• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 50
  • 41
  • 13
  • 8
  • 8
  • 6
  • 4
  • 3
  • 2
  • 2
  • 1
  • 1
  • Tagged with
  • 137
  • 137
  • 137
  • 44
  • 43
  • 37
  • 36
  • 29
  • 20
  • 20
  • 19
  • 19
  • 18
  • 18
  • 18
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
81

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

Melinda Elise Christensen Unknown Date (has links)
Survival of patients with poor prognosis or relapsed haematopoietic malignancies can be markedly improved by allogeneic haematopoietic stem cell transplantation (HSCT). HSCT reconstitutes the immune and haematopoietic systems after myeloablative conditioning and inhibits the recurrence of the malignancy by a graft-versus-leukaemia (GVL) response mediated by donor T cells. However, significant post-transplant complications such as graft-versus-host disease (GVHD) continue to plague the event-free survival of this curative procedure. GVHD is facilitated by donor T cells that recognise histocompatibility antigens on host antigen presenting cells (APC), such as dendritic cells (DC). Current treatment options for GVHD are focused on these T cells. However, these treatments result in an increased incidence of infection, graft rejection and relapse. A novel means of immunosuppression in GVHD is the use of multi-potent, mesenchymal stromal cells (MSC). MSC are non-immunogenic cells that actively suppress T cell function in vitro, and can resolve steroid-refractory GVHD in the clinic. Despite their use in the clinic, there is a paucity of pre-clinical data. Our aim was to investigate the in vivo efficacy of MSC to control GVHD while maintaining the beneficial GVL effect, and to begin to understand the mechanism by which MSC exert their immunosuppressive effects. We isolated and characterised MSC from murine bone/bone marrow and demonstrated that they suppressed T cell proliferation in vitro, even at low ratios of 1 MSC per 100 T cells. This was true of both donor-derived MSC, and MSC derived from unrelated donors (third party). Importantly, we observed that MSC significantly reduced T cell production of the pro-inflammatory cytokines TNFα and IFNγ in culture supernatants and that IFNγ plays a key role in the ability of MSC to suppress T cell proliferation. In vivo, we examined the effects of donor-derived MSC on GVHD severity and onset in two myeloablative murine models of HSCT. A major histocompatibility complex (MHC)-mismatched donor-recipient pair combination was used as a proof–of-principle model [UBI-GFP/BL6 (H-2b)àBALB/c (H-2d)], and an MHC-matched, minor histocompatibility antigen (miHA) mismatched donor-recipient pair combination was used to mimic MHC-matched sibling transplantation [UBI-GFP/BL6 (H-2b)àBALB.B (H-2b)]. We examined a number of variables related to MSC infusion including timing, dose and route of injection. We found that early post transplant infusion of MSC by the intraperitoneal injection was most effective at delaying death from GVHD, compared to pre-transplant infusion or intravenous injection. Furthermore, we found that the dose of MSC was critical, as infusion of too few MSC was ineffective and infusion of too many MSC exacerbated the development of GVHD. Taken together, these results suggest that timing, dose and route of injection are all important factors to be considered to ensure successful therapeutic outcome. To investigate the in vivo mechanism of action, we conducted timed sacrifice experiments in the MHC-mismatched model to determine if MSC altered cytokine secretion and cellular effectors, such as DC, known to play a key role in GVHD. Despite the fact that MSC given post-HSCT enter an environment full of activated DC and IFNγ levels, by day 3 and 6 post infusion, these activated DC and IFNγ levels are decreased compared to controls or mice infused with MSC pre-transplant (p<0.05). This confirmed our in vitro data that IFNγ played an important role in MSC-mediated immunosuppression. In addition, when we removed a major source of IFNγ production in vivo by administering the T cell depleting antibody KT3 to mice with or without MSC, we found that although T cell depletion prolonged survival, MSC were unable to further enhance this effect. This was also true when MSC were used in combination with the conventional immunosuppressant cyclosporine. Finally, we examined whether the infusion of MSC would compromise the GVL effect. We found that whilst MSC could delay the onset of GVHD, in our model they did not alter the anti-tumour effects of the donor T cells. Overall, we have shown that MSC can delay but not prevent death from GVHD when administered at an appropriate time and dose and that IFNγ is required for MSC-mediated immunosuppression in our model. These data suggest that patients undergoing HSCT should be monitored for IFNγ, and administered MSC when high levels are reached. Whilst MSC may be a promising therapy for patients with severe GVHD, we highlight that further investigation is warranted before MSC are accepted for widespread use in the clinic. The risks and benefits for transplant recipients should be carefully considered before utilising MSC to treat or prevent GVHD.
82

