Spelling suggestions: "subject:"mesenchymal stromal cells"" "subject:"mesenchymal stromal wells""
111 |
Studie av celltillväxt hos kondrocyter och mesenkymala stromaceller från benmärg- en utvärdering av optimala odlingsförhållanden / Study of cell growth in chondrocytes and mesenchymal stroma cells from bone marrow - an evaluation of optimal culture conditionsEmilsson, Sara January 2023 (has links)
Introduktion: Hyalint brosk består av kondrocyter inbäddade i ett extracellulärt matrix som täcker alla kroppens synoviala leder. I dag är skador på hyalint brosk ett folkhälsoproblem med begränsade behandlingsmöjligheter. Mesenkymala stamceller har i flertalet studier visat sig vara en lovande kandidat för framtida behandling av broskskador, eftersom dessa kan differentiera till broskproducerande kondrocyter. Modern behandling av broskskador innefattar odling av patientens egna kondrocyter eller stamceller för att sedan återföra dem till broskskadan. För att göra detta effektivt behöver optimering av odlingsförhållanden för aktuella celler utföras. Syfte: Att undersöka tillväxt hos kondrocyter samt mesenkymala stromaceller från benmärg i varierande odlingsförhållanden, detta med avseende att utvärdera vilka tillsatser som mest gynnar cellernas viabilitet och proliferation. Material & Metod: Framtagning av kondrocyter och mesenkymala stromaceller från benmärg i denna studie utfördes på överblivet ortopediskt material från en individ. Cellerna kollagenasbehandlades innan de odlades ut i ett standardmedium under tre veckor. Därefter odlades de i närvaro av olika tillsatser under sex dagar. Cellernas viabilitet mättes genom relativ fluorescens med CellTiter-Blue® reagens medan proliferationen mättes genom cellräkning med flödescytometri. Resultat: Det fanns en signifikant skillnad (ANOVA, p<0,05) mellan minst två tillsatser inom grupperna för viabilitetsanalys samt cellräkning för både kondrocyter och benmärgderiverade mesenkymala stromaceller (BMMSC). Kondrocyterna som odlades i närvaro av fetalt bovint serum (FBS) hade högre proliferation och viabilitet än de celler som odlades i frånvaro av FBS. FBS i kombination med insulin-transferrin-selen (ITS) och kondrocytfaktorerna (KF) dexametason, prolin, insulin samt transforming growth factor beta hade störst positiv effekt på proliferationen. Vidare visade Minimum Essential Medium α med tillsats av FBS 10 % ha störst effekt, av den panel som undersöktes, på både viabilitet och proliferation hos BMMSC. Slutsats: FBS i kombination med ITS och KF hade störst stimulerande effekt på proliferation hos kondrocyter. Val av standardmedium hade större betydelse än tillsats av faktorer vid odling av BMMSC in vitro. / Introduction: Articular cartilage consists of chondrocytes and covers all the synovial joints in the body. Articular cartilage injuries are today a public health issue with limited orthopedic treatments. Mesenchymal stem cells have shown to be a promising candidate for future treatment of articular cartilage injuries due to their many abilities. Modern treatment of cartilage damage involves cultivation of the patient´s own chondrocytes and stem cells. To make this treatment as effective as possible the culture conditions for chondrocytes and stem cells must be optimized. Aim: To study cell growth in chondrocytes and mesenchymal stromal cells from bone marrow in varying culture conditions, this regarding to evaluate the most optimal culture conditions for cell viability and proliferation. Method: Cell extraction in this study was performed on leftover orthopedic material. The cells were treated with collagenase and then cultured in a standard medium for three weeks before they were cultured in presence of various factors for six days. Cell viability was measured by relative fluorescence while proliferation was measured by cell counting. Result: There was a statistically significant difference (ANOVA, p<0,05) between at least two additives within the groups for viability analysis and cell count for both chondrocytes and bone marrow-derived mesenchymal stromal cells (BMMSC). The chondrocytes cultured in presence of fetal bovine serum (FBS) had a higher proliferation and viability compared to the cells cultured in the absence of FBS. FBS in combination with insulin-transferrin-selenium (ITS) and the chondrocyte factors (CF) dexamethasone, proline, insulin and transforming growth factor beta had the biggest impact on proliferation. Minimum Essential Medium α with FBS 10% additive showed positive effects on both viability and proliferation of BMMSCs. Conclusion: FBS in combination with ITS and CF had a good effect on the proliferation of chondrocytes. The choice of standard medium seems to have a more important role than growth factor additive when studying BMMSCs expansion in vitro.
