• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 3
  • 3
  • 1
  • 1
  • 1
  • Tagged with
  • 9
  • 9
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 3
  • 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.
1

MFG-E8 Blockade Enhances Tumor Immunity in a Murine Breast Cancer Model

Draganov, Dobrin Draganov January 2012 (has links)
Milk fat globule - epidermal growth factor - factor 8 protein (MFG-E8) is an important mediator of the tolerogenic functions of GM-CSF, and a dominant-negative RGE mutant augments the therapeutic potential of irradiated, GM-CSF-secreting tumor vaccines (GVAX) in the MFG-E8-negative B16 melanoma model. The frequent expression of MFG-E8 in various solid and hematological malignancies, however, prompted us to investigate the effect of the RGE mutant in a MFG-E8-positive transplantable breast tumor model. Here, we report that MFG-E8 blockade augmented anti-tumor humoral responses and modulated immune infiltrates at vaccination sites, which was associated with defective phagocytosis and clearance of apoptotic tumor cells. The RGE mutant enhanced the therapeutic potential of two irradiated, GM-CSF-secreting vaccines and improved protection correlated with augmented tumor-specific IgG1 and IgG2a antibody responses as well as increased ratios of T effectors to Tregs in TILs. These findings are consistent with the notion that MFG-E8 blockade potentiates anti-tumor responses through the preferential expansion of effector over regulatory T cells. Our data also validate the use of the RGE mutant to achieve therapeutically effective MFG-E8 blockade even in the context of tumors and vaccines that express high levels of endogenous MFG-E8.
2

Immune Modulation Potential of ESC Extracts on T Cells

AlKhamees, Bodour Abdullah 30 August 2012 (has links)
Embryonic stem cells (ESCs) possess hypo-immunogenic properties and have the capacity to modulate allogeneic immune response. ESCs have been shown to reduce immune activation in response to third party antigen presenting cells (APCs) in vitro and have the capacity to promote allograft survival in vivo. Clinical use of live ESCs to treat immunological disorders, however, risks teratoma or ectopic tissue formation. Accordingly, the way lab is studying the immune modulatory potentials of ESC-derived factors and recently, found that dendritic cells (DCs) treated with human ESC extracts are poor stimulators of purified allogeneic T cells compared to those DCs treated with vehicle or fibroblast extracts. In the present study, I found that ESC-derived extracts directly inhibit T cell proliferation and suppress their activation without inducing cell death. Furthermore, ESC extracts are able to suppress Th1 polarization while increasing the numbers of Foxp3+ CD4+ CD25+ regulatory T cells. Moreover, I found that a protein called Milk fat globule-EGF factor 8 (MFG-E8) appears to be highly expressed in ESCs. Importantly, neutralizing MFG-E8 substantially abrogated the immune suppressive effects of ESC extracts on T cell activation. These findings lead to future studies to further define specific immunomodulatory factors derived from ESCs for potential applications.
3

Immune Modulation Potential of ESC Extracts on T Cells

AlKhamees, Bodour Abdullah January 2012 (has links)
Embryonic stem cells (ESCs) possess hypo-immunogenic properties and have the capacity to modulate allogeneic immune response. ESCs have been shown to reduce immune activation in response to third party antigen presenting cells (APCs) in vitro and have the capacity to promote allograft survival in vivo. Clinical use of live ESCs to treat immunological disorders, however, risks teratoma or ectopic tissue formation. Accordingly, the way lab is studying the immune modulatory potentials of ESC-derived factors and recently, found that dendritic cells (DCs) treated with human ESC extracts are poor stimulators of purified allogeneic T cells compared to those DCs treated with vehicle or fibroblast extracts. In the present study, I found that ESC-derived extracts directly inhibit T cell proliferation and suppress their activation without inducing cell death. Furthermore, ESC extracts are able to suppress Th1 polarization while increasing the numbers of Foxp3+ CD4+ CD25+ regulatory T cells. Moreover, I found that a protein called Milk fat globule-EGF factor 8 (MFG-E8) appears to be highly expressed in ESCs. Importantly, neutralizing MFG-E8 substantially abrogated the immune suppressive effects of ESC extracts on T cell activation. These findings lead to future studies to further define specific immunomodulatory factors derived from ESCs for potential applications.
4