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

Melinda Elise Christensen Unknown Date (has links)
Survival of patients with poor prognosis or relapsed haematopoietic malignancies can be markedly improved by allogeneic haematopoietic stem cell transplantation (HSCT). HSCT reconstitutes the immune and haematopoietic systems after myeloablative conditioning and inhibits the recurrence of the malignancy by a graft-versus-leukaemia (GVL) response mediated by donor T cells. However, significant post-transplant complications such as graft-versus-host disease (GVHD) continue to plague the event-free survival of this curative procedure. GVHD is facilitated by donor T cells that recognise histocompatibility antigens on host antigen presenting cells (APC), such as dendritic cells (DC). Current treatment options for GVHD are focused on these T cells. However, these treatments result in an increased incidence of infection, graft rejection and relapse. A novel means of immunosuppression in GVHD is the use of multi-potent, mesenchymal stromal cells (MSC). MSC are non-immunogenic cells that actively suppress T cell function in vitro, and can resolve steroid-refractory GVHD in the clinic. Despite their use in the clinic, there is a paucity of pre-clinical data. Our aim was to investigate the in vivo efficacy of MSC to control GVHD while maintaining the beneficial GVL effect, and to begin to understand the mechanism by which MSC exert their immunosuppressive effects. We isolated and characterised MSC from murine bone/bone marrow and demonstrated that they suppressed T cell proliferation in vitro, even at low ratios of 1 MSC per 100 T cells. This was true of both donor-derived MSC, and MSC derived from unrelated donors (third party). Importantly, we observed that MSC significantly reduced T cell production of the pro-inflammatory cytokines TNFα and IFNγ in culture supernatants and that IFNγ plays a key role in the ability of MSC to suppress T cell proliferation. In vivo, we examined the effects of donor-derived MSC on GVHD severity and onset in two myeloablative murine models of HSCT. A major histocompatibility complex (MHC)-mismatched donor-recipient pair combination was used as a proof–of-principle model [UBI-GFP/BL6 (H-2b)àBALB/c (H-2d)], and an MHC-matched, minor histocompatibility antigen (miHA) mismatched donor-recipient pair combination was used to mimic MHC-matched sibling transplantation [UBI-GFP/BL6 (H-2b)àBALB.B (H-2b)]. We examined a number of variables related to MSC infusion including timing, dose and route of injection. We found that early post transplant infusion of MSC by the intraperitoneal injection was most effective at delaying death from GVHD, compared to pre-transplant infusion or intravenous injection. Furthermore, we found that the dose of MSC was critical, as infusion of too few MSC was ineffective and infusion of too many MSC exacerbated the development of GVHD. Taken together, these results suggest that timing, dose and route of injection are all important factors to be considered to ensure successful therapeutic outcome. To investigate the in vivo mechanism of action, we conducted timed sacrifice experiments in the MHC-mismatched model to determine if MSC altered cytokine secretion and cellular effectors, such as DC, known to play a key role in GVHD. Despite the fact that MSC given post-HSCT enter an environment full of activated DC and IFNγ levels, by day 3 and 6 post infusion, these activated DC and IFNγ levels are decreased compared to controls or mice infused with MSC pre-transplant (p<0.05). This confirmed our in vitro data that IFNγ played an important role in MSC-mediated immunosuppression. In addition, when we removed a major source of IFNγ production in vivo by administering the T cell depleting antibody KT3 to mice with or without MSC, we found that although T cell depletion prolonged survival, MSC were unable to further enhance this effect. This was also true when MSC were used in combination with the conventional immunosuppressant cyclosporine. Finally, we examined whether the infusion of MSC would compromise the GVL effect. We found that whilst MSC could delay the onset of GVHD, in our model they did not alter the anti-tumour effects of the donor T cells. Overall, we have shown that MSC can delay but not prevent death from GVHD when administered at an appropriate time and dose and that IFNγ is required for MSC-mediated immunosuppression in our model. These data suggest that patients undergoing HSCT should be monitored for IFNγ, and administered MSC when high levels are reached. Whilst MSC may be a promising therapy for patients with severe GVHD, we highlight that further investigation is warranted before MSC are accepted for widespread use in the clinic. The risks and benefits for transplant recipients should be carefully considered before utilising MSC to treat or prevent GVHD.
83

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

Melinda Elise Christensen Unknown Date (has links)
Survival of patients with poor prognosis or relapsed haematopoietic malignancies can be markedly improved by allogeneic haematopoietic stem cell transplantation (HSCT). HSCT reconstitutes the immune and haematopoietic systems after myeloablative conditioning and inhibits the recurrence of the malignancy by a graft-versus-leukaemia (GVL) response mediated by donor T cells. However, significant post-transplant complications such as graft-versus-host disease (GVHD) continue to plague the event-free survival of this curative procedure. GVHD is facilitated by donor T cells that recognise histocompatibility antigens on host antigen presenting cells (APC), such as dendritic cells (DC). Current treatment options for GVHD are focused on these T cells. However, these treatments result in an increased incidence of infection, graft rejection and relapse. A novel means of immunosuppression in GVHD is the use of multi-potent, mesenchymal stromal cells (MSC). MSC are non-immunogenic cells that actively suppress T cell function in vitro, and can resolve steroid-refractory GVHD in the clinic. Despite their use in the clinic, there is a paucity of pre-clinical data. Our aim was to investigate the in vivo efficacy of MSC to control GVHD while maintaining the beneficial GVL effect, and to begin to understand the mechanism by which MSC exert their immunosuppressive effects. We isolated and characterised MSC from murine bone/bone marrow and demonstrated that they suppressed T cell proliferation in vitro, even at low ratios of 1 MSC per 100 T cells. This was true of both donor-derived MSC, and MSC derived from unrelated donors (third party). Importantly, we observed that MSC significantly reduced T cell production of the pro-inflammatory cytokines TNFα and IFNγ in culture supernatants and that IFNγ plays a key role in the ability of MSC to suppress T cell proliferation. In vivo, we examined the effects of donor-derived MSC on GVHD severity and onset in two myeloablative murine models of HSCT. A major histocompatibility complex (MHC)-mismatched donor-recipient pair combination was used as a proof–of-principle model [UBI-GFP/BL6 (H-2b)àBALB/c (H-2d)], and an MHC-matched, minor histocompatibility antigen (miHA) mismatched donor-recipient pair combination was used to mimic MHC-matched sibling transplantation [UBI-GFP/BL6 (H-2b)àBALB.B (H-2b)]. We examined a number of variables related to MSC infusion including timing, dose and route of injection. We found that early post transplant infusion of MSC by the intraperitoneal injection was most effective at delaying death from GVHD, compared to pre-transplant infusion or intravenous injection. Furthermore, we found that the dose of MSC was critical, as infusion of too few MSC was ineffective and infusion of too many MSC exacerbated the development of GVHD. Taken together, these results suggest that timing, dose and route of injection are all important factors to be considered to ensure successful therapeutic outcome. To investigate the in vivo mechanism of action, we conducted timed sacrifice experiments in the MHC-mismatched model to determine if MSC altered cytokine secretion and cellular effectors, such as DC, known to play a key role in GVHD. Despite the fact that MSC given post-HSCT enter an environment full of activated DC and IFNγ levels, by day 3 and 6 post infusion, these activated DC and IFNγ levels are decreased compared to controls or mice infused with MSC pre-transplant (p<0.05). This confirmed our in vitro data that IFNγ played an important role in MSC-mediated immunosuppression. In addition, when we removed a major source of IFNγ production in vivo by administering the T cell depleting antibody KT3 to mice with or without MSC, we found that although T cell depletion prolonged survival, MSC were unable to further enhance this effect. This was also true when MSC were used in combination with the conventional immunosuppressant cyclosporine. Finally, we examined whether the infusion of MSC would compromise the GVL effect. We found that whilst MSC could delay the onset of GVHD, in our model they did not alter the anti-tumour effects of the donor T cells. Overall, we have shown that MSC can delay but not prevent death from GVHD when administered at an appropriate time and dose and that IFNγ is required for MSC-mediated immunosuppression in our model. These data suggest that patients undergoing HSCT should be monitored for IFNγ, and administered MSC when high levels are reached. Whilst MSC may be a promising therapy for patients with severe GVHD, we highlight that further investigation is warranted before MSC are accepted for widespread use in the clinic. The risks and benefits for transplant recipients should be carefully considered before utilising MSC to treat or prevent GVHD.
84

