• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 87
  • 68
  • 23
  • 10
  • 10
  • 10
  • 7
  • 5
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • Tagged with
  • 241
  • 241
  • 152
  • 71
  • 68
  • 62
  • 57
  • 54
  • 46
  • 39
  • 32
  • 30
  • 30
  • 29
  • 28
  • 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.
211

Papel de Notch e NF-kB na regulação de fatores de transcrição durante a diferenciação in vitro de células T a partir de células progenitoras hematopoéticas CD34+ / Role of Notch and NF-kB in the regulation of transcription factors during in vitro differentiation of T cells from CD34+

Josiane Lilian dos Santos Schiavinato 01 April 2011 (has links)
Em estudos anteriores desenvolvidos por este grupo de pesquisa uma expressão mais elevada de alvos transcricionais e componentes da via NF-kB, bem como altos níveis de NOTCH1, foi identificada em células-tronco hematopoéticas (CTH) CD34+ de sangue de cordão umbilical (SCU) quando comparadas às CTH CD34+ de medula óssea (MO). Este grupo verificou ainda, por comparação das células CD34+ com as CD133+ (mais primitivas) que diversos fatores de transcrição (FT) envolvidos com o potencial de hemangioblasto, com a autorenovação das CTH, e com a diferenciação linfóide; como: RUNX1/AML1, GATA3, USF1, TAL1/SCL, HOXA9 e HOXB4 apresentaram-se mais expressos em células mais primitivas. A potencial participação das vias Notch e NF-kB na regulação destes FT tem importância conceitual e prática no entendimento da biologia das CTH, e dos processos envolvidos na diferenciação destas células. Com isto em vista, este projeto teve como objetivo, estudar o papel da via NF-kB e da via Notch na regulação destes FT. Para isso, um modelo experimental in vitro, de diferenciação de CTH CD34+ em linfócitos T, foi utilizado e a influência de fatores agonistas e inibidores farmacológicos destas vias, foram avaliados por citometria de fluxo e PCR em tempo real. Nossos resultados evidenciam o papel da via Notch na regulação transcricional de HOXB4 e GATA3 em células-tronco hematopoéticas CD34+ humanas, o que foi confirmado com base na expressão dos alvos diretos de Notch (HEY1 e HES1). Notamos ainda, que a expressão dos transcritos HES1, GATA3 e HOXB4 é prejudicada pela síntese protéica das CTH, uma vez que quando empregamos o prétratamento com a droga CHX há aumento da transcrição dos mesmos. Também podemos inferir que a ação do TNF- é positiva sobre esses transcritos, já que quando o utilizamos há elevação do nível de expressão desses transcritos, com exceção a HES1. Em relação ao cocultivo das CTH com as células estromais de camundongos, verificamos que apenas a linhagem OP9-DL1 detém a capacidade de promover a diferenciação celular T, e isso foi comprovado pelo surgimento de células comprometidas com a linhagem linfocítica T, através da presença dos marcadores de superfície específico CD7+ e CD1a+. Esses resultados auxiliarão na compreensão dos mecanismos moleculares de regulação transcricional envolvidos não apenas na diferenciação de linfócitos T, mas também na manutenção de um estado mais primitivo das CTH. Este conhecimento pode vir a contribuir com o desenvolvimento ou otimização de protocolos laboratoriais visando à expansão de CTH ou geração de células T para usos terapêuticos. / In previous studies by this research group a higher expression of transcriptional targets and components via NF-kB, as well as high levels of NOTCH1, was identified in hematopoietic stem cells (HSC) CD34 + cells from umbilical cord blood (UCB) compared to CD34 + hematopoietic stem cells from bone marrow (BM). This group also found, by comparing the CD34 + cells with CD133 + (more primitive) that several transcription factors (TF) involved in the potential of hemangioblast, with self-renewal of hematopoietic stem cells and to differentiated lymphocytic; as Runx1 / AML1, GATA3, USF1, TAL1/SCL, HOXB4 and HOXA9 were more expressed in more primitive cells. The potential involvement of Notch signaling pathways and NF-kB in the regulation of FT has conceptual and practical importance in understanding the biology of HSC, and the processes involved in differentiation of these cells. With this in mind, this project aimed to study the role of NF-kB pathway and Notch signaling in the regulation of FT. For this, an experimental model in vitro differentiation of CD34 + hematopoietic stem cells into T lymphocytes, was used and the influence of pharmacological agonists and inhibitors of these pathways were evaluated by flow cytometry and real-time PCR. Our results highlight the role of Notch signaling in the transcriptional regulation of GATA3 and HOXB4 in hematopoietic stem cells CD34 + human, which was confirmed based on the expression of direct targets of Notch (HES1 and HEY1). We also note that the expression of transcripts HES1, GATA3 and HOXB4 protein synthesis is hampered by the HSC, since when we use the pre-treatment with the drug there CHX increased transcription thereof. We can also infer that the action of TNF- is positive about these transcripts, since when we use it for raising the level of expression of these transcripts, except the HES1. In relation to the HSC coculture with stromal cells of mice, we found that only the line-DL1 Op9 has the ability to promote T cell differentiation, and this was evidenced by the appearance of cells committed to the T lymphocyte lineage, through the presence of specific surface markers CD7 + and CD1a +. These results will help understand the molecular mechanisms of transcriptional regulation involved not only in the differentiation of T lymphocytes, but also in maintaining a more primitive state of HSC. This knowledge may contribute to the development or optimization of laboratory protocols aimed at the expansion of HSC or generation of T cells for therapeutic use.
212