Immunomodulation of Embryonic Stem Cell-produced MFG-E8 on T Cells

Tan, Yuan January 2015 (has links)
Embryonic stem cells (ESCs) possess certain immunomodulatory properties; the defined components in ESCs, however, are largely unknown. Based on proteomic database, I report here that milk fat globule epidermal growth factor 8 (MFG-E8) is a key component in ESCs to suppress T cell activation and regulate T cell polarization. MFG-E8 is enriched in undifferentiated ESCs while diminishing in differentiated ESCs. Neutralizing ESC-derived MFG-E8 substantially ameliorates the suppressive effects of ESCs on T cell activation and proliferation. Additionally, MFG-E8 in ESCs is capable of up-regulating T regulatory cells. I further prove that MFG-E8 suppresses T cell activation and regulates T cell polarization through inhibiting PKCθ phosphorylation. In vivo teratoma formation assay reveals an increase in ESC engraftments across allogeneic barriers with less immunologic rejection by up-regulation of MFG-E8 expression in ESCs, further validating the immunosuppressive properties of MFG-E8. Identifying an important immunoregulatory component in ESCs will greatly facilitate stem cell-based therapies.
5

Avaliação da Milk Fat Globule Epidermal Growth Factor 8 (MFG-E8), da integrina αvβ3 e da Leukemia Inhibitory Factor (LIF) na implantação embrionária humana : estudo em modelo in vitro e no endométrio de mulheres com e sem endometriose

Schmitz, Carla Regina January 2015 (has links)
Base teórica: O processo de implantação do embrião no ser humano é extremamente complexo e, ao mesmo tempo, essencial para que a mulher possa engravidar. Neste processo, em que o endométrio precisa sofrer uma série de mudanças para tornar-se receptivo, a adequada expressão de MFG-E8 (milk fat globule epidermal growth factor 8), seu receptor a integrina αvβ3 e LIF (leukemia inhibitory factor) parecem ter um papel importante. Além do mais, mulheres com infertilidade e endometriose podem apresentar a falha de implantação como uma grande barreira para obter seu sucesso terapêutico. Objetivos: Avaliar o papel de MFG-E8 e do seu receptor integrina αvβ3 em um modelo de implantação in vitro com uma linhagem celular trofoblástica e outra de epitélio endometrial. Comparar a expressão de MFG-E8, de integrina αvβ3 e de LIF no endométrio de pacientes férteis e inférteis com endometriose durante a janela de implantação. Métodos: No primeiro ensaio, utilizando-se uma linhagem celular bem diferenciada de adenocarcinoma de endométrio (células Ishikawa) e uma linhagem de coriocarcinoma de trofoblasto, o modelo in vitro de implantação humana foi estabelecido. Para investigação do impacto do bloqueio de MFG-E8 e integrina αvβ3, ambas linhagens celulares foram pré-tratadas com anticorpos contra estas proteínas em diferente concentrações antes do ensaio de adesão. No ensaio subsequente, para comparar a expressão de MFG-E8, de integrina αvβ3 e de LIF no endométrio humano, foram realizadas biópsias no período da janela de implantação (LH+7 a LH+10) com cateter de Pipelle. As amostras foram submetidas a imunohistoquímica, e analisadas através do HSCORE. Resultados: Na avaliação in vitro observamos que as células Ishikawa pré-tratadas com anticorpo anti-MFG-E8 causaram diminuição da adesão das esferas Jar dose-dependente. Por outro lado, o pré-tratamento das esferas Jar não resultou em diminuição significativa da adesão. Pré-tratamento com anticorpos anti-integrina αvβ3, tanto de células Ishikawa como de esferas Jar, causaram inibição significativa, dose-dependente, da adesão das esferas. A