• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 24
  • 23
  • 13
  • 3
  • 3
  • 1
  • Tagged with
  • 73
  • 73
  • 26
  • 24
  • 23
  • 22
  • 17
  • 16
  • 15
  • 15
  • 13
  • 13
  • 13
  • 12
  • 11
  • 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.
51

Stratégies de préservation et d'immunoprotection du greffon dans un modèle de transplantation d'îlots pancréatiques / Strategies for great preservation and immunoprotection in a model of pancreatic islets transplantation

Giraud, Sébastien 10 June 2013 (has links)
Actuellement les transplanteurs sont confrontés à une pénurie de greffons, conduisant à l'élargissement des critères de choix des donneurs. Cette démographie fait place à des greffons plus sensibles aux lésions d'ischémie-reperfusion (I/R). Ces lésions conduisent à des dysfonctions de reprises de fonction des greffons, et participent à l'augmentation de l'immunogénicité du greffon et à l'emergence de rejets aigus et chroniques. Dans un premier temps, il est donc nécessaire de limiter les lésions d'I/R et conserver l'intégrité du greffon. Dans un deuxième temps, il est important de réduire l'immunogénicité du greffon et de contrôler le rejet de greffe tout en maintenant le receveur immunocompétent. Afin de limiter les lésions d'I/R nous avons évalué la solution de préservation SCOT de type extracellulaire contenant 30g/L de PEG 20kDa, dans un modèle murin d'isolement et de transplantation d'îlots pancréatiques. L'amélioration des conditions de conservation a permit de préserver l'intégrité des îlots et de réduire l'immunogénicité du greffon, et ce due aux propriétés immunoprotectrices des PEG 20kDa (effets obtenus pour 10 à 30g/L). Dans ce même modèle notre second objectif était d'établir un état de tolérance périphérique par déplétion transitoire des lymphocytes T alloréactifs. La déplétion des lymphocytes T en division a été induite au moment de l'allotransplantation des îlots, par administration transitoire d'un analogue nucleosidique inductible. La déplétion transitoire a permit d'aboutir à une immunotolérance dominante via l'émergence de lymphocytes T régulateurs CD4+CD25+FoxP3+, cellules ouvrant de nouvelles perspectives dans l'inhibition des rejets d'allogreffes. / Organ and tissue transplantation is affected by a shortage of grafts, leading to enlargement of donor criteria. Consequently, these new marginal organs are more susceptible to ischemia-reperfusion injury (IRI). IRI increases primary graft dysfunctions and contributes to increase graft immunogenicity and consequently the occurence of acute and chronic rejection. Our objectives were : firstly, the necessity to limit I/R damages and preserve graft integrity, secondly, the importance to reduce graft immunogenicity and control the graft rejection while maintaining an immunocompetent recipient. To limit IRI we evaluated the new SCOT preservation extracellular type solution containing PEG 20kDa 30g/L in a murine model of pancreatic islets isolation and transplantation. The improvement of conservation with SCOT permitted to maintain the islets integrity and to reduce graft immunogenicity, due to the immunoprotective properties of PEG 20kDa (effects obtained with PEG 20kDa at 10 to 30g /L). In this same model our second objective was to establish a peripheral immunological tolerance of the graft by transient depletion of alloreactive T cells. This depletion of T cells in division was induced at the time of islet allotransplantation by an administration of an inducible nucleosidic analogue during 14 days. Transient alloreactive T cells depletion induced a dominant immunotolerance marked by the emergence of a persistent regulatory T cells CD4+CD25+FoxP3+ population. Thus, regulation of homeostatic balance between effector and regulatory T cells could open an interesting way to control the immune reaction against allograft.
52

L'activation des cellules T CD8+ et T CD4+ en réponse aux auto-antigènes : du tissu lymphoïde à l'organe cible / Activation of CD8+ and CD4+ T cells in response to self-antigen : from the lymphoid tissue to the target organ