|
112 |
Bone marrow mesenchymal stromal cell-derived extracellular matrix displays altered glycosaminoglycan structure and impaired functionality in Myelodysplastic SyndromesKaur Bains, Amanpreet, Behrens Wu, Lena, Rivière, Jennifer, Rother, Sandra, Magno, Valentina, Friedrich, Jens, Werner, Carsten, Bornhäuser, Martin, Götze, Katharina S., Cross, Michael, Platzbecker, Uwe, Wobus, Manja 22 February 2024 (has links)
Myelodysplastic syndromes (MDS) comprise a heterogeneous group of hematologic malignancies characterized by clonal hematopoiesis, one or more cytopenias such as anemia, neutropenia, or thrombocytopenia, abnormal cellular maturation, and a high risk of progression to acute myeloid leukemia. The bone marrow microenvironment (BMME) in general and mesenchymal stromal cells (MSCs) in particular contribute to both the initiation and progression of MDS. However, little is known about the role of MSC-derived extracellular matrix (ECM) in this context. Therefore, we performed a comparative analysis of in vitro deposited MSC-derived ECM of different MDS subtypes and healthy controls. Atomic force microscopy analyses demonstrated that MDS ECM was significantly thicker and more compliant than those from healthy MSCs. Scanning electron microscopy showed a dense meshwork of fibrillar bundles connected by numerous smaller structures that span the distance between fibers in MDS ECM. Glycosaminoglycan (GAG) structures were detectable at high abundance in MDS ECM as white, sponge-like arrays on top of the fibrillar network. Quantification by Blyscan assay confirmed these observations, with higher concentrations of sulfated GAGs in MDS ECM. Fluorescent lectin staining with wheat germ agglutinin and peanut agglutinin demonstrated increased deposition of N-acetyl-glucosamine GAGs (hyaluronan (HA) and heparan sulfate) in low risk (LR) MDS ECM. Differential expression of N-acetyl-galactosamine GAGs (chondroitin sulfate, dermatan sulfate) was observed between LR- and high risk (HR)-MDS. Moreover, increased amounts of HA in the matrix of MSCs from LR-MDS patients were found to correlate with enhanced HA synthase 1 mRNA expression in these cells. Stimulation of mononuclear cells from healthy donors with low molecular weight HA resulted in an increased expression of various pro-inflammatory cytokines suggesting a contribution of the ECM to the inflammatory BMME typical of LR-MDS. CD34+ hematopoietic stem and progenitor cells (HSPCs) displayed an impaired differentiation potential after cultivation on MDS ECM and modified morphology accompanied by decreased integrin expression which mediate cell-matrix interaction. In summary, we provide evidence for structural alterations of the MSC-derived ECM in both LR- and HR-MDS. GAGs may play an important role in this remodeling processes during the malignant transformation which leads to the observed disturbance in the support of normal hematopoiesis.
|
113 |
Transforming Growth Factor Beta 3-Loaded Decellularized Equine Tendon Matrix for Orthopedic Tissue EngineeringRoth, Susanne Pauline, Brehm, Walter, Groß, Claudia, Scheibe, Patrick, Schubert, Susanna, Burk, Janina 09 February 2024 (has links)
Transforming growth factor beta 3 (TGF3) promotes tenogenic differentiation and
may enhance tendon regeneration in vivo. This study aimed to apply TGF3 absorbed in
decellularized equine superficial digital flexor tendon scaffolds, and to investigate the bioactivity
of scaffold-associated TGF3 in an in vitro model. TGF3 could effectively be loaded onto tendon
scaffolds so that at least 88% of the applied TGF3 were not detected in the rinsing fluid of the
TGF3-loaded scaffolds. Equine adipose tissue-derived multipotent mesenchymal stromal cells
(MSC) were then seeded on scaffolds loaded with 300 ng TGF3 to assess its bioactivity. Both
scaffold-associated TGF3 and TGF3 dissolved in the cell culture medium, the latter serving as
control group, promoted elongation of cell shapes and scaffold contraction (p < 0.05). Furthermore,
scaffold-associated and dissolved TGF3 affected MSC musculoskeletal gene expression in a similar
manner, with an upregulation of tenascin c and downregulation of other matrix molecules, most
markedly decorin (p < 0.05). These results demonstrate that the bioactivity of scaold-associated
TGF3 is preserved, thus TGF3 application via absorption in decellularized tendon scaffolds is a
feasible approach.
|
114 |
A1-reprogrammed mesenchymal stromal cells as a therapeutic vaccine against solid tumorsPereira Gonçalves, Marina 09 1900 (has links)
L'efficacité de la réponse antitumorale repose sur l'activité des cellules T cytotoxiques, qui peut être stimulée par des vaccins contenant des antigènes spécifiques aux tumeurs. Malgré le fait d'être les principales cellules présentatrices d'antigènes (CPA) responsables de l'activation des cellules TCD8, les cellules dendritiques (CD) ont rencontré des défis dans le développement de vaccins contre le cancer, notamment en ce qui concerne leur fabrication et efficacité. Pour combler ces problèmes, cette étude propose d’utiliser des cellules stromales mésenchymateuses (CSM) comme plateforme de vaccination alternative, en exploitant les avantages des CSMs en matière de fabrication, de sécurité et de plasticité. La plasticité remarquable des CSMs leur permet d'acquérir une capacité de présentation croisée sous des stimuli spécifiques. Étant donné que la présentation croisée est essentielle pour induire l'activation des cellules T contre les antigènes tumoraux, cette étude vise à convertir les CSMs en cellules présentatrice d‘antigènes en améliorant l'exportation des antigènes des endosomes vers le cytosol - une étape critique du processus. Dans cette démarche, nous avons examiné une librairie de molécules dérivés de l’Accum, une molécule initialement conçue pour favoriser la destruction de la membrane endosomale. Après avoir évalué leur potentiel à induire la présentation croisée, nous avons sélectionné la molécule A1 pour des investigations subséquentes. Les études mécanistiques ont démontré qu'A1 déclenchait des processus cellulaires essentiels favorisant une présentation croisée efficace, notamment une augmentation de la capture, dégradation et évasion des antigènes des endosomes ainsi que la production de d’espèces oxygénés réactifs. L'efficacité thérapeutique des CSMs reprogrammées par A1 (ARM) en tant que vaccin anticancéreux a été évaluée chez des souris ayant des tumeurs, en monothérapie et en combinaison avec l’anti-PD-1. La thérapie combinée ARM a induit une régression tumorale et a augmenté les taux de survie dans les modèles de tumeurs solides. En conclusion, cette étude présente une stratégie innovante pour transformer les CSM en cellules à présentation croisée en déclenchant l'échappement endosomal de l'antigène. Les cellules ARMs en association avec des inhibiteurs des points de contrôle immunitaire présentent un potentiel en tant que plateforme de vaccination contre les tumeurs solides. De plus, ces résultats soulignent l'importance de l'évasion des endosomes dans la présentation croisée d‘antigènes et ouvrent la voie à de nouvelles plateformes de vaccins contre le cancer. / The effectiveness of antitumoral response relies on cytotoxic T-cell activity, which can be stimulated through vaccines carrying tumor-specific antigens. Despite being the primary antigen-presenting cells (APCs) responsible for CD8 T-cell activation, dendritic cells (DCs) have encountered challenges in cancer vaccine