análise imunohistoquímica das biópsias realizadas durante a janela de implantação mostrou uma expressão aumentada de MFG-E8 em pacientes com endometriose e infertilidade. Além do mais, houve expressão diminuída de LIF no grupo em estudo. Contudo, não houve diferença estatisticamente significativa na expressão de integrina αvβ3 entre os grupos em estudo. Conclusão: Este estudo demonstrou que, quando se bloqueia MFG-E8 ou seu receptor integrina αvβ3 em células Ishikawa em um modelo in vitro, ocorre uma diminuição de adesão das células Jar. Além do mais, bloqueando-se a integrina αvβ3 nas esferas Jar, também ocorre uma diminuição da adesão destas nas células Ishikawa. No entanto, quando estudamos o endométrio in vivo de pacientes com endometriose e infertilidade, encontramos a expressão aumentada de MFG-E8 e diminuída de LIF durante a janela de implantação no endométrio. / Background: The human implantation process is very complex and, at the same time, it is essential for women to achieve pregnancy. In this process, where the human endometrium must go through a lot of changes in order to become receptive, an adequate expression of MFG-E8 (milk fat globule epidermal growth factor 8), integrin αvβ3 and LIF (leukemia inhibitory factor) appear to play an important role. Furthermore, women with endometriosis and infertility may have in their implantation process the key to achieve pregnancy. Objectives: To investigate the role of MFG-E8 and its receptor integrin αvβ3 in the attachment of trophoblast cells to the endometrial epithelium, in an in vitro model. To compare endometrial expression of MFG-E8, integrin αvβ3 and LIF between fertile patients and patients with endometriosis and infertility during the window of implantation. Methods: In our first assay, by using a well-differentiated endometrial adenocarcinoma cell line (Ishikawa cells) and choriocarcinoma human trophoblast cells (Jar cells), an in vitro model mimicking human implantation was established. To investigate the impact of blocking MFG-E8 and integrin αvβ3, the cell lines were pretreated with antibodies against those proteins at different concentrations before the attachment assay. Moreover, to compare endometrial expression of MFG-E8, integrin αvβ3 and LIF, endometrial biopsies were performed during the window of implantation (LH+7 to LH+10) with the Pipelle catheter. The samples were submitted immunochemistry, and analyzed with HSCORE. Results: Pretreatment of Ishikawa cells with anti-MFG-E8 antibody caused a dosedependent and significant inhibition of attachment is our in vitro assay. On the other hand, pretreatment of Jar spheroids did not result in a significant effect on the attachment rate. Pretreatment of Ishikawa cells as well as Jar spheroids with anti-integrin avb3 antibodies resulted in a dose-dependent, significant inhibition of attachment. The immunochemistry analysis of the endometrial biopsies performed during the window of implantation showed increased MFG-E8 expression in patients with endometriosis and infertility. Moreover, there was lower LIF expression in the study group. Conclusion: This study showed that blocking MFG-E8 and its receptor integrin αvβ3 in Ishikawa cells diminishes Jar spheroid attachment in an in vitro model. Moreover, blocking integrin αvβ3 in the trophoblastic cells also diminished their attachment to the Ishikawa monolayer. Nevertheless, when we studied the endometrium of patients with endometriosis and infertility, we saw an increased expression of MFG-E8 and decreased expression of LIF during the window of implantation.
6