Espinosa Carrasco, Gabriel 07 October 2016 (has links)
Le système immunitaire comporte différents mécanismes de tolérance périphérique permettant de contrôler la réponse des cellules T CD8+. Dans certaines conditions encore peu connues, des cellules T potentiellement auto-réactives peuvent contourner les mécanismes de tolérance et se différencier en cellules effectrices, capables d’attaquer différentes organes de l’organisme, dans un processus d’auto-réactivité. En utilisant une souris transgénique exprimant un antigène modèle dans les cellules bêta du pancréas, j’ai étudié deux processus fondamentaux impliqués dans la différenciation des cellules T CD8+ en réponse aux antigènes du soi.1) Rôle de la translocation des lipopolysaccharides (LPS) dans la rupture de la tolérance. Nous avons préalablement démontré dans le laboratoire que des protocoles de lympho-déplétions, tels l’irradiation, étaient capables d’induire une rupture de la tolérance périphérique dans les cellules T CD8+. L’irradiation provoque la translocation des LPS des bactéries commensales vers la circulation sanguine, ce qui induit une activation du système immunitaire inné. Mes données ont montré que la translocation des LPS était corrélée avec l’activation systémique des cellules dendritiques (DC) CD11c+, en particulier les DC CD8+, responsables de la cross-présentation des auto-antigènes pancréatiques dans les tissus lymphoïdes. Alors que le traitement par des antibiotiques avant l’irradiation permet de prévenir la translocation des LPS, l’activation des DC n’est que partiellement affectée, et le développement de l’auto-immunité résultant d’une rupture de la tolérance périphérique des cellules T CD8+ ne peut pas être empêchée par le traitement.2) Visualisation de la coopération entre cellules T CD4+ et CD8+ effectrices dans la destruction des cellules bêta pancréatiques in vivo. En utilisant la microscopie intra-vitale à 2-photons, j’ai pu analyser, pour la première fois, la dynamiques des cellules T CD4+ et CD8+ auto-réactives exprimant un marqueur fluorescent, lors de l’infiltration du pancréas et du développement du diabète auto-immun. J’ai mis en évidence que l’infiltration des cellules T était accompagnée d’un remodelage de la matrice extracellulaire du pancréas, permettant la migration dirigée des lymphocytes. De plus, j’ai montré que l’arrêt MHC classe II-dépendant des cellules T CD4+, dû à des interactions avec des cellules présentatrices d’antigène recrutées au site d’inflammation et impliquant dans certains cas également les cellules T CD8+, contribuait au maintien des fonctions effectrices des cellules T CD8+. / The immune system has evolved multiple mechanisms of peripheral tolerance to control CD8+ T cell responses. Under particular conditions that are not yet well understood, potentially autoreactive T cells may override tolerance and differentiate into effector cells capable of targeting the own components of the organism resulting in self-reactivity. Utilizing transgenic mice expressing a model antigen in the beta cells of the pancreas, I have studied two important processes involved in CD8+ T cells differentiation in response to self-antigens. 1) Role of lipopolysaccharides (LPS) translocation in the breakdown of CD8+ T cell tolerance. It has been previously shown in our laboratory that lymphodepleting protocols, such as total body irradiation, promote breakdown of peripheral CD8+ T cell tolerance. Irradiation induces translocation of commensal bacteria LPS, a potent innate immune system activator, into the bloodstream. My data demonstrated that LPS translocation correlated with systemic activation of CD11c+ dendritic cells (DC), in particular CD8+ DC, responsible for pancreatic self-antigen cross-presentation, in lymphoid tissue. While antibiotic treatment of mice before irradiation prevented LPS translocation, DC activation was only partially affected, and onset of autoimmunity and breakdown of CD8+ T cell tolerance could not be prevented.2) Intra-vital visualization of effector CD8+ and CD4+ T cell cooperation in beta cell destruction in the pancreas. Using two-photon microscopy, I have been able, for the first time, to simultaneously analyze dynamics of fluorescently tagged autoreactive CD8+ and CD4+ T cells as they infiltrated the pancreas and induced autoimmune diabetes. I found that T cell infiltration promoted extracellular matrix remodeling in the pancreas, which in turn served as a scaffold for T cell migration. In addition, I showed that MHC class II dependent arrest of effector CD4+ T cells, due to interactions with antigen presenting cells, occasionally also implicating CD8+ T cells, provided help to effector CD8+ T cells in maintaining their effector functions.
53