Influência do microambiente no prognóstico do câncer da mama / Influence of the microenvironment on breast cancer prognosis

Fabiana Baroni Alves Makdissi 19 February 2014 (has links)
Introdução: Os cânceres de mama subtipos Luminal A e B (HER2 negativo) podem apresentar prognóstico variável, a depender do índice de proliferação, avaliado pelo Ki67. As células malignas e as células estromais adjacentes (fibroblastos e células de resposta imune ) podem interagir tanto pelo contato célula a célula como por fatores secretados por elas, ambas influenciando no comportamento tumoral. Já foi demonstrado que as células estromais podem aumentar a proliferação das células do câncer da mama. Objetivo: Nosso objetivo foi avaliar o perfil de expressão gênica de células do estroma em câncer de mama luminal A e luminal B e analisar se este se correlaciona com o prognóstico da doença. Pacientes e Métodos/ Resultados: Amostras de tumores de 11 pacientes na pós menopausa foram analisadas, todas elas HER2 negativas. A expressão de Ki67 foi <= 10 % em 5 pacientes (luminal A) e >= 30 % em outras 6 amostras(Luminal B ). Células estromais foram microdissecadas para a extração de RNA, que posteriormente foi hibridizado na plataforma de microarray Agilent G485 -1A GE 8x60K. Após a normalização, 50 % dos genes com a maior variância foram selecionados para análise por SAM duas classes desemparelhado (software TMEV ) e aceitando FDR 14.1%, 35 sequências foram identificadas como diferencialmente expressas, incluindo 16 genes conhecidos, entre as células estromais das amostras de Luminal A versos Luminal B, todos mais expressos nas amostras B. Dentre as funções biológicas enriquecidas em genes diferencialmente expressos encontram-se regulação positiva do sistema imune, incluindo genes como ZAP70 (proteína quinase 70kDa associada a cadeia zeta (TCR)), CD38 (molécula CD38); UBASH3A (ubiquitina associada e SH3 domínio que contém A); PLA2G7 (fosfolipase A2, grupo VII (fator acetil ativador de plaquetas no plasma)); NCR3 (citotoxicidade natural, provocando receptor 3). Nosso próximo passo foi avaliar se a expressão de alguns genes selecionados estava associada com prognóstico de tumores luminais. Para tal selecionamos amostras de outro grupo de 89 pacientes com seguimento de pelo menos 5 anos, cujos tumores eram ER(+), HER2(-), para análise de expressão proteica em Tissue microarray. Caracterizamos os fibroblastos destas amostras com 3 marcadores de fibroblastos: actina de músculo liso (AML), S100A4 e caveolina-1 (CAV1) e analisamos a marcação da proteína ZAP70. Correlacionamos a expressão proteica de todos os marcadores com as características anatomopatológicas da amostra. Observamos que fibroblastos de todas as amostras de tumor de mama expressam AML, S100A4 e CAV1, em diferentes proporções, entretanto não detectamos diferença entre os tumores luminais A e B. Também não obsevamos diferença de expressão de AML, S100A4 e CAV1 em relação a grau histológico, comprometimento linfonodal e estadiamento clínico. Nestas amostras não detectamos expressão proteica de ZAP70 em fibroblastos tumorais. Conclusão: Houve expressão diferencial de 16 genes relacionados a processos imunes, todos eles mais expressos em células estromais de tumores Luminal B em relação a luminal A / Introduction: Luminal breast cancer subtypes A and B (HER2 negative) may present a variable prognosis, depending on tumor proliferation index, evaluated by Ki67 expression. Malignant cells and adjacent stromal cells (fibroblasts and immune response cells) may interact by both cell contact and secreted factors and influence tumor behavior. It was shown that stromal cells may enhance breast cancer cells proliferation. Objective: Our aim was to evaluate stromal cells gene expression profile in luminal A and luminal B tumors and to evaluate whether selected transcripts expressed in stromal cells may be associated with prognosis in breast cancer. Material/ Methods and Results: Hormone receptor positive tumor samples from 11 post menopausal patients were analyzed, all of them Her2 negative. Ki67 expression <= 10% (luminal A) was observed in five and Ki67 >= 30% (luminal B) in six samples. Stromal cells were microdissected for RNA extraction, which was hybridized in Agilent G485-1A GE 8x60K microarray platform. After normalization, 50% of the genes with the highest variance were selected for further analysis by two class unpaired SAM (TMEV software) and accepting FDR 14,1%, 35 sequences, including 16 known genes, were found differentially expressed between stromal cells from luminal A vs luminal B breast cancer samples, all of them more expressed in luminal B. Among biological functions enriched in genes found differentially expressed were positive regulation of immune system process, including genes as: ZAP70 (zeta-chain (TCR) associated protein kinase 70kDa); CD38 (CD38 molecule); UBASH3A (ubiquitin associated and SH3 domain containing A); PLA2G7 (phospholipase A2, group VII (platelet-activating factor acetylhydrolase, plasma); NCR3 (natural cytotoxicity triggering receptor 3). Our next step was evaluate whether expression of selected genes was associated with prognosis in another group of patients. Tumor samples from 89 patients with at least 5 years of follow up, all of them estrogen receptor positive and HER2 negative, were selected. Tissue microarray was prepared with stromal tumor compartment from paraffin embedded tumor samples. Fibroblasts were characterized for the expression of 3 fibroblasts markers (alfa-SMA, alpha smooth muscel actin; S100A4 and CAV1, caveolin 1), and ZAP70. Correlation of expression of these markers with prognostic variables was determined. Expression of alfa-SMA, S100A4 and CAV1 was detected in fibroblasts from all tumor samples in different proportions, however no differential expression was observed between luminal A and B tumors. Neither difference was detected on the expression of these proteins in relation with histological grade, lymph node involvement and clinical stage. Conclusion: A differential expression of 16 genes involved in immune process was found, all of them more expressed in fibroblasts from luminal B as compared with luminal A tumors
213