Avaliação da Milk Fat Globule Epidermal Growth Factor 8 (MFG-E8), da integrina αvβ3 e da Leukemia Inhibitory Factor (LIF) na implantação embrionária humana : estudo em modelo in vitro e no endométrio de mulheres com e sem endometriose

Schmitz, Carla Regina January 2015 (has links)
Base teórica: O processo de implantação do embrião no ser humano é extremamente complexo e, ao mesmo tempo, essencial para que a mulher possa engravidar. Neste processo, em que o endométrio precisa sofrer uma série de mudanças para tornar-se receptivo, a adequada expressão de MFG-E8 (milk fat globule epidermal growth factor 8), seu receptor a integrina αvβ3 e LIF (leukemia inhibitory factor) parecem ter um papel importante. Além do mais, mulheres com infertilidade e endometriose podem apresentar a falha de implantação como uma grande barreira para obter seu sucesso terapêutico. Objetivos: Avaliar o papel de MFG-E8 e do seu receptor integrina αvβ3 em um modelo de implantação in vitro com uma linhagem celular trofoblástica e outra de epitélio endometrial. Comparar a expressão de MFG-E8, de integrina αvβ3 e de LIF no endométrio de pacientes férteis e inférteis com endometriose durante a janela de implantação. Métodos: No primeiro ensaio, utilizando-se uma linhagem celular bem diferenciada de adenocarcinoma de endométrio (células Ishikawa) e uma linhagem de coriocarcinoma de trofoblasto, o modelo in vitro de implantação humana foi estabelecido. Para investigação do impacto do bloqueio de MFG-E8 e integrina αvβ3, ambas linhagens celulares foram pré-tratadas com anticorpos contra estas proteínas em diferente concentrações antes do ensaio de adesão. No ensaio subsequente, para comparar a expressão de MFG-E8, de integrina αvβ3 e de LIF no endométrio humano, foram realizadas biópsias no período da janela de implantação (LH+7 a LH+10) com cateter de Pipelle. As amostras foram submetidas a imunohistoquímica, e analisadas através do HSCORE. Resultados: Na avaliação in vitro observamos que as células Ishikawa pré-tratadas com anticorpo anti-MFG-E8 causaram diminuição da adesão das esferas Jar dose-dependente. Por outro lado, o pré-tratamento das esferas Jar não resultou em diminuição significativa da adesão. Pré-tratamento com anticorpos anti-integrina αvβ3, tanto de células Ishikawa como de esferas Jar, causaram inibição significativa, dose-dependente, da adesão das esferas. A análise imunohistoquímica das biópsias realizadas durante a janela de implantação mostrou uma expressão aumentada de MFG-E8 em pacientes com endometriose e infertilidade. Além do mais, houve expressão diminuída de LIF no grupo em estudo. Contudo, não houve diferença estatisticamente significativa na expressão de integrina αvβ3 entre os grupos em estudo. Conclusão: Este estudo demonstrou que, quando se bloqueia MFG-E8 ou seu receptor integrina αvβ3 em células Ishikawa em um modelo in vitro, ocorre uma diminuição de adesão das células Jar. Além do mais, bloqueando-se a integrina αvβ3 nas esferas Jar, também ocorre uma diminuição da adesão destas nas células Ishikawa. No entanto, quando estudamos o endométrio in vivo de pacientes com endometriose e infertilidade, encontramos a expressão aumentada de MFG-E8 e diminuída de LIF durante a janela de implantação no endométrio. / Background: The human implantation process is very complex and, at the same time, it is essential for women to achieve pregnancy. In this process, where the human endometrium must go through a lot of changes in order to become receptive, an adequate expression of MFG-E8 (milk fat globule epidermal growth factor 8), integrin αvβ3 and LIF (leukemia inhibitory factor) appear to play an important role. Furthermore, women with endometriosis and infertility may have in their implantation process the key to achieve pregnancy. Objectives: To investigate the role of MFG-E8 and its receptor integrin αvβ3 in the attachment of trophoblast cells to the endometrial epithelium, in an in vitro model. To compare endometrial expression of MFG-E8, integrin αvβ3 and LIF between fertile patients and patients with endometriosis and infertility during the window of implantation. Methods: In our first assay, by using a well-differentiated endometrial adenocarcinoma cell line (Ishikawa cells) and choriocarcinoma human trophoblast cells (Jar cells), an in vitro model mimicking human implantation was established. To investigate the impact of blocking MFG-E8 and integrin αvβ3, the cell lines were pretreated with antibodies against those proteins at different concentrations before the attachment assay. Moreover, to compare endometrial expression of MFG-E8, integrin αvβ3 and LIF, endometrial biopsies were performed during the window of implantation (LH+7 to LH+10) with the Pipelle catheter. The samples were submitted immunochemistry, and analyzed with HSCORE. Results: Pretreatment of Ishikawa cells with anti-MFG-E8 antibody caused a dosedependent and significant inhibition of attachment is our in vitro assay. On the other hand, pretreatment of Jar spheroids did not result in a significant effect on the attachment rate. Pretreatment of Ishikawa cells as well as Jar spheroids with anti-integrin avb3 antibodies resulted in a dose-dependent, significant inhibition of attachment. The immunochemistry analysis of the endometrial biopsies performed during the window of implantation showed increased MFG-E8 expression in patients with endometriosis and infertility. Moreover, there was lower LIF expression in the study group. Conclusion: This study showed that blocking MFG-E8 and its receptor integrin αvβ3 in Ishikawa cells diminishes Jar spheroid attachment in an in vitro model. Moreover, blocking integrin αvβ3 in the trophoblastic cells also diminished their attachment to the Ishikawa monolayer. Nevertheless, when we studied the endometrium of patients with endometriosis and infertility, we saw an increased expression of MFG-E8 and decreased expression of LIF during the window of implantation.
7

Avaliação da Milk Fat Globule Epidermal Growth Factor 8 (MFG-E8), da integrina αvβ3 e da Leukemia Inhibitory Factor (LIF) na implantação embrionária humana : estudo em modelo in vitro e no endométrio de mulheres com e sem endometriose