development, particularly in manufacturing and efficiency. To address this gap, this study proposes mesenchymal stromal cells (MSCs) as an alternative vaccine platform, leveraging the advantages in manufacturing, safety profile, and plasticity of MSCs. The remarkable plasticity of MSCs enables them to acquire cross-presentation capacity under specific stimuli. Given that cross-presentation is pivotal for inducing T-cell activation against tumor antigens, this study aims to convert MSCs into antigen cross-presenting cells by enhancing the export of antigens from endosomes to the cytosol—a critical step in the process. In this pursuit, we screened a library of Accum and variant molecules designed to promote endosomal disruption. After evaluating their potential to induce cross-presentation, we selected the molecule A1 for further investigation. Mechanistic studies demonstrated that A1 triggers essential cellular processes supporting efficient cross-presentation, including enhanced antigen uptake, processing, endosomal escape, and reactive oxygen species production. The therapeutic efficacy of A1-reprogrammed MSCs (ARMs) as an anticancer vaccine was evaluated in tumor-bearing mice, as monotherapy and combined with anti-PD-1. In solid tumor models, ARMs combination therapy induced tumor regression and increased survival rates. In conclusion, this study presents an innovative strategy to transform MSCs into cross-presenting cells by triggering antigen endosomal escape. ARM cells in combination with immune checkpoint inhibitors hold potential as a vaccination platform against solid tumors. These findings underscore the importance of endosomal escape on antigen cross-presentation and pave the way for new cancer vaccine platforms.
|
115 |
Avaliação dos efeitos da sinvastatina via Inibidor do Ativador do Plasminogênio 1 (PAI-1) sobre a terapia celular com células estromais mesenquimais / Evaluation of the effects of simvastatin in mesenchymal stromal cell therapy through Plasminogen Activator inhibitor 1 (PAI-1)Faria, Carolina Arruda de 08 August 2016 (has links)
A Doença Pulmonar Obstrutiva Crônica (DPOC) é caracterizada pela limitação persistente de trocas gasosas, usualmente progressiva e associada a uma resposta inflamatória crônica exacerbada das vias aéreas a partículas e gases nocivos. Apesar de prevenível e tratável, não se logrou até o presente uma terapêutica eficaz, que resulte na cura da doença. Neste cenário, a terapia celular apresenta-se como uma alternativa terapêutica potencialmente promissora em DPOC, bem como em outras doenças pulmonares degenerativas e de caráter inflamatório. Porém, vários aspectos da terapia celular carecem de um melhor entendimento. Um dos principais desafios ao sucesso da terapia celular são as baixas taxas de sobrevivência das células transplantadas. O Inibidor do Ativador de Plasminogênio 1 (Plasminogen Activator Inhibitor 1 - PAI-1) pode representar um potencial mediador da sobrevivência de células estromais mesenquimais (CTM) pós-transplante, pois tem sido proposto que anticorpos neutralizadores do PAI-1 auxiliam no aumento da sobrevivência de CTM no tecido-alvo da terapia celular. Desta forma, a diminuição dos níveis de PAI-1 possui um potencial terapêutico interessante, ao modular os principais processos envolvidos na criação de um ambiente pouco propício ao \"homing\" celular durante o processo de injúria. A diminuição dos níveis de PAI-1 é promovida, pela sinvastatina, fármaco da família das estatinas. Desta forma, objetivou-se com este trabalho analisar os efeitos da sinvastatina sobre a expressão do PAI-1, bem como sua influência na sobrevivência das células infundidas para terapia celular de enfisema pulmonar em modelo murino. Camundongos da linhagem FVB foram submetidos à instilação intranasal de elastase para indução de enfisema pulmonar e, posteriormente, tratados com CTM do tecido adiposo e sinvastatina. Os resultados mostraram que, quanto aos aspectos morfológicos e funcionais, considerando-se a análise conjunta de ambos os pulmões, não houve diferença estatisticamente significativa entre os grupos submetidos à instilação intranasal de elastase e submetidos à terapia celular com CTM tratados ou não com sinvastatina. Quando porém os pulmões foram analisados individualmente constatou-se que não houve diferença estatisticamente significativa entre os grupos controle e os resultados referentes ao lado direito do pulmão dos animais tratados com elastase e que receberam sinvastatina e infusão de CTM. Diferenças anatômicas entre os lados direito e esquerdo do pulmão, levaram a uma maior deposição de células no lado direito, como evidenciado pelos resultados obtidos nos ensaios de bioluminescência. Pode-se, portanto, inferir que a recuperação morfológica no lado direito do pulmão de animais com DPOC/enfisema poderia ser decorrente de um efeito regenerativo parácrino das CTM associadas à sinvastatina. / Chronic Obstructive Pulmonary Disease - COPD is characterized by the persistent limitation of gas exchange, is usually progressive, and associated to a chronic augmented inflammatory response of the airways to particles and noxious gases. Despite preventable e treatable, an effective, curative therapeutic approach is yet to be achieved. In this context, cell therapy presents itself as a promising therapeutic approach for COPD and other pulmonary inflammatory and degenerative diseases. However, many aspects of cell therapy with stem cells remain unclear. One of the major challenges to the success of cell therapy are the low survival rates of transplanted cells. The Plasminogen Activator Inhibitor 1 (PAI-1) is a potential mediator of the survival of mesenchymal stromal cells (MSC) after transplantation, since PAI-1 neutralizing antibodies have been shown to increase the survival rate of MSC in the target tissue of cell therapy. Thus, the decreased levels of PAI-1 has an interesting therapeutic potential to modulate key processes involved in creating an inhospitable environment, during the process of injury, to the homing of transplanted cells. Decreased levels of PAI-1 are promoted by simvastatin, a drug of the statins family. Thus, the goal of this work was to evaluate the effect of simvastatin in vivo on the expression of PAI-1, as well as its influence on the survival rate of infused cells in mice model of cell therapy for pulmonary COPD/emphysema. FVB mice were submitted pulmonary emphysema induction by means of intranasal instillation of elastase and than treated with adipose-derived mesenchymal stem cells and simvastatin. The results regarding morphological and functional aspects, when considering the analisys of both lungs, presented no statistically significant difference among the groups submitted to intranasal instillation of elastase and cell therapy with MSC, treated or not with simvastatin. However, when the lungs where analyzed individually, it was found that there was no statistically significant difference between the control group and the results regarding the right lung of animals treated with elastase and that received simvastatin and MSC infusion. Anatomical differences between the right and left sides of the lung lead to a higher deposition of cells in the right side, as observed in the bioluminescence assays. Thus, it is possible to infer that the morphological recovery in the right side of the lung of animals with DPOC/emphysema could be due to a regenerative paracrine effect of mesenchymal stem cells associated with simvastatin.