Comment deux lignées cellulaires stromales mésenchymateuses humaines récapitulent in vitro le microenvironnement hématopoïétique ? : Intérêt en ingénierie / No title available

Ishac, Nicole 01 July 2015 (has links)
L’hématopoïèse se déroule dans un microenvironnement spécialisé appelé niche où les cellules souches hématopoïétiques (CSH) sont en contact étroit avec les cellules stromales mésenchymateuses. Cette interaction cellulaire associée à d’autres facteurs environnementaux, comme la présence des espèces réactives à l’oxygène, est cruciale pour la régulation des CSH normales, mais aussi leucémiques. Pour étudier ce microenvironnement, il est donc important de développer un modèle in vitro de niche humaine qui mime la physiologie in vivo. Nous avons choisi comme modèle deux lignées mésenchymateuses stromales humaines HS-27a et HS-5, très peu décrites dans la littérature. Le premier objectif a été de déterminer la qualité de cette niche tant du point de vue cellulaire, moléculaire que fonctionnel. Nos résultats montrent clairement que les cellules HS-27a participent à la formation d’une niche « quiescente » alors que les cellules HS-5 représentent une niche « proliférative ». Le deuxième objectif a été de créer une niche contrôlée pour le métabolisme oxydatif en régulant l’expression d’une protéine antioxydante, la glutathion peroxydase 3 ou GPx3. L’originalité de ce travail repose sur l’utilisation d’une méthode non virale de transfert de gène par le transposon piggyBac. Le plasmide porteur du gène d'intérêt a été apporté sous forme d’ADN et une source de transposase, enzyme catalysant la réaction d'intégration sous forme d’ARNm. Notre travail montre que GPx3 est un régulateur clé de l’homéostasie hématopoïétique favorisant le maintien des progéniteurs immatures. Pour la première fois, nous créons par ingénierie in vitro une niche hématopoïétique « calibrée » capable de mimer le microenvironnement normal et leucémique. Ce modèle permet non seulement d’identifier les acteurs clés de la régulation des cellules médullaires, mais aussi de développer des stratégies thérapeutiques ciblées. / Hematopoiesis occurs in a hypoxic microenvironment or niche in which hematopoietic stem cells (HSCs) are in close contact with mesenchymal stromal cells. Cellular interactions as well as microenvironmental factors such as reactive oxygen species are crucial for the maintenance of normal and leukemic HSCs. Developing an in vitro human culture system that closely mimcs marrow physiology is therefore essential to study the niche. Here, we present a model using two human stromal cell lines, HS-27a and HS-5. Previously poorly described in the literature, we have further characterized both of these cell lines. The first objective was to assess the quality of HS-27a and HS-5 niches by investigating their cellular, molecular and functional characteristics. Our results clearly show that HS-27a cells display features of a “quiescent” niche whereas HS-5 cells rather represent a “proliferative” niche. The second objective was to engineer a hematopoietic niche where the oxidative metabolism is optimized for the expression of an antioxidant protein, glutathione peroxidase 3 (GPx3). The originality of this work is the use of a non-viral gene transfer system by using the transposon piggyBac. This strategy was achieved by delivering a DNA plasmid carrying the gene of interest, and an mRNA source of transposase, the enzyme which catalyzes the transgene integration. Functionally, GPx3 was shown to be a key regulator for sustaining hematopoietic homeostasis by maintaining immature progenitor cells. For the first time, an original non-viral gene transfer has been used to create an in vitro hematopoietic niche that recapitulates the complexity of normal and leukemic microenvironment. This niche not only provides a platform to identify regulatory factors controlling medullary cells, but may also help in the development of targeted therapeutic strategies.
85