Schmitz, Carla Regina January 2015 (has links)
Base teórica: O processo de implantação do embrião no ser humano é extremamente complexo e, ao mesmo tempo, essencial para que a mulher possa engravidar. Neste processo, em que o endométrio precisa sofrer uma série de mudanças para tornar-se receptivo, a adequada expressão de MFG-E8 (milk fat globule epidermal growth factor 8), seu receptor a integrina αvβ3 e LIF (leukemia inhibitory factor) parecem ter um papel importante. Além do mais, mulheres com infertilidade e endometriose podem apresentar a falha de implantação como uma grande barreira para obter seu sucesso terapêutico. Objetivos: Avaliar o papel de MFG-E8 e do seu receptor integrina αvβ3 em um modelo de implantação in vitro com uma linhagem celular trofoblástica e outra de epitélio endometrial. Comparar a expressão de MFG-E8, de integrina αvβ3 e de LIF no endométrio de pacientes férteis e inférteis com endometriose durante a janela de implantação. Métodos: No primeiro ensaio, utilizando-se uma linhagem celular bem diferenciada de adenocarcinoma de endométrio (células Ishikawa) e uma linhagem de coriocarcinoma de trofoblasto, o modelo in vitro de implantação humana foi estabelecido. Para investigação do impacto do bloqueio de MFG-E8 e integrina αvβ3, ambas linhagens celulares foram pré-tratadas com anticorpos contra estas proteínas em diferente concentrações antes do ensaio de adesão. No ensaio subsequente, para comparar a expressão de MFG-E8, de integrina αvβ3 e de LIF no endométrio humano, foram realizadas biópsias no período da janela de implantação (LH+7 a LH+10) com cateter de Pipelle. As amostras foram submetidas a imunohistoquímica, e analisadas através do HSCORE. Resultados: Na avaliação in vitro observamos que as células Ishikawa pré-tratadas com anticorpo anti-MFG-E8 causaram diminuição da adesão das esferas Jar dose-dependente. Por outro lado, o pré-tratamento das esferas Jar não resultou em diminuição significativa da adesão. Pré-tratamento com anticorpos anti-integrina αvβ3, tanto de células Ishikawa como de esferas Jar, causaram inibição significativa, dose-dependente, da adesão das esferas. A análise imunohistoquímica das biópsias realizadas durante a janela de implantação mostrou uma expressão aumentada de MFG-E8 em pacientes com endometriose e infertilidade. Além do mais, houve expressão diminuída de LIF no grupo em estudo. Contudo, não houve diferença estatisticamente significativa na expressão de integrina αvβ3 entre os grupos em estudo. Conclusão: Este estudo demonstrou que, quando se bloqueia MFG-E8 ou seu receptor integrina αvβ3 em células Ishikawa em um modelo in vitro, ocorre uma diminuição de adesão das células Jar. Além do mais, bloqueando-se a integrina αvβ3 nas esferas Jar, também ocorre uma diminuição da adesão destas nas células Ishikawa. No entanto, quando estudamos o endométrio in vivo de pacientes com endometriose e infertilidade, encontramos a expressão aumentada de MFG-E8 e diminuída de LIF durante a janela de implantação no endométrio. / Background: The human implantation process is very complex and, at the same time, it is essential for women to achieve pregnancy. In this process, where the human endometrium must go through a lot of changes in order to become receptive, an adequate expression of MFG-E8 (milk fat globule epidermal growth factor 8), integrin αvβ3 and LIF (leukemia inhibitory factor) appear to play an important role. Furthermore, women with endometriosis and infertility may have in their implantation process the key to achieve pregnancy. Objectives: To investigate the role of MFG-E8 and its receptor integrin αvβ3 in the attachment of trophoblast cells to the endometrial epithelium, in an in vitro model. To compare endometrial expression of MFG-E8, integrin αvβ3 and LIF between fertile patients and patients with endometriosis and infertility during the window of implantation. Methods: In our first assay, by using a well-differentiated endometrial adenocarcinoma cell line (Ishikawa cells) and choriocarcinoma human trophoblast cells (Jar cells), an in vitro model mimicking human implantation was established. To investigate the impact of blocking MFG-E8 and integrin αvβ3, the cell lines were pretreated with antibodies against those proteins at different concentrations before the attachment assay. Moreover, to compare endometrial expression of MFG-E8, integrin αvβ3 and LIF, endometrial biopsies were performed during the window of implantation (LH+7 to LH+10) with the Pipelle catheter. The samples were submitted immunochemistry, and analyzed with HSCORE. Results: Pretreatment of Ishikawa cells with anti-MFG-E8 antibody caused a dosedependent and significant inhibition of attachment is our in vitro assay. On the other hand, pretreatment of Jar spheroids did not result in a significant effect on the attachment rate. Pretreatment of Ishikawa cells as well as Jar spheroids with anti-integrin avb3 antibodies resulted in a dose-dependent, significant inhibition of attachment. The immunochemistry analysis of the endometrial biopsies performed during the window of implantation showed increased MFG-E8 expression in patients with endometriosis and infertility. Moreover, there was lower LIF expression in the study group. Conclusion: This study showed that blocking MFG-E8 and its receptor integrin αvβ3 in Ishikawa cells diminishes Jar spheroid attachment in an in vitro model. Moreover, blocking integrin αvβ3 in the trophoblastic cells also diminished their attachment to the Ishikawa monolayer. Nevertheless, when we studied the endometrium of patients with endometriosis and infertility, we saw an increased expression of MFG-E8 and decreased expression of LIF during the window of implantation.
8

Rôle du microenvironnement apoptotique tissulaire et du MFG-E8 dans la modulation de la réponse inflammatoire