|
116 |
Análise da expressão gênica por microarrays de células-tronco hematopoéticas e mesenquimais de pacientes com esclerose múltipla / Gene expression profiles of hematopoietic stem cells and mesenchymal stromal cells obtained from multiple sclerosis patients and detected by microarrays.Oliveira, Gislane Lelis Vilela de 22 February 2013 (has links)
As células-tronco hematopoéticas (CTHs) e estromais mesenquimais multipotentes (CTMs) isoladas da medula óssea vêm sendo utilizadas como fonte autóloga no tratamento de doenças autoimunes, como a esclerose múltipla (EM). As CTHs dão origem a todas as células dos sistemas hematopoético e imunológico e as CTMs possuem propriedades imunomoduladoras pela liberação de fatores solúveis e interação célula-célula. Existem trabalhos que sugerem que as doenças autoimunes sejam provenientes de defeitos intrínsecos nas células-tronco precursoras da medula óssea. Com o intuito de avaliar se as CTHs e CTMs de pacientes com EM possuem alterações intrínsecas, o objetivo geral deste trabalho foi avaliar o perfil de expressão gênica diferencial por microarrays de CTHs e CTMs de pacientes com EM, além de avaliar o perfil de expressão gênica de CTMs após o transplante autólogo de CTHs e a capacidade imunomoduladora in vitro das CTMs de pacientes. As CTHs e CTMs foram isoladas da medula óssea de pacientes com EM e doadores saudáveis, após consentimento informado. As CTHs foram isoladas por colunas imunomagnéticas e as CTMs foram isoladas por gradiente de densidade e submetidas à caracterização morfológica, imunofenotípica e capacidade de diferenciação em adipócitos e osteócitos. O RNA das CTHs e CTMs foi extraído e purificado e o perfil de expressão gênica foi avaliado por microarrays, utilizando hibridações em lâminas contendo 44.000 sondas. A capacidade imunomoduladora das CTMs de pacientes e controles foi avaliada por ensaios de cocultivo com linfócitos alogênicos e as citocinas foram quantificadas no sobrenadante por CBA flex e ELISA. Este estudo foi aprovado pelo comitê de ética do Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto. Os resultados mostraram que as CTHs de pacientes possuem perfis de expressão gênica diferentes dos controles, com 2.722 genes diferencialmente expressos, envolvidos em vias de sinalização importantes para manutenção/proliferação das CTHs e diferenciação em linhagens específicas durante a hematopoese. Dentre essas sinalizações estão incluídas as vias da apoptose, Wnt, Notch, mTOR, PI3K/Akt e Ca/NFAT, sugerindo que as CTHs de pacientes com EM possuam alterações intrínsecas que podem estar relacionadas com a patogenia da doença autoimune. As CTMs isoladas de pacientes com EM exibiram aparência senescente e reduzida expressão de marcadores imunofenotípicos. Com relação à expressão gênica, as CTMs de pacientes possuem perfil diferente das CTMs controle, sendo detectados 618 genes diferencialmente expressos, incluindo genes relacionados à sinalização FGF, HGF, sinalização de moléculas de adesão e moléculas envolvidas nos processos de imunorregulação, como IL10, IL6, TGFB1, IFNGR1, IFNGR2 e HGF. O perfil de expressão gênica das CTMs de pacientes pós-transplante assemelhou-se ao perfil das CTMs pré-transplante. Ensaios de cocultivo de CTMs com linfócitos alogênicos mostraram que as CTMs de pacientes possuem capacidade antiproliferativa reduzida em relação às CTMs controle, e ainda, secreção reduzida de TGF- e IL-10 no sobrenadante das coculturas. Esses dados sugerem que as CTMs isoladas de pacientes com EM possuam alterações fenotípicas, transcricionais e funcionais. Embasados nesses achados, concluímos que as CTHs e as CTMs de pacientes com EM possuem alterações intrínsecas que podem estar relacionadas com a patogenia da doença. Uma vez que as CTMs sejam células com grande potencial terapêutico para controle da EM em pacientes refratários aos tratamentos convencionais, as alterações encontradas sugerem que CTMs de doadores saudáveis sejam mais adequadas em aplicações clínicas. / Bone marrow hematopoietic stem cells (HSCs) and mesenchymal stromal cells (MSCs) have been used as an autologous source to treat autoimmune diseases, such as multiple sclerosis (MS). HSC give rise to all hematopoietic and immune system cells, and MSCs exhibit immunomodulatory properties by releasing soluble factors and by cell-cell interactions. Evidence indicates that bone marrow stem cells obtained from patients with autoimmune diseases may present intrinsic defects. To assess whether or not HSC and MSC of MS patients have intrinsic defects, the main objective of this study was to evaluate the differential gene expression profiles of HSC and MSC from MS patients before and after autologous HSC transplantation, and additionally, to evaluate the in vitro immunomodulatory ability of patient MSCs. Bone marrow HSC and MSCs were isolated from MS patients and healthy donors. HSCs were isolated by immunomagnetic columns and MSCs were isolated by gradient density and cultured until the third passage. MSCs were characterized according to morphology, immunophenotypic markers and cell differentiation into adipocytes and osteocytes. HSC and MSCs mRNAs were extracted, purified, and the gene expression profile was evaluated by microarray hybridizations, using a platform containing 44.000 probes. The immunomodulatory activity of patient and control MSCs was assessed by coculture assays with allogeneic lymphocytes. Cytokines were quantified in coculture supernatants by ELISA and CBA flex. This study