Expressão gênica diferencial das células estromais obtidas de medula óssea na presença ou ausência de célula tumoral oculta em pacientes com câncer de mama / Differential gene expression of bone marrow stromal cells from breast cancer patients in the presence or abscence of occult tumor cells

Cintia Milani 21 September 2006 (has links)
A célula estromal pode influenciar o desenvolvimento do tumor no sítio primário e secundário, mas pouco é conhecido sobre as características moleculares das células estromais presentes na medula óssea de pacientes com câncer de mama. Nosso objetivo foi avaliar a expressão gênica diferencial entre as células estromais oriundas de medula óssea na presença ou ausência de célula tumoral oculta. Coletamos dez aspirados de medula óssea das pacientes com câncer de mama. A classificação do comprometimento da medula por células tumorais ocultas foi realizada pela detecção da expressão de CK19 por Nested-RT-PCR e quatro entre dez pacientes apresentaram presença de célula tumoral na medula óssea. Estabelecemos culturas primárias de células estromais de todas as amostras e, selecionamos amostras originárias de duas pacientes contendo linfonodos comprometidos e presença de célula tumoral oculta em medula e também de duas pacientes que não apresentavam linfonodos comprometidos e nem célula tumoral oculta na medula. As pacientes selecionadas eram pós-menopausadas com diagnóstico de carcinoma ductal invasor e expressão imunohistoquímica positiva para receptor de estrógeno e progesterona. Realizamos avaliação do perfil de expressão gênica entre estes dois grupos, o que nos revelou 21 genes diferencialmente expressos dentre os 4.608 genes imobilizados em lâmina de cDNA microarray; nove genes hiperexpressos em célula estromal de medula comprometida (PTHLH, TLOC1, NCOA6, C17orf57, ANAPC11, MAST4, POLR3E, CPNE1 e B4GALT5) e doze genes hipoexpressos em célula estromal de medula comprometida (MRPL2, NAT10, DAP, RNF2, FLOT2, FKBP10, SLIT3, EBNA1BP2, SLC35B2, MICAL2, GPR3, TSPAN17). Nossos dados sugerem que apesar da expressão gênica de células estromais oriundas de medula óssea comprometida ou não por micrometástases ser semelhante, algumas diferenças podem ser identificadas. / Stromal cells may influence tumor development in primary and secundary sites, however, molecular characteristics of bone marrow stromal cells from breast cancer patients are almost unknown. Our aim was to evaluate the differential gene expression of bone marrow stromal cells from breast cancer patients in the presence or abscence of occult tumor cells. Bone marrow (BM) aspirates were obtained from 10 breast cancer patients. The presence of occult bone marrow disseminated tumor cells was detected by CK19 expression quantified by reverse transcriptase polymerase chain reaction (RT-PCR). Presence of tumoral cell was detected in four of ten BM samples. Stromal cells primary cultures were established and samples from two patients with positive lymph nodes and presence of occult tumor cells in bone marrow and samples from two patients with negative lymph nodes and abscence of occult tumor cells in bone marrow were selected. All the included patients were postmenopausal with invasive ductal carcinoma and positive estrogen and progesterone receptors detected by immunohistochemical analysis. Gene profile evaluated in cDNA microarray slides containing 4.608 spotted genes revealed 21 differencially expressed genes, nine upregulated (PTHLH, TLOC1, NCOA6, C17orf57, ANAPC11, MAST4, POLR3E, CPNE1 e B4GALT5) and twelve downregulated (MRPL2, NAT10, DAP, RNF2, FLOT2, FKBP10, SLIT3, EBNA1BP2, SLC35B2, MICAL2, GPR3, TSPAN17) in stromal cell derived from bone marrow in the presence of tumor breast cancer cell. Our data suggest that gene expression from bone marrow derived stromall cells in the presence or abscence of occult tumor cells seems similar, however small differences may be identified.
214

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 patients

Paulo Roberto Del Valle 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
215