Brissette, Marie-Joëlle 09 1900 (has links)
L’inflammation fait partie des processus réactionnels de défense dont dispose l’organisme en réponse aux agressions, assurant l’intégrité de l’hôte. En réponse au dommage tissulaire, plusieurs médiateurs inflammatoires interviennent dans le processus de l’inflammation. Lors de ces dommages, des signaux de dangers provenant de cellules endommagées sont relâchés dans l’environnement tissulaire, pouvant causer des dommages cellulaires et tissulaires. Les macrophages, tout comme d’autres cellules, peuvent être activés par ces signaux de danger, menant à la sécrétion de molécules telles que des cytokines et des chimiokines pouvant modifier le microenvironnement tissulaire. Les insultes au tissu sain peuvent entrainer la mort cellulaire telle que l’apoptose. Les molécules pouvant être relâchées lors de celle-ci contribuent au microenvironnement, notamment de par l’influence de celles-ci sur le macrophage. Parmi ces médiateurs, nous avons identifié le Milk Fat Globule-Epidermal growth factor 8 (MFG-E8), un acteur important dans la résolution de l’inflammation, comme étant relâché spécifiquement par les cellules apoptotiques. Nous avons émis l’hypothèse que le microenvironnement apoptotique tissulaire, via la relâche de MFG-E8, module le phénotype du macrophage, modifiant le microenvironnement, la réponse inflammatoire ainsi que le devenir de l’insulte tissulaire. Nos objectifs sont 1) de caractériser ce microenvironnement apoptotique tissulaire et la cinétique de relâche du MFG-E8 par les cellules apoptotiques, 2) d’en évaluer son rôle dans la modulation du phénotype du macrophage ainsi que 3) d’en étudier, in vivo, son influence sur l’environnement inflammatoire et le devenir tissulaire. Dans le premier article présenté, nous avons démontré que les cellules endothéliales apoptotiques relâchent le MFG-E8 de façon Caspase-3 dépendante. La stimulation des macrophages par l’environnement conditionné par les cellules endothéliales apoptotiques mène à l’adoption d’un profil macrophagien davantage anti-inflammatoire et moindrement pro-inflammatoire. Ce phénotype est réduit par l’inhibition de la Caspase-3 et il dépend de la présence de MFG-E8. De plus, le potentiel du MFG-E8 à la reprogrammation du macrophage pro-inflammatoire a été démontré via un modèle expérimental de péritonite. Ce changement phénotypique médié par MFG-E8 implique une signalisation STAT3. Ayant démontré que les cellules épithéliales apoptotiques, à l’instar des cellules endothéliales apoptotiques, relâchent elles aussi de façon apoptose-dépendante le MFG-E8, nous avons étudié plus exhaustivement un modèle in vivo riche en apoptose épithéliale, l’obstruction urétérale unilatérale. Dans ce deuxième article présenté, nous rapportons l’implication bénéfique de MFG-E8 dans ce modèle de pathologie rénale obstructive. Nous avons constaté que la présence ou l’administration de MFG-E8 réduit le dommage tissulaire et la fibrose. La protection conférée par MFG-E8 est médiée via la modulation de l’activation de l’inflammasome. De plus, nos résultats illustrent l’importance du phénotype anti-inflammatoire du macrophage médié par le MFG-E8 dans la régulation négative de l’activation de l’inflammasome rénal et du dommage tissulaire. Cette thèse présente la première description de la relâche Caspase-3-dépendante de MFG-E8 par les cellules apoptotiques. Elle démontre également l’importance du MFG-E8 dans le microenvironnement apoptotique inflammatoire dans l’atténuation du phénotype pro-inflammatoire du macrophage. De plus, nous avons démontré son rôle protecteur dans des modèles in vivo de transplantation aortique et de réparation tissulaire, de même que dans un modèle de maladie rénale chronique où nous avons