Estudo sobre condições do cultivo de células-tronco mesenquimais para aplicações clínicas

Valim, Vanessa de Souza January 2012 (has links)
Introdução: Células-troco mesenquimais (CTM) vêm mostrando seus benefícios na doença do enxerto-versus-hospedeiro (DECH), observada no transplante de células tronco hematopoéticas (TCTH), existem três questões em aberto: (1) Expansão de CTM em meio de cultura suplementado com soro fetal bovino (SFB), pelo o risco de xenorreação; (2) Otimização de condições de cultura para a obtenção, em tempo hábil, de um numero que permita de 4 a 6 infusões de 2x106cells/kg do receptor; (3) Obter células do doador de medula óssea, evitando assim a utilização de um terceiro doador. Objetivos: Este estudo foi desenhado para comparar o lisado de plaquetas (LP) e o SFB na expansão de CTM, a densidade de plaqueamento das células e os dias entre cada passagem, e para investigar se as células nucleadas totais obtidas da bolsa e filtro do TCTH, podem ser utilizadas para expansão de CTM para utilização clínica. Métodos: Células residuais foram removidas do filtro e da bolsa utilizados para o TCTH, plaqueadas e depois da primeira passagem foram cultivadas em diferentes concentrações com SFB ou LP e observado o número de dias que levaram para chegar a 80% de confluência. Em seguida, as culturas com as mesmas densidades de plaqueamento foram suplementadas com LP ou SFB e depois de sete dias contou-se o número de células para analisar o quanto elas cresceram nesse período. Resultados: A proliferação de CTM, na presença de LP e SFB foi em média 11,88 e 2,5 vezes, respectivamente, num período de 7 dias. A concentração mais elevada de células usando LP demorou menos tempo para atingir a confluência, em comparação com os três inferiores. Este estudo sugere que o LP é a melhor escolha como suplemento para expandir CTM, e permite a proliferação de um número suficiente de CTM de doadores para uso clínico. / Introduction: Mesenchymal stromal cells (MSC) have shown their benefits in graft-versus-host disease (GVHD), with three unsettled matters:(1) MSCs expansion in medium with Fetal Calf Serum (FCS) and its risk of xenoreaction; (2) The number of cells indicated for therapy is 2x106cells/Kg with the need to optimize expansion, number and time wise; and (3) the utilization of third party donors. Aims: This study was designed to compare the platelet lysate (LP) and FCS on the expansion of MSC, the optimal cell plating density and days between each pass, and to investigate if donor total nucleated cells (TNC) obtained from the washouts of hematopoietic stem cell transplantation (HSCT) explants can be expanded to be used at clinical grade. Methods: TNC were removed, plated and after the first passage were cultivated in different concentrations with FCS or PL and the number of days reach 80% of confluence was observed. Next, cultures with the same plating density were fed either with PL or FCS and after seven days counted to analyze how much they have grown in that period. Results: The proliferation of mesenchymal stromal cells in the presence of PL and SFB was averaged 11.88 and 2.5 times, respectively, in a period of 7 days. The highest concentration of plating cells using PL, took less time to reach confluence as compared with the three lower ones. This study suggests that the PL is the best choice as a supplement to expand MSC, and allows the proliferation of a sufficient number of donors MSC at P2 for clinical use.
86

Avaliação do potencial imunomodulador de células-tronco mesenquimais isoladas a partir de polpa dental, tecido adiposo e medula óssea

Rodrigues, Felipe Valle Fortes January 2015 (has links)
Introdução: Células tronco mesenquimais (CTM) são uma população residente nos tecidos adultos de origem mesodérmica, com funções regenerativas de manutenção da integridade tecidual, com destaque no desempenho imunomodulador. Esse aspecto levou as CTM a tornarem-se ferramentas terapêuticas valiosas da pesquisa à assistência ao paciente em doenças autoimunes e de cunho inflamatório. Além disso, CTM podem ser isoladas de materiais tidos como descarte de procedimentos, como dentes decíduos, filtros de transplante de medula óssea e gordura. Nesse panorama, torna-se necessário estabelecer o efeito que a origem tecidual tem na eficiência imunoreguladora e na possível aplicabilidade clínica destas células. Objetivo: Comparar o potencial imunomodulador de células mesenquimais isoladas a partir de filtros descartados após a infusão de medula óssea, de lipoaspirado e de polpa de dentes decíduos. Métodos: Foi realizada a comparação da capacidade proliferativa de CTMs, cultivadas na presença de lisado plaquetário, das diversas fontes através do cálculo de population doubling das CTM em co-cultura com linfócitos T isolados em coluna magnética e com células mononucleares de sangue periférico, estimuladas com fitohemaglutinina; e determinado por citometria de fluxo o efeito das CTM das diversas fontes sobre as subpopulações linfocitárias. Resultados: CTM das três fontes foram capazes de inibir a proliferação de linfócitos e CTM de tecido adiposo foram mais eficientes em induzir o fenótipo de células T reguladoras e na diminuição de células T citotóxicas. Conclusão: comparadas à CTM isoladas de medula óssea e de polpa dentária, as CTM originadas de tecido adiposo exibem efeito imunomodulador mais acentuado. / Background: Mesenchymla stromal cells (MSC) reside in most adult tissue of mesenchymal origen, with a broad functions envolving cell repopulation and maintenence of tissue homeostasis, trough immunemmodulatory action. MSC are valuable terapêutic instruments applied from research to autoimune and inflamatory diseases. MSC can be isolated from diverse discarted biological matherials, like lipoaspirate, exfoliated deciduous teeeth and boné marrow ransplant filters. There so it´s necessary to stablish how source can impact MSC efficiency and possible clinical aplications. Objective: Compare immunomodulatory potential of adipose MSC and dental pulp MSC to boné marrow MSC. Methods: MSC from three selected sources were cocultured with phytohemaglutinin stimulated and magnetically isolated T cells and peripheral blood mononuclear cells; immunephenotype of cocultivated lymphocytes were also conducted. Results: MSC from all analyzed sources were capable to inhibit lymphocyte proliferation. Adipose MSC were capable to induce Treg phenotype and decrease T CD8+ limphocytes. Conclusion: Cell culture and therapy with MSC present many paradigms and we address to some of those to elucidate the possible most efficient source.
87