Développement de modèles précliniques humanisés autologues en immuno-oncologie

Moquin-Beaudry, Gaël 08 1900 (has links)
La reconnaissance de l’implication du système immunitaire dans le cancer a guidé l’industrie vers de développement d’immunothérapies nombreuses et prometteuses. Or, à l’ère de l’immuno-oncologie, on constate un manque criant de modèles précliniques capables de simuler les interactions immunitaires entre un patient et sa tumeur. Pour remédier à cette situation, nous avons développé des modèles de souris humanisées combinant la reconstitution immunitaire de souris immunodéficiente et l’injection de lignées tumorales issues d’un même donneur. L’utilisation de cellules souches pluripotentes induites (iPSC) a permis notamment le développement de multiples lignées tumorales à partir d’un seul donneur sain, facilitant ainsi l’accès aux cellules immunitaires nécessaires à l’humanisation des souris. La transformation des cellules primaires ou dérivées d’iPSC a été faite par la transduction lentivirale des proto-oncogènes de la télomérase (hTERT), de Ras oncogénique (HRASV12) et de la région précoce du viruse simen 40 (SV40ER) encodant les gros et petits antigènes T (LgT et SmT). Cette approche permis de générer des tumeurs de haut grade, agressives et peu différenciées à l’aide de fibroblastes primaires et de cellules hépatiques, de cellules souches neurales et d’astrocytes dérivés d’iPSC. Dans tous les cas, les tumeurs ainsi générées ont été efficacement reconnues, infiltrées et souvent rejetées par le système immunitaire autologue implanté. Le rejet partiel de la plupart de ces tumeurs ouvre toutefois la porte à l’évaluation préclinique d’immunothérapies diverses reposant sur les réactions immunitaires anti-tumorales de l’hôte. Par exemple, nous avons pu étudier l’impact d’un traitement d’inhibition du point de contrôle immunitaire PD-1 sur la croissance de tumeurs d’origine fibroblastique où une augmentation marquée du taux d’infiltration immunitaire humaine a été observé sans toutefois mener à une réduction significative du fardeau tumoral. Nous avons aussi pu produire, de façon autologue, des lymphocytes T exprimant un récepteur d’antigène chimérique (CAR) contre le ganglioside GD2, un antigène tumoral préalablement identifié et détecté sur les tumeurs de cellules souches neurales générées par notre approche. L’efficacité cytotoxique de ces CAR a ainsi pu être validée in vitro dans un système autologue. Finalement, nous avons utilisé le modèle de tumeurs fibroblastiques dans des contextes immunitaires autologues et allogéniques pour déterminer si le potentiel immunomodulateur des cellules stromales mésenchymateuses (MSC) pouvait affecter la croissance tumorale. Selon nos résultats, les MSC n’auraient aucun effet ni sur le taux d’émergence et de croissance tumoral, ni sur l’infiltrat immunitaire, suggérant que leur utilisation thérapeutique serait sécuritaire en ce qui concerne ce type de tumeurs ayant préalablement un microenvironnement tumoral immunosuppresseur. En somme, les modèles innovateurs décrits dans cette thèse visent à améliorer la qualité prédictive des modèles murins précliniques en immuno-oncologie en récapitulant certaines interactions immunitaires entre un patient et sa tumeur. La grande flexibilité de cette approche permettra d’adapter aisément le modèle aux problématiques d’intérêt, tant fondamentales que précliniques. / Identification of the human’s immune system implication in cancer has guided the biotech industry towards the development of numerous and promising cancer immunotherapies. However, in the era of immuno-oncology, a distinct lack preclinical models can simulate the interactions between a patient’s tumor and immune cells. To tackle this issue, we developed humanized mouse models combining immune reconstitution of immunodeficient mice and injection of tumor cells lines from the same human donor. The use of induced pluripotent stem cells (iPSC) allowed the generation of multiple tumorigenic cell lines from a single donor, facilitating access to autologous immune cells necessary for mouse immune humanization. The transformation of primary or iPSC-derived cell lines was done using lentiviral transduction of proto-oncogenes telomerase (hTERT), oncogenic Ras (HRASV12) and simian virus 40 early region (SV40ER) encoding large and small T antigens (LgT and SmT). This approach allowed to generate high grade, aggressive and undifferentiated tumors from primary fibroblasts and iPSC-derived hepatic cells, neural stem cells and astrocytes. In all cases, such tumors were efficiently recognized, infiltrated and often rejected by the implanted autologous immune system. However, partial rejection of most tumors allows for preclinical evaluation of targeted immunotherapies relying on the hosts’ pre-existing immune response. For instance, we could study the impact of PD-1 checkpoint blockade inhibition on tumor growth in fibroblastic tumors where a significant increase in tumor infiltration was observed, but without an associated decrease in tumor burden. We could also produce autologous chimeric antigen receptor (CAR)-expressing T lymphocytes against GD2 ganglioside, a previously described tumor antigen detected on our neural stem cell-derived tumor cells. Cytotoxic efficiency of these autologous CAR T cells could thus be validated in vitro. Finally, we used our fibroblast-derived tumor models in autologous and allogeneic settings to determine if mesenchymal stem cells’ (MSC) immunomodulatory potential could impact tumor growth. Our results showed that MSC had no effect neither on tumor emergence and growth nor on immune infiltration, suggesting therapeutic use of these cells should be safe regarding such tumors already harboring a strongly immunodeficient microenvironment. Overall, the novel models described in this thesis aim at improving the predictive capacity of mouse pre-clinical models in immuno-oncology by recapitulating some immune interactions between a patient and its tumor. The great flexibility of this approach will allow for easy adaptation to many research problematics both preclinical and fundamental.
216

Generation of a stem cell driven in vitro culture of polarized cells to study gastric tissue homeostasis and response to infections