was approved by the Ethics Committee of the University Hospital of the School of Medicine of Ribeirão Preto. The results showed that the patient HSCs exhibited a distinctive gene expression profile when compared to healthy HSCs, yielding 2.722 differentially expressed genes, involved in essential HSC signaling pathways for maintenance, proliferation and differentiation into specific lineages during hematopoiesis. Among these signaling pathways were included, apoptosis, Wnt, Notch, mTOR, PI3K/Akt and Ca/NFAT, suggesting that patient HSCs have significant intrinsic transcriptional alterations that may be associated with MS pathogenesis. Regarding MSCs isolated from MS patients, they exhibited senescence appearance, decreased expression of immunophenotypic markers, and also exhibited a distinctive gene expression profile in relation to healthy MSCs, yielding 618 genes differentially expressed genes, included in FGF and HGF signaling pathways, adhesion molecules, and genes involved in immunoregulation processes, such as IL-10, IL-6, TGFB1, IFNGR1, IFNGR2 and HGF. Coculture assays of control or patient MSCs with allogeneic lymphocytes showed that patient cells exhibited reduced antiproliferative activity as compared with controls, and also exhibited reduced secretion of TGF- and IL-10 cytokines in coculture supernatants. These data suggest that MSCs isolated from MS patients have phenotypic, functional and transcriptional defects, highlighting genes related to MSC maintenance, adhesion and immunomodulatory effects. According to these results, we concluded that patient HSCs and MSCs have intrinsic defects that may be associated with the disease per se. Considering that MSCs exhibit great therapeutic potential to control MS patients refractory to conventional treatment, the major MSCs alterations observed in this study indicate that healthy MSCs may be more suitable for MS cell therapy.
|
117 |
Perfil transcricional de fibroblastos de tumor primário, linfonodo e medula óssea de pacientes com câncer de mama / Transcriptional profile of fibroblasts obtained from primary tumor, lymph node and bone marrow of breast cancer patientsDel Valle, Paulo Roberto 01 March 2013 (has links)
Introdução: Em câncer de mama, existem evidências de que o microambiente pode influenciar o desenvolvimento do tumor no sítio primário, bem como em metástases regionais e a distância. Neste contexto, fibroblastos são importantes células estromais que podem influenciar a proliferação e a migração de células do câncer e podem prover um nicho apropriado para o desenvolvimento tumoral. Objetivos:O principal objetivo deste trabalho é comparar células estromais obtidas do tumor primário (PT), metástase linfonodal (N+) e medula óssea (BM) de pacientes com câncer de mama, através do perfil de expressão gênica. Pacientes e Métodos: Foi analisada a expressão gênica de fibroblastos (cultura primária) de 11 pacientes com câncer de mama. O perfil de expressão foi determinado em PT (n=4), N+(n=3) e BM (n=4) através de uma plataforma de cDNA microarray customizada (contendo 4.800 sequencias imobilizadas, representando cerca de 4600 genes), e os genes diferencialmente expressos foram identificados pelo teste SAM multiclasse, seguido pelo teste SAM de duas classes (TMEV, FDR 0%). A análise funcional foi realizada pelo software DAVID v6.7. Validação técnica foi realizada em 6 amostras previamente analisadas no microarray e a validação biológica em fibroblastos obtidos de outros 16 pacientes utilizando-se de RT-qPCR. Resultados: O perfil de expressão gênica dos fibroblastos obtidos de diferentes sítios mostraram 267 genes diferencialmente expressos, os quais apropriadamente agruparam os fibroblastos de acordo com suas origens (PT vs. N+ vs. BM). Apesar das diferenças entre PT e N+ serem representadas por 20 genes, as diferenças entre PT vs. BM e N+ vs BM foram mais significantes (235 e 245 genes diferencialmente expressos respectivamente). Análise funcional dos genes diferencialmente expressos mostrou enriquecimento de funções relacionadas ao desenvolvimento e morfogênese.A seguir, a expressão de alguns genes selecionados foi analisada em uma série diferente de amostras (validação biológica). Desse modo observamos que NOTCH2 confirmou uma alta expressão em N+ (vs. PT), e ADCY2, HECTD1, HNMT, LOX, MACF1 e USP16 confirmaram alta expressão em BM (vs PT). Conclusão:Em pacientes com câncer de mama, células estromais obtidas de diferentes origens apresentam um perfil de expressão gênica diferencial, o qual pode influenciar o comportamento do tumor / may influence tumor development in the primary site of breast cancer, as well as in regional and distant metastatic sites. In this context, fibroblasts are important stromal cells which influence proliferation and migration of cancer cells and may also provide an appropriate niche to tumor development. Objectives: The main objective of this work is the comparison of stromal cells from the primary tumor (PT), lymph node metastasis (N+) and bone marrow (BM) obtained from breast cancer patients, through gene expression profile. Patients and Methods: The gene expression profile was analyzed in fibroblasts primary culture from 11 breast cancer patients. The expression profiles of PT cells (n=4), N+ cells (n=3) and BM cells (n=4) were determined through a customized cDNA microarray platform (containing 4800 immobilized sequences which represents 4600 genes approximately). The analysis were performed by SAM multiclass (TMEV; FDR 0%), followed