montré que cette protection conférée par MFG-E8 est médiée par la régulation négative de l’inflammasome tissulaire. Nos résultats suggèrent ainsi que le MFG-E8 pourrait être considéré comme un interrupteur inflammatoire et ainsi comme une cible potentielle dans la modulation de maladies inflammatoires. / Inflammation is an important component of the « response to injury » process, allowing host integrity. In response to injury, released inflammatory mediators from damaged cells play a crucial role in the modification of the inflammatory microenvironment, which can lead to more cellular and tissue damages. Macrophages can be activated by those danger signals, leading to a spectrum of cytokines and chemokines secretion and modulating the tissular microenvironment. Tissue injuries can lead to cell death such as apoptosis. Mediators released during apoptosis contribute to the nature of the microenvironment, by their influence on macrophage amongst others. We have identified that Milk Fat Globule-Epidermal growth factor 8 (MFG-E8), an important actor in inflammation resolution, is specifically released by apoptotic cells. We hypothesized that tissular apoptotic microenvironment, through MFG-E8 release, modulates macrophage phenotype, resulting in the modification of microenvironment, inflammatory response and tissu injury outcome. Thus, our objectives were to 1) characterize this tissular apoptotic microenvironment by studying MFG-E8 release kinetic by apoptotic cells, to 2) evaluate its role in macrophage phenotype modulation and to 3) study, in vivo, its influence on inflammatory environment and tissu damage outcome. In the first study, we demonstrated that MFG-E8 is released by apoptotic endothelial cells in a caspase-3-dependent manner. When macrophages were exposed to conditioned media from apoptotic endothelial cells, they adopt a high anti-inflammatory, low pro-inflammatory cytokine/chemokine secreting phenotype that is lost if apoptosis is inhibited or if MFG-E8 is absent from the media. Furthermore, MFG-E8 potential to anti-inflammatory macrophage reprogramming has been demonstrated in the experimental peritonitis model. This MFG-E8-mediated reprogramming of macrophages occurs through increased phosphorylation of STAT-3. As apoptotic endothelial cells, apoptotic epithelial cells also release MFG-E8 in an apoptotic-dependent manner. Thus, we investiguated more exhaustively an in vivo epithelial apoptosis rich model, the unilateral ureteral obstruction. In this second study, we report the positive impact of MFG-E8 in this renal obstructive model. MFG-E8 administration reduced kidney damage and fibrosis compared to the control, whereas its absence in MFG-E8 KO mice was associated with more severe disease. Moreover, we demonstrated that the protective role of MFG-E8 is mediated through inflammasome activation modulation in the kidney. Furthermore, our results showed the importance of the anti-inflammatory macrophage phenotype that results in decreased inflammasome activation, preventing severe tissue damage. This thesis presents the first description of apoptosis-dependent release of MFG-E8 by apoptotic cells. It also demonstrate the importance of MFG-E8 in inflammatory apoptotic microenvironment, leading to pro-inflammatory macrophage phenotype attenuation. Moreover, we demonstrated MFG-E8 protective role in an aortic transplantation and a tissue repair models, as well as in a chronic kidney disease model where we showed that this MFG-E8 confered protection is mediated by negative regulation of tissular inflammasome activation. These data provide valuable insight for identifying MFG-E8 as a novel target in the modulation of inflammatory diseases and could be considerate as an inflammatory switch.
9