Wölffling, Sarah 07 September 2020 (has links)
In der humanen Magenschleimhaut regulieren eine Vielzahl von Interaktionen zwischen verschiedenen Zellpopulationen die Verdauung und die Überwachung von Infektionen. Epithelzellen in der Schleimhaut differenzieren in spezialisierte Zelltypen, die schützenden Mukus, Magensäure, Verdauungsenzyme oder Hormone produzieren. Eine Infektion mit Helicobacter pylori kann die Gewebehomöostase fehlregulieren, was die Wahrscheinlichkeit erhöht, dass an der Infektionsstelle ein Magengeschwür, ein Adenokarzinom oder letztendlich Magenkrebs auftritt. In dieser Arbeit wird die Entwicklung eines neuartigen in vitro Kulturmodells für humane primäre Magenepithelzellen, die sogenannte Mukosoidkultur, gezeigt. Die Mukosoidkulturen sind repräsentativ für Epithelbarrieren und rekapitulieren die meisten Funktionen der menschlichen Magenschleimhaut in vivo, einschließlich der Schleimproduktion, und ermöglichen eine langfristige und stabile Kultivierung von Epithelzellen sowie Infektionsstudien mit H.pylori. Mukosoidkulturen aus Corpus wurden verwendet, um die Nischenfaktoren zu untersuchen, die die Differenzierung von Oberflächenepithelzellen, Hauptzellen und Parietalzellen fördern. EGF erwies sich zusammen mit BMP/Noggin als ein wichtiger Regulator bei der Differenzierung. Stromazellen sind Teil der Lamina propria der Magenschleimhaut. Über die Wechselwirkung mit dem Epithel unter homöostatischen Bedingungen und bei bakteriellen Infektionen mit H.pylori ist nur sehr wenig bekannt. Die Co-Kultur von humanen primären Stromazellen des Magens mit Epithelzellen unter Verwendung des Mukosoidkultur-Modells zeigte die aktive Signalübertragung zwischen beiden Zelltypen auf. Darüber hinaus wurden Mukosoidkulturen erfolgreich mit H.pylori infiziert. Die Ergebnisse zeigen, dass Stromazellen aktiv mit Cytokin- und Chemokinexpression auf eine epitheliale Infektion reagieren. Gleichzeitig erhöhten Stromazellen die NFκB-gesteuerte Entzündungsreaktion in Epithelzellen. / In the human gastric mucosa, multiple interactions between different cell populations regulate digestion and surveillance of infections. Epithelial cells in the mucosa differentiate into specialized cell types to produce protective mucins, gastric acid, digestive enzymes or hormones. Infection with Helicobacter pylori dysregulates the tissue homeostasis increasing the chance to develop a gastric ulcer, adenocarcinoma or ultimately gastric cancer at the site of infection. In this thesis, the development of a novel in vitro culture model for human primary gastric epithelial cells, called the mucosoid culture, is shown. The mucosoid cultures are representative of epithelial barriers and recapitulate most of the functions of the human gastric mucosa in vivo, including mucus production, and allow long-term and stable cultivation of epithelial cells as well as infection studies with H.pylori. Corpus derived mucosoids were used to investigate the niche factors that promote the differentiation of foveolar cells, chief cells, and parietal cells. EGF was found to be a major regulator in differentiation together with BMP/Noggin. Stromal cells are part of the lamina propria of the gastric mucosa. Very little is known about the interaction with the epithelium under homeostatic conditions and during bacterial infections with Helicobacter pylori. The co-culture of human primary gastric stromal cells with epithelial cells using the mucosoid culture model demonstrated the active signaling between both cell types. Furthermore, mucosoid cultures were successfully infected with H. pylori. The results revealed that stromal cells actively respond to epithelial infection with cytokine and chemokine expression. Concurrently stromal cells increased the NFκB-driven inflammatory response in epithelial cells.
217

Mezenchymální stromální buňky a biologické scaffoldy pro regeneraci nervové tkáně / Mesenchymal stromal cells and biological scaffolds for neural tissue regeneration

Kočí, Zuzana January 2018 (has links)
Despite tremendous progress in medicine, injuries of the adult central neural system remain without satisfactory solution. Regenerative medicine employs tissue engineering, cellular therapies, medical devices, gene therapy, or growth factors with the aim to bridge the lesion, re-establish lost connections and enhance endogenous repair in order to restore neural function. The aim of my thesis was to evaluate therapeutic potential of two approaches, transplantation of human mesenchymal stromal cells (hMSCs) and biological scaffolds derived from extracellular matrix (ECM) for neural regeneration, particularly in models of spinal cord injury (SCI). First, hMSCs from various sources - bone marrow (BM), adipose tissue (AT) and Wharton's jelly (WJ) - were isolated and characterized in vitro. All cell types met the minimal criteria for MSC phenotype and displayed similar properties in terms of their surface marker expression, differentiation potential, migratory capacity, and secretion of cytokines and growth factors. On the other hand, the cell yield from WJ and AT was significantly higher, and MSCs isolated from these tissues proliferated better than from BM. Therapeutic effect of intrathecal application of hWJ-MSCs was then evaluated in SCI compression model in rats. The effect of low (0.5 million) and...
218

Adheze, růst a diferenciace osteoblastů a kmenových stromálních buněk na povrchu biokompatibilních nanomateriálů / Adhesion, growth and differentiation of osteoblasts and mesenchymal stromal cells on biocompatible nanomaterial surfaces

Brož, Antonín January 2017 (has links)
The thesis is based on articles describing the fundamental research of carbon based nanomaterials for their possible utilization in biomedicine. The aim of this thesis was to describe the way how human osteoblasts (SAOS-2 cell line) and primary human mesenchymal stem cells (hMSC) adhere, grow and behave on surfaces made of several carbon allotropes - nanocrystalline diamond (NCD), single walled carbon nanotubes (SWCNTs) films and graphene. The utilization of carbon as the basic material promised good biocompatibility and possibility of useful surface modifications. The NCD had modified surface nanotopography (nanoroughness and nanostructuring prepared by dry ion etching). All the materials had modified surface atomic termination with oxygen and hydrogen which changes the surface electrical conductivity, surface charge and wettability. It was hypothesized that the surface termination can also influence the cell adhesion and growth. It turned out that all the studied materials were suitable as substrates for cultivation of mentioned cell types. Various nanoroughnesses of NCD surface had different effect on the cell adhesion and cell metabolic activity. Nanostructuring of the NCD influenced the formation of focal adhesions. The surface terminations of NCD and the other studied nanomaterials in...
219

Targeting the leukemic stem cell niche: An opportunity for novel therapeutic treatment options