Effets des cellules stromales mésenchymateuses dans la chimiorésistance des cancers ovariens par sécrétion de chimiokines et polarisation des macrophages / Protumoral effect of mesenchymal stromal cells in ovarian cancer by cytokine secretions and activation of the innate immune response

Le Naour, Augustin 27 October 2017 (has links)
Le cancer ovarien est la 1ère cause de décès par cancer gynécologique. A ce jour, le traitement le plus efficace consiste en une exérèse la plus complète possible de la tumeur associée à une chimiothérapie à base de sel de platine et de taxanes. Cependant, la survie globale des patientes à 5 ans reste faible (environ 40%), due à un fort taux de récidive et au développement d'une résistance aux traitements. Les interactions entre les cellules tumorales ovariennes (CTO) et le microenvironnement semblent être impliquées dans la progression de la maladie et l'acquisition de cette chimiorésistance. Parmi les cellules du microenvironnement tumoral, nous avons pu isoler des cellules stromales mésenchymateuses (MSC) à partir de biopsies de carcinome ovarien. Ces MSC associées au cancer (CA-MSC) ont la capacité d'induire une résistance des CTO au traitement par carboplatine. Afin de mieux comprendre les mécanismes conduisant des MSC à sécréter des facteurs pro-tumoraux, dans un contexte de cancer ovarien, nous avons mis en place un modèle à partir de MSC issues de donneurs ne présentant pas de cancer. Nous avons observé que la présence d'un environnement tumoral ovarien modifie le phénotype de MSC de moelle osseuse (BM-MSC) physiologiques, en entrainant notamment la sécrétion par ces CA-MSC " induites " de facteurs chimioprotecteurs pour les CTO. Parmi les facteurs sécrétés, nous avons identifié des chimiokines telles que CXCL1, CXCL2 et IL-8 comme des cibles thérapeutiques intéressantes pour lutter contre la résistance à la chimiothérapie. En effet, les CA-MSC et les CA-MSC " induites " sécrètent plus de CXCL1, CXCL2 et d'IL-8 que les BM-MSC. En outre, l'inhibition des récepteurs de ces chimiokines (CXCR1 et CXCR2) sensibilise les CTO au carboplatine, même en présence des sécrétions des CA-MSC et des CA-MSC " induites ". Ces expériences in vitro ont été confirmées in vivo dans un modèle expérimental murin. En effet, la co-injection à des souris immunodéprimées de MSC avec des CTO humaines entraine une protection des CTO vis-à-vis du carboplatine comparée à l'injection de CTO seules. Le co-traitement avec un inhibiteur de CXCR1 et de CXCR2 a permis de sensibiliser les CTO au carboplatine et d'empêcher la chimiorésistance induite par les MSC. En effectuant une étude rétrospective évaluant la concentration de ces chimiokines au moment du diagnostic, nous avons confirmé que les patientes qui seront a posteriori " résistantes " au carboplatine ont une concentration sérique de ces chimiokines qui est plus élevées que les patientes du groupe " sensibles " au carboplatine. Outre leur rôle direct dans l'acquisition de la chimiorésistance, les chimiokines comme CXCL1, CXCL2 et IL-8 peuvent être impliquées dans la régulation du système immunitaire. Nous avons montré que les CA-MSC étaient capables de modifier le phénotype des macrophages vers un phénotype M2 décrit comme pro-tumoral. En effet, ces macrophages ainsi polarisés ont un pouvoir cytotoxique dirigé contre les CTO inférieur aux macrophages non stimulés. L'ajout d'un inhibiteur de CXCR1 et de CXCR2 restaure le pouvoir cytotoxique initial des macrophages, même en présence des sécrétions des CA-MSC. Ainsi nos travaux suggèrent que les CA-MSC pourraient provenir de MSC physiologiques qui, au contact d'un environnement tumoral ovarien, vont acquérir un phénotype capable d'induire la sécrétion de facteurs chimioprotecteurs pour les CTO et de polariser les macrophages vers un phénotype moins cytotoxique pour les CTO. Ces deux phénomènes pro-tumoraux peuvent être inhibés par l'utilisation d'un inhibiteur des récepteurs de CXCR1 et de CXCR2. Ainsi ces récepteurs des chimiokines, semblent être des cibles thérapeutiques intéressantes afin de sensibiliser les CTO au carboplatine et traiter plus efficacement la tumeur. Ceci pourrait permettre d'éviter les récidives des cancers ovariens qui sont, à l'heure actuelle, observées chez plus de 70% des patientes. / Ovarian cancer is the leading cause of gynecological cancer death. To date, the most effective treatment consists of the complete excision of the tumor associated with chemotherapy based on platinum salts and taxanes. However, the 5-year overall survival remains low (close to 40%) due to a high rate of recurrence and development of resistance to treatments. Disease progression and the acquisition of this chemoresistance seem to be due to interactions between ovarian tumor cells (OTC) and the microenvironment. Amidst the cells of the tumor microenvironment, we were able to isolate mesenchymal stromal cells (MSC) from tumor biopsies of patients with ovarian adenocarcinoma. These cancer-associated MSC (CA-MSC) have the ability to induce resistance to carboplatin in OTC. In order to understand the mechanisms leading to the secretion of pro-tumoral factors by the CA-MSC in the context of ovarian cancer, we have developed a model based on the in vitro MSC culture of from healthy donors in tumor conditioning media. We have observed that an ovarian tumor environment modifies the physiological phenotype of bone marrow MSC (BM-MSC), leading in particular to the secretion by these "induced" CA-MSC of chemoprotective factors for OTC. Among these secreted factors, we have identified chemokines such as CXCL1, CXCL2 and IL-8 as therapeutic targets in order to control drug resistance. In fact, CA-MSC and "induced" CA-MSC secrete more CXCL1, CXCL2 and IL-8 than BM-MSC and the use of an inhibitor of their receptors (CXCR1 and CXCR2) sensitized OTC to carboplatin even in the presence of CA-MSC and " induced " CA-MSC secretions. These in vitro experiments have been confirmed in an experimental mouse model in vivo. Indeed, the co-injection of MSC with OTC yielded a greater protection of OTC to carboplatin compared with the OTC injection alone. Co-treatment with a CXCR1 and CXCR2 inhibitor resulted in sensitization of OTC to carboplatin and prevention of MSC-induced chemoresistance. We conducted a retrospective study evaluating the concentration of these chemokines at the time of diagnosis. We thus showed that patients who are a posteriori "resistant" to carboplatin have a higher concentration of chemokines than patients belong to the "sensitive" group to carboplatin. In addition to their direct role concerning the acquisition of chemoresistance, chemokines such as CXCL1, CXCL2 and IL-8 may be involved in the immune system regulation. In this context, we showed that CA-MSC were able to modify the phenotype of macrophages into a M2 phenotype described in literature to have a pro-tumoral activity. Indeed, these polarized macrophages present a lower cytotoxic capacity against OTC than unstimulated macrophages. CXCR1 and CXCR2 inhibitor restores the initial cytotoxic activity of macrophages even in the presence of CA-MSC secretions. Thus, our work suggests that CA-MSC could originate from physiological MSC which, in contact with an ovarian tumor environment, acquire a phenotype capable of inducing the secretion of chemoprotective factors for CTO and of polarizing macrophages into a less cytotoxic phenotype for OTC. These two pro-tumoral mechanisms can be inhibited by the use of CXCR1 and CXCR2 receptor inhibitors emphasizing the role of these chemokines in the development of a chemoresistance and showing how important is to go further is this study. Finally, these chemokines receptors seem to be therapeutic targets in order to sensitize OTC to carboplatin and to potentialize actual treatments. This could prevent the recurrence of ovarian cancers that are presently observed in more than 70% of patients.
88