Étude du rôle de l'inflammation dans l’insulte cérébrale précoce associée à l'hémorragie sous-arachnoïdienne.

Gris, Typhaine 04 1900 (has links)
L’hémorragie sous-arachnoïdienne (HSA) est une pathologie redoutable résultant fréquemment de la rupture d’un anévrisme intracrânien. Elle est associée à une mortalité élevée et d’importants déficits neurologiques. Le saignement entraîne l’augmentation de la pression intracrânienne, la diminution du flux cérébral sanguin, l’apparition de l’inflammation cérébrale et la mort neuronale. Ces évènements de la phase d’insulte cérébrale précoce (72 premières heures) conditionnent le devenir du patient. Le vasospasme était initialement considéré comme la cause principale des ischémies cérébrales retardées (DCI), mais sa diminution pharmacologique n’a montré aucun bénéfice pour les patients. Cependant, l’infiltration rapide des leucocytes dans le SNC suivant le saignement semble impliquée dans le développement des DCI. Notre hypothèse est que l’activation précoce du système immunitaire à la suite de l’HSA est responsable de la mort neuronale retardée et de la survenue des déficits constatés chez les patients souffrant d’HSA. Nos buts étaient de caractériser la contribution des leucocytes dans l’inflammation cérébrale et la mort neuronale dans un modèle murin d’HSA, de moduler cette inflammation par l’utilisation de la protéine MFG-E8, une protéine anti-inflammatoire favorisant la clairance apoptotique, et de confirmer la présence d’une signature immunologique comparable chez les patients HSA. Notre modèle murin nous a permis d’induire chirurgicalement l’HSA et d’injecter par voie intrapéritonéale la protéine MFG-E8. La composition cellulaire du sang (humain et murin) et du cerveau des souris a été analysée par cytométrie en flux. Le plasma (humain et murin) et le liquide céphalo-rachidien (LCR) des patients ont été analysés par dosage cytokinique. Certains cerveaux de souris étaient inclus en paraffine pour l’imagerie par microscopie confocale. La lignée cellulaire de microglie nous a permis d’étudier la modulation de la capacité de phagocytose et de production de ROS par l’exposition au sérum ou au LCR de patients HSA. Dans une première étude, nous avons démontré le rôle de l’inflammation cérébrale précoce dans le développement de la mort neuronale et des symptômes chez les souris HSA. Nous avons également caractérisé la présence de marqueurs inflammatoires systémiques chez les patients HSA. Dans une deuxième étude, nous avons montré que le traitement par la protéine MFG-E8 chez les souris HSA entraînait la diminution de l’inflammation périphérique ainsi que de la présence des marqueurs M1, de l’activation des astrocytes et de la mort neuronale dans le cerveau aboutissant à la diminution de la sévérité des symptômes. L’étude de l’activation immunitaire chez les patients HSA, nous a permis d’observer une signature immunologique similaire à notre modèle murin. Nous avons montré que les patients HSA présentaient une augmentation des cellules immunitaires innées et une immunodépression lymphocytaire en comparaison avec des donneurs sains. Nous avons également décrit l’importance du grade et du genre des patients par la caractérisation d’un profil inflammatoire plus sévère chez les patients hauts gradés et chez les hommes. Finalement, nos résultats confirment l’existence d’une signature immunologique similaire entre les patients HSA et notre modèle murin aboutissant, dans les deux cas, à l’augmentation de l’activation de l’inflammation systémique et cérébrale. Cette signature immunologique est dépendante du sexe et du grade des patients. La diminution de la gravité des symptômes par le traitement avec la protéine MFG-E8 dans notre modèle souris confirme l’implication incontestable de l’inflammation dans l’apparition des déficits moteurs secondaires à la mort neuronale, et le potentiel thérapeutique de cette protéine MFG-E8 dans le développement de nouvelles thérapies. / Subarachnoid hemorrhage (SAH) is a redoubtable pathology resulting frequently from the rupture of an intracranial aneurysm. It is associated to an important mortality and severe neurologic deficits. The bleeding leads to an increase in the intracranial pressure, to a decrease in cerebral blood flow, to the development of cerebral inflammation and to neuronal death. These events of early brain injury (first 72 hours) determine the patient’s prognosis. The vasospasm was first thought to be the main cause of delayed cerebral ischemia (DCI), but its pharmacological decrease was not being associated to any benefits for the patients. However, rapid leucocytic infiltration in the CNS secondary to the bleeding seems implicated in the development of DCI. Our hypothesis is that the early immune system activation in SAH is responsible for delayed neuronal death and for the onset of symptoms in SAH patients. Our goals were to characterize the contribution of leucocytes in cerebral inflammation and neuronal death in our SAH mice model, to modulate the inflammation by using MFG-E8 protein, an anti-inflammatory protein promoting the apoptotic clearance, and to confirm this similar immunologic signature in SAH patients. Our mouse model allows us to surgically induce SAH and to inject the MFG-E8 protein by intraperitoneal injection. The cellular composition of blood (human and mouse) and of mouse brains were analyzed by flow cytometry. The plasma (human and mouse) and the cerebrospinal fluid (CSF) were analyzed by cytokine assay. Some mice brains were paraffin-embedded for confocal microscopy imaging. Microglia cell lines allowed us to evaluate the modulation of phagocytosis and reactive oxygen species (ROS) production secondary to the exposition to SAH patients’ serum and CSF. In the first study, we have demonstrated the impact of early cerebral inflammation on neuronal death and the occurrence of symptoms in SAH mice. We also have characterized the presence of systemic inflammatory markers in SAH patients. In the second study, we have shown that MFG-E8 protein treatment in our SAH mice model is linked to a decrease in peripheric inflammation as well as to a decrease of M1 markers, astrocytic activation and neuronal death in the brain leading to a decrease of symptoms severity. iv The study of immune activation in SAH patients allowed us to observe an immune signature like in our mouse model. We have revealed that SAH patients have an increase in innate immune cells and in lymphocytic immunosuppression in comparison to healthy donors. We have also described the importance of gender and SAH grade by the characterization of a more severe inflammatory profile in high-grade and in male patients. To conclude, our results confirm the existence of a similar immune signature between SAH patients and our mouse model leading in both cases to an increase in systemic and cerebral inflammation. This immunologic signature depends on the patients’ gender and on the grade of SAH. The decrease of symptom severity with MFG-E8 protein treatment in our mice model confirms the unquestionable implication of inflammation in the occurrence of motor deficits secondary to neuronal death and the therapeutic potential of MFG-E8 for the development of new therapies.

Page generated in 0.0456 seconds