Fusenig, Maximilian 09 June 2022 (has links)
Acute myeloid leukemia (AML) presents the deadliest form of blood cancer which leads to abrupt, premature deaths. Current therapeutic treatment options in AML are unspecific, resulting in high relapse rates and poor clinical responses in patients. Therapy-resistant, stem cell-like AML cells are believed to be protected by proximal stromal cells in their microenvironment, the leukemic stem cell niche. In part A of this work, an innovative first-of-its-kind arrayed endoribonuclease-prepared siRNA (esiRNA) screen was established for the targeted identification of stromal-derived, AML-supportive genes. Immortalized bone-marrow derived mesenchymal stromal cells (SCP-1) were subjected to individual esiRNA-mediated target gene knockdowns (KD) and subsequently cocultured with AML cell lines MV4-11, OCI-AML3, MOLM-13 and HL-60. AML proliferation and therapy resistance to cytostatic agents Cytarabine or Daunorubicin and tyrosine kinase inhibitor Midostaurin were assessed in direct cocultures. In SCP-1, several secreted, membrane-associated and intracellular molecules were identified which, upon esiRNA-mediated KD, resulted in proliferation inhibition and enhanced treatment response of cocultured AML cells. Carbonic anhydrase 9 (CA9), a stabilizer of intracellular pH, was identified as a supportive factor in proliferation and resistance of leukemic cells to Daunorubicin treatment whilst CA9-KD exerted only a comparably low toxicity in SCP-1 cells. Excitingly, published data by Chen and colleagues (Blood, 2017, Vol. 130, Suppl. 1, 2521) indicated an upregulation of CA9 in hypoxic ex vivo cultures of leukemic cells, measured an anti-leukemic effect of pharmacological CA9 inhibition and identified a synergistic effect on leukemic cells via combinatorial treatment of CA9-inhibition and Cytarabine under hypoxic culture conditions. Taken together, an arrayed esiRNA screen identified CA9 and other stromal-derived factors which potentially open up new avenues for selective therapeutic treatments targeting the leukemic microenvironment in AML. Currently, preclinical leukemia research relies on artificial suspension cultures of AML cells and highly sophisticated, patient-derived xenograft (PDX) mouse models that are marked by suboptimal translation of findings of PDX experiments into the clinic. Recent developments in complex three-dimensional (3D) hydrogel star-shaped poly(ethylene glycol) (starPEG)-heparin cocultures of leukemic and stromal cells of human origin showed promising results in proliferation and drug response studies. Therefore, in part B of this work, a high throughput screening (HTS)-compatible 3D hydrogel culture setup of human stromal cells was established in 384-well plates. Implementation of design of experiments (DoE) enabled an efficient, cost-effective optimization of hydrogel monocultures of human umbilical vein endothelial cells (HUVECs). Optimized culture conditions favored angiogenic sprouting of hydrogel-embedded HUVECs which responded to angiogenic inhibitors Axitinib, AZD4547 and Bevacizumab in a dose-dependent manner. A coculture with bone marrow derived MSCs altered the angiogenic network formation of endothelial CD31+ vessel-like structures. The hydrogel coculture was further stabilized by extensive hydrogel degradation and ECM deposition of MSCs. Stromal MSC networks were illustrated as highly interconnected and elongated F-Actin filament structures (CD31- F-Actin+) that were closely associating with CD31+ F-Actin+ endothelial vessel-like structures. Excitingly, the established 3D hydrogel HTS platform of primary human stromal cells enables future addition of patient-derived leukemic cells for targeted leukemic vulnerability screens in an ex vivo cell culture model of the perivascular stem cell niche. / Akute myeloische Leukämie (AML) gilt als die tödlichste Form der Blutkrebserkrankungen, welche untherapiert zum abrupten, vorzeitigen Tod führt. Etablierte therapeutische Verfahren der AML sind unspezifisch, welche durch heterogene Behandlungseffekte gekennzeichnet sind und zu hohen Rückfallquoten führen. Man vermutet, dass therapie-resistente, stammzellähnliche leukämische Zellen von proximal residierenden Stromazellen in ihrem Mikromilieu, in der sogenannten leukämischen Stammzellnische, vor