by SAM two classes test (TMEV; FDR 0%). Functional analysis was performed using DAVID v6.7. Technical validation was performed in same 6 samples that were previously analyzed in microarray experiments and biological validation was performed in fibroblasts obtained from other group of 16patients by RT-qPCR Results: The expression profile of fibroblasts obtained from three sites revealed 267 differentially expressed genes, which appropriately clustered fibroblasts in three different branches, in accordance with their origin (PT vs. N+ vs. BM). Although the differences between PT and N+ were represented by 20 genes, differences between PT vs. BM and N+ vs. BM were more significant (235 and 245 differentially expressed genes respectively). Functional analysis revealed enrichment of functions related to development and morphogenesis. Afterwards, the expression of some selected genes were analyzed in a different batch of samples (biological validation).Thereby, NOTCH2 confirmed high expression in N+ (vs. PT), and ADCY2, HECTD1, HNMT, LOX, MACF1 and USP16 confirmed high expression in BM (vs. PT). Conclusion: In breast cancer patients, stromal cells obtained from different origins present a differential gene expression profile, which may influence tumor behavior
|
118 |
Effet de l’association des basses concentrations d’O2 et des cellules stromales mésenchymateuses sur l’expansion ex vivo des cellules souches et progénitrices hématopoïétiques / Effect of the combination of low 02 concentration and mesenchymal stroml cells on ex vivo expansion of hematopoietic stem and progenitor cellsHammoud, Mohammad 02 October 2012 (has links)
Afin d’améliorer au mieux le greffon placentaire, nous suggérons de réaliser sa culture d’expansion ex vivo dans des conditions proches de l’environnement des cellules souches hématopoïétiques in vivo. Ainsi, nous proposons que la co-culture de cellules CD34+ placentaires avec des cellules stromales mésenchymateuses (CSM) à basses concentrations d’O2 (BC-O2) pourrait contribuer à équilibrer les processus d’autorenouvellement et de différenciation afin d’obtenir un greffon optimisé. Sur le plan fonctionnel, nos résultats confirment un effet bénéfique de notre modèle expérimental par rapport aux conditions où figure la culture simple et/ou l’oxygénation atmosphérique (20%) en termes du maintien de progéniteurs (PH) primitifs (pré-CFC) et de cellules souches Scid-Repopulating Cells (SRC). Sur le plan quantitatif, l’amplification des cellules CD34+ et des PH engagés, bien qu’elle soit en retrait dans nos conditions de référence par rapport à la condition de 20% d’O2, elle demeure néanmoins importante. Par ailleurs, le rôle de l’IL-3 exogène se montre crucial à BC-O2 notamment en co-culture à 1,5% d’O2 où elle permet non seulement de préserver mais aussi d’amplifier le taux de SRC par rapport au témoin de cellules CD34+ de J0. Enfin, l’étude de la sécrétion des facteurs solubles et l’expression des marqueurs phénotypiques sur les CSM montre que l’IL-6, le VEGF et l’IL-8 sont plus sécrétés et les CD146, CD49a, CD54, CD200 et CD105 sont plus exprimés après incubation à 5% d’O2. Cependant, l’implication réelle de ces facteurs et antigènes dans l’effet paracrine et/ou de contact cellulaire direct menés par les CSM dans notre protocole requiert de nouvelles investigations / To optimize at best the hematopoietic engraftment, we suggest in this work to improve the ex vivo expansion conditions by moving them closer to physiology. Indeed, we propose to culture placental CD34+ (HSC/PH) on MSC layer in combination with LO2-C to ensure the amplification of HP together with the maintenance/expansion of HSC. Compared to the single culture and/or atmospheric oxygenation, our experimental model allows a better maintenance of primitive HP (Pre-CFC) and HSC together with a quite good amplification of total cells, CD34+ cells and committed HP despite of lower than control condition. Moreover, exogenous IL-3 shows crucial effect in co-culture at LO2-C (1.5% O2) since its addition better preserves and even increases the number of HSC compared to the CD34+ cells control from D0. We then studied the secretion of soluble factors in culture supernatants and found that IL-6, VEGF and IL-8 were present in larger quantities at LO2-C in both co-culture and MSC culture. Finally, the CD146, CD49a, CD54, CD200 and CD105 membrane antigens appear to be up-regulated in MSCs when incubated at 5% O2. However, the involvement of these factors and antigens in paracrine effect and/or direct cell to cell contact mechanisms at LO2-C requires further investigations. In conclusion, the combination of LO2-C and MSC would be promising in the field of HSC/PH grafts expansion to achieve its main objective of reducing the post-transplant cytopenia period together with maintaining the long-term graft potential
|
119 |
Imunomodulação promovida pelo transplante de células tronco mesenquimais derivadas de medula óssea em lesões no sistema nervoso central / Immunomodulation promoted by bone marrow derived mesechymal stem cells transplanted in central nervous system injuriesGalindo, Layla Testa [UNIFESP] 22 February 2011 (has links) (PDF)
Made available in DSpace on 2015-07-22T20:50:21Z (GMT). No. of bitstreams: 0
Previous issue date: 2011-02-22. Added 1 bitstream(s) on 2015-08-11T03:26:13Z : No. of bitstreams: 1