Avaliação da expansão de células estromais mesenquimais em biorreator de fibra oca

Santos, Diogo Peres dos 28 February 2013 (has links)
Made available in DSpace on 2016-06-02T19:56:51Z (GMT). No. of bitstreams: 1 5178.pdf: 2466586 bytes, checksum: be50519a129b12383d84e69ae25b54db (MD5) Previous issue date: 2013-02-28 / Universidade Federal de Sao Carlos / The use of mesenchymal stromal cells (MSCs) for clinical therapy has been limited by the low amount of cells that can be obtained directly from tissue, making it necessary to develop techniques for in vitro cell number expansion. The current methods of expansion are laborintensive, exhibit unfavorable environments for cell growth, show still modest levels of expansion and low yield in the recovery of these cells. In the search for better alternatives, several types of bioreactors have been assessed, however, with results still discreet. A littlestudied system, which has showed itself very effective in the use with other types of animal cells, is the hollow fiber bioreactor. This bioreactor has relatively homogeneous culture environment, low level of hydrodynamic stress on cells and the process control is made through manipulation external to the culture. Thus, it is proposed in this work the study of the in vitro expansion of MSCs in 15 mL hollow fiber prototype bioreactor designed and built with a configuration specifically conceived for expansion of MSCs for use in therapeutic applications. The inoculum was prepared with MSCs precultured adhered to microcarrier Cultispher-S at concentration of 4 g/L in spinner flask containing 50 mL of &#945;-MEM culture medium with 15% v/v fetal bovine serum. The preculture was performed in CO2 incubator at pH close to 7.3 and temperature of 37°C. For bioreactor expansion cultures, it was used the same culture medium, with addition of 12 g/L of alginate and 4.25-4.50 mM of CaCl2 as gelling agents to immobilize and keep in suspension the microcarriers, in the conditions of pH and temperature used in the preculture. The oxygenation of the culture medium continuously recirculated through the intracapilar space was carried out by air bubbling in an external flask. The oxygenation levels were of 70 to 90% of saturation with air. The experimental results obtained show that the used configuration of hollow fiber bioreactor promoted good conditions for expansion of MSCs without cell aggregation, reaching 15.3-fold expansion and cell recovery levels of 82%. These results also demonstrate the possibility of improving the efficiency of MSCs expansion through the renewal of medium to maintain suitable levels of arginine, nutrient present in limiting amounts, and ammonium, growth inhibitor metabolite. / A utilização de células estromais mesenquimais (MSCs em inglês) para a terapia clínica tem sido limitada pela baixa quantidade de células que podem ser obtidas diretamente do tecido, tornando necessário o desenvolvimento de técnicas de expansão do número de células in vitro. Os métodos atuais de expansão apresentam necessidade de intensa mão de obra, ambientes desfavoráveis para o crescimento celular, níveis de expansão ainda modestos e baixo rendimento na recuperação destas células. Na procura de melhores alternativas, diversos tipos de biorreatores vêm sendo avaliados, porém, com resultados ainda discretos. Um sistema pouco estudado que tem se mostrado muito eficiente no uso com outros tipos de células animais é o biorreator de fibra oca. Este biorreator apresenta ambiente de cultura relativamente homogêneo, baixo nível de forças hidrodinâmicas sobre as células e o controle do processo é feito através de manipulação externa à cultura. Assim, é proposto neste trabalho o estudo da expansão in vitro de MSCs num protótipo de biorreator de fibra oca de 15 mL projetado e construído com uma configuração especialmente concebida para expansão de MSCs a serem utilizadas em aplicações terapêuticas. O inóculo foi preparado com MSCs précultivadas aderidas ao microcarregador Cultispher-S na concentração de 4 g/L em frasco spinner contendo 50 mL de meio de cultura &#945;-MEM com 15% v/v de soro fetal bovino. O précultivo foi realizado em incubadora de CO2 a pH próximo a 7,3 e temperatura de 37°C. Para os cultivos de expansão no biorreator foi utilizado o mesmo meio de cultura, com adição de 12 g/L de alginato e 4,25-4,50 mM de CaCl2 como agentes geleificantes para imobilizar e manter suspensos os microcarregadores, nas condições de pH e temperatura utilizadas no précultivo. A oxigenação do meio de cultura continuamente recirculado pelo espaço intracapilar foi realizada mediante borbulhamento de ar em um frasco externo. Os níveis de oxigenação foram de 70 a 90% da saturação com ar. Os resultados experimentais obtidos mostram que a configuração utilizada propiciou boas condições para a expansão sem agregação celular das MSCs, chegando-se a fatores de expansão estimados de 15,3 vezes e níveis de recuperação de células de 82%. Esses resultados também evidenciam a possibilidade de melhora da eficiência da expansão das MSCs através da renovação do meio de cultivo para a manutenção de níveis adequados de arginina, nutriente presente em quantidades limitantes, e amônia, metabólito inibidor de crescimento.
89