therapeutischen Behandlungen geschützt werden. In Teil A dieser Arbeit wurde ein innovativer, neuartiger Screen basierend auf Endoribonuklease-generierten kleinen, interferierenden Ribonukleinsäuren (esiRNAs) für eine gezielte Identifikation von AML-supportiven, stromalen Faktoren etabliert. Immortalisierte, mesenchymale Stromazellen aus dem Knochenmark (SCP-1) wurden in einem Array mit spezifischen esiRNAs transfiziert, um esiRNA-basierende inhibierende Effekte (Knockdown) auf die Genexpression von Zielgenen in SCP-1 zu studieren und indirekte Auswirkungen auf Proliferationsrate und Therapieresistenz von kokultivierten leukämischen Zelllinien, MV4-11, OCI-AML3, MOLM-13 und HL-60, bei Behandlung mit Cytarabin, Daunorubicin und Midostaurin, zu studieren. Mehrere sezernierte, membranständige und intrazelluläre Faktoren wurden in SCP-1 identifiziert, deren esiRNA-vermittelter Knockdown zu einer Proliferationsminderung sowie verstärkten Toxizitätseffekten von applizierten Therapeutika in Leukämiezellen führten. Beispielhaft wurde Carboanhydrase (CA9), ein Enzym welches den intrazellularen pH einer Zelle stabilisert, als Target identifiziert. Ein Knockdown von CA9 in SCP-1 resultierte in einer Proliferationsminderung von kokultivierten Leukämiezellen, welche des Weiteren in einer Behandlung mit Daunorubicin verstärkt abgetötet wurden. Publizierte Daten von Chen et al. (Blood, 2017, Vol. 130, Suppl. 1, 2521) zeigten, dass CA9 in hypoxischen ex vivo Kulturen in leukämischen Zellen hochreguliert war und, dass dessen pharmakologische Inhibition einen anti-leukämischen Effekt aufwies. Zudem wurde ein synergistischer Therapieffekt, bei einer Kombinationstherapie mit einem CA9-Inhibitor und Cytarabin, auf AML Zellen in hypoxischer Zellkultur festgestellt. Zusammenfassend wurden in einem esiRNA-Screen CA9 und weitere stromal-exprimierte Faktoren identifiziert, die das Potential besitzen neuartige Therapiestrategien zu ermöglichen, welche auf die leukämische Stammzellnische als Zielstruktur ausgerichtet sind. In der präklinischen Forschung von hämatologischen Erkrankungen werden vorrangig artifizielle zweidimensionale Suspensionskulturen von Leukämiezellen verwendet oder ausgefeilte, patienten-derivierende Xenograft (PDX) Mausmodelle eingesetzt. Bedauerlicherweise weisen Erkenntnisse aus Mausmodellen eine geringe Translationseffizienz in die klinische Forschung auf. Neuste Entwicklungen mit komplexen, dreidimensionalen Hydrogelkulturen, bestehend aus sternförmigem Polyethylenglykol (starPEG) und Heparin, von stromalen und leukämischen Zellen humanen Ursprungs zeigten vielversprechende Ergebnisse in präklinischen Proliferations- und Vulnerabilitätsstudien. Daher wurde in Teil B dieser Arbeit ein hochdurchsatzfähiges dreidimensionales Kultursystem von humanen Stromazellen in Hydrogelen entwickelt. Per statistischer Versuchsplanung wurde eine effiziente, kostengünstige Optimierung von etablierten Hydrogelkulturen für die Hochdurchsatz-kompatible Kultur von humanen venösen Endothelzellen aus Nabelschnuren (HUVECs) durchgeführt. Optimierte Kulturbedingungen führten zur Angiogenese von Hydrogel-eingebetteten HUVECs, welche des Weiteren auf die Angiogenese-Inhibitoren Axitinib, AZD4547 und Bevacizumab in einer konzentrationsabhängigen Weise mit verminderter Bildung von gefäßähnlichen Strukturen reagierten. Eine Kokultur von HUVECs mit primären, mesenchymalen Stromazellen aus dem Knochenmark (MSCs) beeinflusste die Bildung von CD31+ gefäßähnlichen Strukturen. Die Hydrogel-Kokultur wurde des Weiteren durch verstärkte Degradation des Hydrogels und Deposition von Komponenten der extrazellulären Matrix via MSCs verändert und dadurch zusätzlich stabilisiert. Geformte Netzwerkstrukturen von MSCs und HUVECs wurden mittels F-Actin Färbung identifiziert, wodurch ersichtlich wurde, dass Strukturen von MSCs (CD31- F-Actin+) in enger räumlicher Distanz zu HUVEC Strukturen (CD31+ F-Actin+) gebildet wurden. Spannenderweise ermöglicht die, in dieser Arbeit etablierte, Hochdurchsatz-kompatible Kokultur von humanen Stromazellen die Möglichkeit auch leukämische Zellen in die Hydrogelmatrix einzubetten. Eine humane AML-Stroma Kokultur in Hydrogelen wird gezielte Vulnerabilitätsscreens von AML Zellen in einem komplexen ex vivo Zellkulturmodel der perivaskulären Stammzellnische ermöglichen.
220