Publico-12887.pdf: 1856396 bytes, checksum: 42de9f60d780e16634f25ebf960a3c24 (MD5) / Lesões no sistema nervoso central (SNC) levam a permeabilidade da barreira hematoencefálica, o que permite a entrada de células do sistema imune e a ativação das células da glia, principalmente microglia e astrócitos. Esse processo desencadeia a secreção de mediadores inflamatórios por essas células. As citocinas são as principais moléculas da resposta neuroinflamatória e são críticas para a regulação desta resposta, exercendo uma variedade de ações no SNC. Células tronco mesenquimais (CTMs), que possuem potencial proliferativo e são capazes de originar linhagens celulares distintas e especializadas, também secretam essas moléculas, caracterizando um poder imunomodulador. As CTMs, particularmente as derivadas da medula óssea, promovem o reparo tecidual pela secreção de fatores que aumentam a regeneração do tecido, estimulando proliferação, migração e diferenciação de progenitores endógenos encontrados na maioria dos tecidos, diminuindo a resposta imune e inflamatória e a apoptose. A habilidade de essas células alterarem o microambiente através de sua influência trófica pode contribuir mais significativamente para o reparo do tecido que a transdiferenciação. Nossa hipótese é que as citocinas secretadas pelas CTMs poderiam participar da atração de células tronco neurais endógenas para um local de lesão no SNC, criando um microambiente favorável para essas células. Tendo isso em vista, esta tese teve como objetivo estudar os efeitos dos fatores secretados pelas CTMs sobre células tronco neurais (CTNs) in vitro, e analisar a expressão de citocinas por CTMs in vivo em um modelo de lesão traumática no SNC. Primeiramente, avaliamos os efeitos dos fatores secretados pelas CTMs sobre apoptose, proliferação e diferenciação de CTNs adultas derivadas da zona subventricular e cultivadas como neuroesferas. Para isso, cultivamos as neuroesferas em meio condicionado por CTMs derivadas de medula óssea. Além disso, foram realizadas lesões no córtex motor primário dos animais, seguidas da injeção de CTMs no local da lesão. Nossos resultados indicam que os fatores secretados pelas CTMs não induzem nem previnem a apoptose das CTNs, aumentam a proliferação dessas células e induzem maior expressão do gene GFAP in vitro, o que indicaria uma tendência a diferenciação em astrócitos. Nos experimentos in vivo, nossos resultados mostram que a injeção das CTMs em um modelo de lesão aguda no SNC diminui a expressão de citocinas pró-inflamatórias no tecido lesado, indicando que os fatores solúveis secretados por CTMs podem modular a inflamação no local lesado, o que pode ser interessante para a criação de um microambiente favorável para CTNs endógenas e conseqüentemente para o reparo do tecido lesado. / Central nervous system (CNS) injury breakes the impermeability of the blood brain barrier, this allows the invasion of immune cells and activation of glial cells, mainly microglia and astrocytes. This process triggers the secretion of inflammatory mediators by these cells. Cytokines are the main molecules in neuroinflammatory response and are critical for its regulation, exerting a variety of actions in the CNS. Furthermore, mesenchymal stem cells (MSC) which have proliferative potential and are able to originate different and specialized cell lineages, also secrete these molecules, characterizing its immunomodulatory function. MSC, particularly those derived from bone marrow, promote tissue repair by secreting factors that enhance tissue regeneration stimulating proliferation, migration and differentiation of endogenous stem-like progenitors found in most tissues, decreasing inflammatory and immune reactions and apoptosis. The ability of such cells to alter tissue microenvironment through its trophic influence may contribute more significantly than their capacity for transdifferentiation in effecting tissue repair.Our hypothesis is that MSC secreted cytokines could take part in the attraction of endogenous neural stem cells (NSC) to an injury site in the CNS, providing a favorable microenvironment for these cells. Our aim was to study the effects of factors secreted by MSC on NSC in vitro and to analyse the MSC cytokines expression in vivo in a model of CNS traumatic injury. We first evaluated the effects of MSC secreted factors on apoptosis, proliferation and differentiation of adult NSC derived from the subventricular zone and cultured as neurospheres. Neurospheres were cultured in MSC conditioned medium (MSC-CM), which was obtained from bone marrow-derived MSC cultures. Besides a traumatic injury was performed at the primary motor cortex of mice and MSCs were injected at the injury site. Our results show that MSC secreted factors do not induce or prevent NSC apoptosis, increase NSC proliferation and induce bigger expression of GFAP gene in vitro, this could indicate a tendency of differentiation to astrocytes. In vivo experiments show that MSC injection at an acute model of injury diminishes pro-inflamatory cytokines in the injured tissue, suggesting that MSC secreted factors may modulate the inflammation at the injury site, which may be interest to the development favorable microenvironment for endogenous NSC and consequently repair of the injured tissue. / TEDE / BV UNIFESP: Teses e dissertações
|
120 |
Geração e caracterização de linhagens de células-tronco mesenquimais de camundongo geneticamente modificadas para expressão ectópica de hIGF-1 ou hG-CSFGonçalves, Gabrielle Viana Martins Gonçalves January 2015 (has links)
Submitted by Ana Maria Fiscina Sampaio (fiscina@bahia.fiocruz.br) on 2016-02-19T13:49:54Z