Biorreator wave como alternativa para expansão de células estromais mesenquimais

Silva, Juliana de Sá da 05 March 2015 (has links)
Made available in DSpace on 2016-06-02T19:56:57Z (GMT). No. of bitstreams: 1 6615.pdf: 4694610 bytes, checksum: 288df2441cd04d5e5e4c36da49c66cb9 (MD5) Previous issue date: 2015-03-05 / Financiadora de Estudos e Projetos / Mesenchymal stromal cells (MSCs) are required by the scientific community in the development and enhancement of therapeutic techniques in different fields of medicine. The MSCs are present in small concentrations in tissues, which makes necessary the expansion in vitro for enable studies and therapeutic applicability. These are cells with high sensitivity to environmental conditions of cultivation. So, for increase productivity in vitro is used the technology of bioreactors in the development of processes in order to produce high cell densities in less time, with reduce use of resources and maintaining a safe operation. The new concepts of "disposable bioreactors", as the wave-induced motion bioreactor or Wave bioreactor, with possibility operating in a closed system, controlled and automated, reduced investment cost and operation, less risk of contamination, higher level biosecurity, added to the fact of being a underexplored technology and already approved by the FDA (Food and Drugs Administration) becomes a highly attractive alternative bioprocessing for cultivation of animal cells in large scale. In this context, the present work aims to develop a protocol for cultivation of MSCs in the Wave Bioreactor System 2/10. Experiments were performed to characterize the CEMs's culture behavior in the Wave bioreactor to obtain high cell productivity while maintaining the therapeutic potential of the CEMs. The experiments were carried out with 2 L Cellbag and Cultispher-S microcarrier with 300 ml of &#945;-MEM medium culture supplemented with glucose, glutamine, and arginine and 15% v/v fetal bovine serum at 37 ° C and pH between 6,9-7,4. In the preliminary experiments it was verified that most of the inoculated cells did not adhere to the microcarriers. It was shown that such behavior is due to low relation between adhesion area (AMC = total projected area of the microcarriers) and wet surface area of Cellbag (ASMCellbag), which in the normal condition of operation results an adhesion between 25,7 and 61,7% of the inoculated cells. To solve the problem were performed experiments reducing ACellbag which enabled improvements in cell adhesion by up to 100%. It was also found low performance of the cell expansion phase, presumably linked to operational problems like: microcarriers segregation in certain regions of the bioreactor causing depletion of nutrients, formation of aggregates of MCs colonized with cells and adhesion of MCs to Cellbag. In addition, it was observed that reducing CEM/MC ratio at the start of the culture, the cell expansion factor could be increased to values equal to or greater than 10. These results show that the Wave bioreactor has good potential for expansion of MSCs and that the same can be improved. / As células estromais mesenquimais (CEMs) estão sendo visadas pela comunidade científica no desenvolvimento e aprimoramento de técnicas terapêuticas em diferentes ramos da medicina. As CEMs estão presentes em pequenas concentrações nos tecidos, o que torna necessário a sua expansão in vitro para viabilizar pesquisas e a aplicabilidade terapêutica. Tratam-se de células com elevada sensibilidade em relação às condições do ambiente de cult ivo. Assim, para o aumento da produtividade in vitro utiliza-se a tecnologia de biorreatores no desenvolvimento de processos com objetivo de produzir altas densidades celulares em curto tempo, de forma econômica e respeitando as normas impostas pelos órgãos reguladores. O novo conceito de biorreator descartável, como o do biorreator com movimento induzido em forma de ondas, ou biorreator Wave, apresenta possibilidade de operação em sistema fechado segundo as boas práticas de fabricação (BPF), controlado e automatizado. O custo de investimento e operação reduzido, com menor risco de contaminação, maior nível de biossegurança, somado ao fato de utilizar uma tecnologia pouco explorada e já aprovada pelo FDA (Food and Drugs Administration) se transforma numa alternativa de bioprocessamento altamente atrativa para cultivo de células animais em larga escala. Nesse contexto, o presente trabalho tem por meta avaliar o desempenho do biorreator Wave 2/10 na expansão das CEMs. Para tal, foram realizados experimentos visando caracterizar o comportamento do cultivo nesse biorreator a fim de obter alta produtividade celular mantendo a potencialidade terapêutica das CEMs. Os experimentos foram realizados com saco plástico (doravante Cellbag) de 2 L e microcarregador (MC) Cultispher-S com 300 mL me io de cultivo &#945;-MEM suplementado com glicose, glutamina e arginina e 15% v/v de soro fetal bovino a 37°C e pH entre 6,9-7,4. Nos experimentos preliminares constatou-se que grande parte das células inoculadas não aderiam aos microcarregadores. Comprovou-se que tal comportamento se devia à baixa relação entre área de adesão (AMC = área total projetada dos microcarregadores) e área de superfície molhada da Cellbag (ASMCellbag) que na condição normal de operação resultava numa adesão entre 25,7 e 61,7% das células inoculadas. Para melhorar a adesão foram realizados experimentos reduzindo a ASMCellbag, o que possibilitou melhoria na adesão celular em até 100%. Na etapa de expansão celular verificou-se baixo desempenho, presumivelmente vinculado a problemas de operação como: segregação de microcarregadores em determinadas regiões do biorreator provocando o esgotamento de nutrientes, formação de agregados de MCs colonizados com células e adesão dos MCs à Cellbag. Em adição, notou-se que diminuindo a relação CEM/MC no início do cultivo a expansão celular podia ser aumentada para valores iguais ou maiores que 10. Ao todo, os resultados mostraram que o biorreator Wave possui bom potencial para a expansão de CEMs e que o mesmo ainda pode ser melhorado.
90