Terapias innovadoras basadas en vesículas extracelulares derivadas de células madre mesenquimales modificadas genéticamente

Gómez Ferrer, Marta 02 May 2022 (has links)
[ES] Las células mesenquimales estromales (MSC) poseen una serie de cualidades inmunológicas, pro-angiogénicas y regenerativas que las convierten en un excelente candidato para el tratamiento de diversas patologías. A pesar de las pruebas contundentes obtenidas en modelos preclínicos que demuestran la actividad terapéutica de las MSC, los ensayos clínicos no han podido mostrar hasta ahora un beneficio consistente, probablemente debido a deficiencias metodológicas y a la falta de estandarización, así como a la variabilidad genética intrínseca a los estudios en humanos. Debido a ello, ha sido necesario profundizar en los mecanismos responsables del beneficio terapéutico y rediseñar las estrategias clínicas. En los últimos años, se ha observado que la reparación tisular mediada por las MSC se produce de forma paracrina, y se ha constatado que las vesículas extracelulares (EVs) secretadas por las MSC (EVMSC) son capaces de recapitular las propiedades inmunosupresoras de las células parentales. Además, las estrategias terapéuticas basadas en vesículas tienen grandes ventajas en términos de bioseguridad y producción en condiciones de grado clínico, reduciendo significativamente el coste de dichas terapias. Sin embargo, la dosis efectiva en grandes mamíferos, incluidos los humanos, es bastante elevada y la producción industrial de EVs se ve dificultada en parte, por la senescencia proliferativa que afecta a las MSC durante la expansión celular masiva. En este trabajo hemos intentado solventar los principales escollos de la terapia con EVs incrementando su potencial inmunosupresor y reduciendo por tanto la dosis efectiva. Este incremento se ha conseguido gracias a la sobreexpresión del factor inducible por hipoxia 1-alpha y al desarrollo de un medio de acondicionamiento en cultivo basado en citoquinas. Además, la inmortalización de las células secretoras mediante la transducción del gen de la telomerasa humana ha permitido tanto la estandarización del producto como su producción a gran escala. La eficacia de estas EVs ha sido testada en diferentes poblaciones celulares in vitro: linfocitos T, monocitos, células Natural Killer, macrófagos, células endoteliales y fibroblastos; y en dos modelos de ratón: hipersensibilidad retardada y colitis aguda inducida por TNBS. En conclusión, hemos desarrollado una fuente de EVs de larga duración que secreta grandes cantidades de vesículas con mayor capacidad inmunosupresora y antiinflamatoria, facilitando un producto terapéutico más estándar y fácil de producir para el tratamiento de enfermedades inflamatorias inmunomediadas. / [CA] Les cèl·lules mesenquimals estromals (MSC) posseeixen una sèrie de qualitats immunològiques, pro-angiogèniques i regeneratives que les converteixen en un excel·lent candidat per al tractament de diverses patologies. Tot i les proves contundents obtingudes en models preclínics que demostren l'activitat terapèutica de les MSC, els assaigs clínics no han pogut mostrar fins ara un benefici consistent, probablement degut a deficiències metodològiques i a la manca d'estandardització, així com a la variabilitat genètica intrínseca als estudis en humans. A causa d'això, ha calgut aprofundir en els mecanismes responsables del benefici terapèutic i redissenyar les estratègies clíniques. En els últims anys, s'ha observat que la reparació tissular intervinguda per les MSC es produeix de forma paracrina, i s'ha constatat que les vesícules extracel·lulars (EVs) secretades per les MSC (EVMSC) són capaços de recapitular les propietats immunosupressores de les cèl·lules parentals. A més, les estratègies terapèutiques basades en vesícules tenen grans avantatges en termes de bioseguretat i producció en condicions de grau clínic, reduint significativament el cost d'aquestes teràpies. No obstant això, la dosi efectiva en grans mamífers, inclosos els humans, és bastant elevada i la producció industrial de les EVs es veu dificultada en part, per la senescència proliferativa que afecta les MSC durant l'expansió cel·lular massiva. En aquest treball hem intentat solucionar els principals esculls de la teràpia amb EVs incrementant el seu potencial immunosupressor i reduint per tant la dosi efectiva. Aquest increment s'ha aconseguit gràcies a la sobreexpressió del factor induïble per hipòxia 1-alpha i a el desenvolupament d'un mitjà de condicionament en cultiu basat en citoquines. A més, la immortalització de les cèl·lules secretores mitjançant la transducció del gen de la telomerasa humana ha permès tant l'estandardització del producte com la seva producció a gran escala. L'eficàcia d'aquestes EVs ha estat testada en diferents poblacions cel·lulars in vitro: limfòcits T, monòcits, cèl·lules Natural Killer, macròfags, cèl·lules endotelials i fibroblasts; i en dos models de ratolí: hipersensibilitat retardada i colitis aguda induïda per TNBS. En conclusió, hem desenvolupat una font de EVs de llarga durada que secreta grans quantitats de vesícules amb major capacitat immunosupressora i antiinflamatòria, facilitant un producte terapèutic més estàndard i fàcil de produir per al tractament de malalties inflamatòries inmunomediades. / [EN] Mesenchymal stromal cells (MSC) possess several immunological, pro-angiogenic and regenerative qualities that make them an excellent candidate for the treatment of various pathologies. Despite compelling evidence from preclinical models demonstrating the therapeutic activity of MSCs, clinical trials have so far failed to show consistent benefit, probably due to methodological shortcomings and lack of standardisation, as well as the genetic variability intrinsic to human studies. As a result, it has been necessary to further investigate the mechanisms responsible for therapeutic benefit and to redesign clinical strategies. In recent years, it has been observed that MSC-mediated tissue repair occurs in a paracrine pathway, and it has been confirmed that extracellular vesicles (EVs) secreted by MSC (EVMSC) are able to recapitulate the immunosuppressive properties of the parental cells. Moreover, vesicle-based therapeutic strategies have great advantages in terms of biosafety and production under clinical-grade conditions, significantly reducing the cost of such therapies. However, the effective dose in large mammals, including humans, is quite high and the industrial production of EVs is hampered in part by the proliferative senescence that affects MSC during massive cell expansion. In this work, we have attempted to overcome the main challenges of EVs therapy by increasing their immunosuppressive potential and thus reducing the effective dose. This increase has been achieved by overexpression of hypoxia-inducible factor 1-alpha and the development of a cytokine-based culture conditioning medium. In addition, immortalization of secretory cells by transduction with the human telomerase gene has allowed both product standardisation and large-scale production. The efficacy of these EVs has been tested in different cell populations in vitro: T lymphocytes, monocytes, Natural Killer cells, macrophages, endothelial cells and fibroblasts; and in two mouse models: delayed-type hypersensitivity and TNBS-induced acute colitis. In conclusion, we have developed a long-lasting source of EVs that secretes large amounts of vesicles with enhanced immunosuppressive and anti-inflammatory capacity, providing a more standard and easier-to-produce therapeutic product for the treatment of immune-mediated inflammatory diseases. / Gómez Ferrer, M. (2022). Terapias innovadoras basadas en vesículas extracelulares derivadas de células madre mesenquimales modificadas genéticamente [Tesis doctoral]. Universitat Politècnica de València. https://doi.org/10.4995/Thesis/10251/182560 / TESIS

Page generated in 0.0544 seconds