No. of bitstreams: 1
Gabrielle Viana Martins Gonçalves Geração...2015.pdf: 6199357 bytes, checksum: 605427028fc638e77cf5015ba759917c (MD5) / Approved for entry into archive by Ana Maria Fiscina Sampaio (fiscina@bahia.fiocruz.br) on 2016-02-19T13:50:09Z (GMT) No. of bitstreams: 1
Gabrielle Viana Martins Gonçalves Geração...2015.pdf: 6199357 bytes, checksum: 605427028fc638e77cf5015ba759917c (MD5) / Made available in DSpace on 2016-02-19T13:50:09Z (GMT). No. of bitstreams: 1
Gabrielle Viana Martins Gonçalves Geração...2015.pdf: 6199357 bytes, checksum: 605427028fc638e77cf5015ba759917c (MD5)
Previous issue date: 2015-01 / Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz. Salvador, BA, Brasil / As células-tronco mesenquimais (CTM) constituem uma ferramenta promissora para o campo
de terapia celular. Além de seu potencial de diferenciação em diferentes tipos celulares, as
CTM apresentam a habilidade de secretar moléculas bioativas e, assim, exercer múltiplos
efeitos biológicos, tais como indução da regeneração de tecidos lesionados, redução de fibrose
e modulação do sistema imune. A superexpressão dos fatores de crescimento G-CSF e IGF-1,
conhecidos por seus efeitos sobre os processos de imunomodulação, sobrevivência celular e
reparo tecidual, pode ampliar as ações terapêuticas das CTM. O objetivo deste trabalho
consiste em gerar e caracterizar linhagens de CTM de camundongo superexpressando hGCSF
ou hIGF-1. Um sistema lentiviral de segunda geração foi utilizado para modificação de
CTM para expressão ectópica dos genes de interesse. As sequências codificantes de hG-CSF e
hIGF-1 foram amplificadas por PCR e subclonadas em um vetor lentiviral de transferência,
contendo um promotor constitutivo. As partículas lentivirais foram produzidas a partir da cotransfecção
de células da linhagem HEK293FT com os vetores constituintes do sistema
lentiviral. Em seguida, as CTM obtidas da medula óssea de camundongos transgênicos para
proteína fluorescente verde (GFP) foram transduzidas com partículas lentivirais infectantes
contendo hG-CSF ou hIGF-1. A expressão gênica de hG-CSF ou hIGF-1 pelas linhagens
geradas foi quantificada por qRTPCR, e a produção da proteína por ELISA. As linhagens
foram caracterizadas por imunofenotipagem e avaliadas quanto ao seu potencial de
diferenciação celular. Foram geradas duas linhagens de CTM superexpressando hG-CSF e
três linhagens superexpressando hIGF-1. Todas demonstraram por qRTPCR, estar
efetivamente expressando os genes de interesse. Foi possível detectar e quantificar a síntese
proteica de G-CSF e IGF-1. Todas as linhagens geradas foram capazes de se diferenciar em
osteócitos, condrócitos e adipócitos, demonstrando a manutenção de seu fenótipo estromal.
Neste contexto, este trabalho resultou em ferramentas funcionais para a avaliação dos efeitos
terapêuticos de IGF-1 e G-CSF combinados à CTM, em modelos de lesões animais, em
comparação com CTM não-modificadas geneticamente. Além disso, estas ferramentas
poderão ser empregadas em estudos de pesquisa básica, para melhor compreensão dos efeitos
de hIGF-1 e hG-CSF sobre a biologia das CTM. / Mesenchymal stem cells (MSCs) are a promising tool for the cell therapy field. In addition to
their potential for differentiation into different cell types, MSCs have the ability to secrete
bioactive molecules and thus exert multiple biological effects such as induction of the injured
tissue regeneration, fibrosis reduction and modulation of the immune system. The
overexpression of the growth factors G-CSF and IGF-1, known for their effects on immune
modulation processes, cell survival and tissue repair, can result in a magnification of MSCs'
therapeutic actions. The objective of this work is to generate and characterize mouse MSCs
lines overexpressing hG-CSF or hIGF-1. A second generation lentiviral system was used to
modify MSCs derived from mice for the ectopic expression of the genes of interest. The
coding sequences of hG-CSF and hIGF-1 were amplified by PCR and subcloned into a
lentiviral transfer vector containing a constitutive promoter. The lentiviral particles were
produced from the co-transfection of HEK293FT lineage cells with the lentiviral vectors.
Subsequently, MSCs obtained from the bone marrow of transgenic mice for green fluorescent
protein (GFP) were transduced with infectious lentiviral particles containing hG-CSF or
hIGF-1. The gene expression of hG-CSF or hIGF-1 by the generated cell lines was quantified
by qRTPCR, and the protein production by ELISA. The lineages were characterized by
immunophenotyping and evaluated for their potential of cellular differentiation. Two lines of
MSCs overexpressing hG-CSF and three lines overexpressing hIGF-1 were generated. All the
cell lines demonstrated to be effectively expressing the genes of interest by qRTPCR. It was
possible to detect and quantify the protein synthesis of G-CSF and IGF-1. Moreover, all the
generated lines were capable of differentiating into osteocytes, chondrocytes and adipocytes,
indicating the conservation of their stromal phenotype even after genetic modification. In this
context, this study resulted in functional tools for evaluating the IGF-1 and G-CSF
therapeutic effects when combined with MSCs, to be tested in experimental animal models in
comparison to non-genetically modified MSCs. Furthermore, these tools may be employed
for basic research studies, for a better understanding of the effects of hIGF-1 and hG-CSF on
MSCs' biology
|
Page generated in 0.0841 seconds