Estudo sobre condições do cultivo de células-tronco mesenquimais para aplicações clínicas

Valim, Vanessa de Souza January 2012 (has links)
Introdução: Células-troco mesenquimais (CTM) vêm mostrando seus benefícios na doença do enxerto-versus-hospedeiro (DECH), observada no transplante de células tronco hematopoéticas (TCTH), existem três questões em aberto: (1) Expansão de CTM em meio de cultura suplementado com soro fetal bovino (SFB), pelo o risco de xenorreação; (2) Otimização de condições de cultura para a obtenção, em tempo hábil, de um numero que permita de 4 a 6 infusões de 2x106cells/kg do receptor; (3) Obter células do doador de medula óssea, evitando assim a utilização de um terceiro doador. Objetivos: Este estudo foi desenhado para comparar o lisado de plaquetas (LP) e o SFB na expansão de CTM, a densidade de plaqueamento das células e os dias entre cada passagem, e para investigar se as células nucleadas totais obtidas da bolsa e filtro do TCTH, podem ser utilizadas para expansão de CTM para utilização clínica. Métodos: Células residuais foram removidas do filtro e da bolsa utilizados para o TCTH, plaqueadas e depois da primeira passagem foram cultivadas em diferentes concentrações com SFB ou LP e observado o número de dias que levaram para chegar a 80% de confluência. Em seguida, as culturas com as mesmas densidades de plaqueamento foram suplementadas com LP ou SFB e depois de sete dias contou-se o número de células para analisar o quanto elas cresceram nesse período. Resultados: A proliferação de CTM, na presença de LP e SFB foi em média 11,88 e 2,5 vezes, respectivamente, num período de 7 dias. A concentração mais elevada de células usando LP demorou menos tempo para atingir a confluência, em comparação com os três inferiores. Este estudo sugere que o LP é a melhor escolha como suplemento para expandir CTM, e permite a proliferação de um número suficiente de CTM de doadores para uso clínico. / Introduction: Mesenchymal stromal cells (MSC) have shown their benefits in graft-versus-host disease (GVHD), with three unsettled matters:(1) MSCs expansion in medium with Fetal Calf Serum (FCS) and its risk of xenoreaction; (2) The number of cells indicated for therapy is 2x106cells/Kg with the need to optimize expansion, number and time wise; and (3) the utilization of third party donors. Aims: This study was designed to compare the platelet lysate (LP) and FCS on the expansion of MSC, the optimal cell plating density and days between each pass, and to investigate if donor total nucleated cells (TNC) obtained from the washouts of hematopoietic stem cell transplantation (HSCT) explants can be expanded to be used at clinical grade. Methods: TNC were removed, plated and after the first passage were cultivated in different concentrations with FCS or PL and the number of days reach 80% of confluence was observed. Next, cultures with the same plating density were fed either with PL or FCS and after seven days counted to analyze how much they have grown in that period. Results: The proliferation of mesenchymal stromal cells in the presence of PL and SFB was averaged 11.88 and 2.5 times, respectively, in a period of 7 days. The highest concentration of plating cells using PL, took less time to reach confluence as compared with the three lower ones. This study suggests that the PL is the best choice as a supplement to expand MSC, and allows the proliferation of a sufficient number of donors MSC at P2 for clinical use.

Page generated in 0.0923 seconds