same human T-cell clone can possess the phenotype of Teff and Treg cells with specificity to identical foreign epitopes and suggest that Treg-induction in the periphery is supported by frequent antigen-exposure.
179

Mecanismos embrionários de diferenciação de precursores coronários: princípios para aplicação em terapia celular. / Embryonic mechanisms of coronary precursor differentiation: principles for cell therapy.

Azambujá, Ana Paula 17 August 2009 (has links)
As coronárias derivam do proepicárdio, uma estrutura formada por precursores dos constituintes de vasos coronários, células endoteliais e musculares lisas (CoSMC). In vivo observa-se um marcante atraso entre a diferenciação endotelial e a integração de CoSMC à parede do vaso. O objetivo deste trabalho foi identificar os mecanismos que inibem a diferenciação a CoSMC in vivo. Baseados na perda progressiva da expressão de raldh2, a principal enzima de síntese de ácido retinóico (AR), nós exploramos a sinalização por AR como um possível inibidor da diferenciação a CoSMC. Através de um vetor adenoviral de expressão de raldh2 e da inibição in vivo da síntese de AR nós demonstramos que a sinalização por AR bloqueia a diferenciação a CoSMC dos precursores coronários. Nós também identificamos o VEGF como um fator chave no controle da diferenciação a CoSMC. Em conjunto, nossos dados suportam o modelo que a síntese de AR e VEGF durante o desenvolvimento cardíaco foi co-optada para o bloqueio da diferenciação a CoSMC até o estabelecimento de uma vasta malha vascular. / Coronary vessels derive from the proepicardium (PE), a structure formed by precursor of coronary vessels cells, endothelial and smooth muscle cells (CoSMC). In vivo there is a clear gap between the endothelial differentiation and the integration of CoSMC into the vascular tubes. The aim of this work was to understand the mechanisms controlling the delayed in vivo CoSMC differentiation. Based on the progressive loss of expression of raldh2, the main retinoic acid (RA) synthesizing enzyme, we explored the RA signaling as a possible candidate inhibitor of CoSMC differentiation. Using a adenoviral raldh2 expression system and in vivo inhibition of RA synthesis we showed that RA signaling act as a brake to slow CoSMC differentiation in PE-derived cells. We also identified VEGF as key factor acting on the control of CoSMC differentiation. Together our results support a model that AR and VEGF synthesis during cardiac development was co-opted to block the CoSMC differentiation of coronary precursors before an extensive endothelial network of tubes is established.
180

Cultura e caracterização de células do trofoblasto extraviloso (TEV) derivado da placenta humana a termo / Culture and characterization of extravillous trophoblast cells (EVT) derived from human term placenta

Fernandes, Isabella Rodrigues 14 December 2010 (has links)
A placenta é um anexo embrionário que tem atraído grande interesse como fonte de células-tronco para medicina regenerativa, devido à plasticidade fenotípica de alguns dentre os vários tipos celulares isolados a partir deste tecido. Apesar de terem a mesma origem, não fazem parte do embrião, portanto o uso da placenta como fonte de células embrionárias não provoca debates éticos. Uma característica que vale a pena mencionar, é que a placenta está envolvida na manutenção da tolerância do feto pelo organismo materno, pois contém células que apresentam propriedades imunomoduladoras. Por fim, o tecido placentário é disponibilizado após o parto e é geralmente descartado. Estas características tornam esse tecido de grande interesse para protocolos de terapia celular, tanto que tem surgido bancos de célulatronco de placenta humana. Alguns trabalhos demonstraram plasticidade de células extraídas da placenta, porém existe ainda a necessidade de se definir melhor a região de coleta e os métodos de extração e isolamento dessas células. Nosso grupo estabeleceu a cultura de células derivadas da região do trofoblasto extraviloso (TEV) de placenta humana a termo, que são as células responsáveis pelos mecanismos de imunotolerância materno-fetal. As células TEV apresentam os marcadores de pluripotencia Oct-4 e Nanog, e, portanto, podem reter mesmo extraídas da placenta a termo, alguma plasticidade celular, caracterizando-as como células-tronco. Entretanto, nossa experiência no cultivo destas células mostrou que existem limitações relacionadas ao tempo de cultivo celular e capacidade de proliferação das TEV, que certamente fornece argumentos sólidos para limitar o seu uso em protocolos de terapia celular em medicina regenerativa. / The placenta is attached embryo that has attracted great interest as a source of stem cells for regenerative medicine due to phenotypic plasticity of some of the various cell types isolated from this tissue. Despite having the same origin, they are not part of the embryo, so the use of placenta as a source of embryonic cells does not provoke ethical debates. A feature worth mentioning is that the placenta is involved in maintaining tolerance of the fetus by the mother, because it contains cells that have immunomodulatory properties. Finally, the placental tissue is present after birth and is usually discarded. These characteristics make this fabric of great interest for cell therapy protocols, which has emerged both banks of stem cells from human placenta. Some studies have demonstrated plasticity of cells extracted from the placenta, but there is still a need to better define the catchment area and the methods of extraction and isolation of these cells. Our group has established a culture of cells derived from the extravillous trophoblast region (TEV) from human placenta at term, which are the cells responsible for the mechanisms of maternalfetal immunotolerance. TEV cells have the pluripotency markers Oct-4 and Nanog, and therefore can retain, even extracted from the placenta at term, some cellular plasticity, characterizing them as stem cells. However, our experience in growing these cells showed that there are limitations related to the time of cell culture and proliferation capacity, which certainly provides strong arguments for limiting their use in cell therapy protocols in regenerative medicine.

Page generated in 6.4345 seconds