AIRE-exprimující buňky v imunitní toleranci ve zdraví a nemoci / AIRE-expressing cells in immune tolerance in health and disease

Vobořil, Matouš January 2020 (has links)
The process of self-nonself discrimination by the immune system is a fundamental attribute of healthy organisms. Since T-cell receptors (TCRs) are generated by the random process of somatic recombination without regard to its targets, the newly developed T-cell clones could recognize either self or nonself antigens. The mechanisms of central tolerance robustly limit the self-reactive repertoire within the T-cell population via deletion of clones that express self-reactive TCRs or their deviation into the regulatory T-cells (Tregs). These processes occur mainly in the thymic medulla where the TCR reactivity to self-antigens is tested by various types of antigen-presenting cells, mainly medullary thymic epithelial cells (mTECs), dendritic cells (DCs), and B-cells. The cooperation between these cell-types has been shown to be essential for the establishment of thymic tolerance. A key molecule regulating the production of self-antigens is the autoimmune regulator (AIRE), which is thought to be expressed primarily by mTECs and its mutations are associated with the development of severe autoimmune disorders. In this context, the presented thesis describes the novel regulatory pathways important for the development of a functional and "harmless" repertoire of T-cells and for enforcement of tolerance....
54

TLR Activation Prevents Hematopoietic Chimerism Induced by Costimulation Blockade: A Dissertation

Miller, David M. 20 May 2008 (has links)
Costimulation blockade based on a donor-specific transfusion and anti-CD154 mAb is effective for establishing mixed allogeneic hematopoietic chimerism and inducing transplantation tolerance. Despite its potential, recent evidence suggests that the efficacy of costimulation blockade can be reduced by environmental perturbations such as infection or inflammation that activate toll-like receptors (TLR). TLR agonists prevent costimulation blockade-induced prolongation of solid organ allografts, but their effect on the establishment of hematopoietic chimerism has not been reported. In this dissertation, we hypothesized that TLR activation during costimulation blockade would prevent the establishment of mixed hematopoietic chimerism and shorten skin allograft survival. To test this hypothesis, costimulation blockade-treated mice were co-injected with TLR2 (Pam3Cys), TLR3 (poly I:C), or TLR4 (LPS) agonists and transplanted with allogeneic bone marrow and skin grafts. Supporting our hypothesis, we observed that TLR agonists administered at the time of costimulation blockade prevented the establishment of mixed hematopoietic chimerism and shortened skin allograft survival. To investigate underlying cellular and molecular mechanisms, we first determined that LPS administration during costimulation blockade did not increase production of alloantibodies or activate natural killer cells. Similarly, costimulation blockade-treated mice depleted of CD4+ or CD8+ cells did not become chimeric when co-injected with LPS. In contrast, mice depleted of both CD4+ and CD8+cell subsets were resistant to the effects of LPS. We next observed that alloreactive T cells were activated by TLR agonists in mice treated with costimulation blockade, and this activation correlated with LPS-induced maturation of donor and host alloantigen-presenting cells. In contrast, TLR4-deficient mice treated with costimulation blockade and LPS did not upregulate costimulatory molecules on their APCs, and mixed chimerism and permanent skin allograft survival were readily achieved. We further observed that injection of recombinant IFN-β recapitulated the detrimental effects of LPS, and that LPS-injected mice deficient in the type I IFN receptor were partially protected. Importantly, alloantigen-presenting cells did not upregulate costimulatory molecules in response to LPS, and mixed chimerism and permanent skin allograft survival were readily established in type I IFN receptor and MyD88 double deficient mice treated with costimulation blockade. We conclude that the TLR4 agonist LPS prevents the establishment of mixed hematopoietic chimerism and shortens skin allograft survival in mice treated with costimulation blockade by inducing the production of type 1 IFN and MyD88-dependent factors that upregulate costimulatory molecules on APCs, leading to the generation of activated alloreactive T cells.
55

Sensitization of CD8 T Cells During Acute Viral Infections Impacts Bystander and Latecomer CD8 T Cell Responses : A Dissertation

Marshall, Heather D. 19 October 2009 (has links)
Many virus infections induce a transient state of immune suppression in the infected host. Virus-induced T cell suppression can be caused by T cell activation-induced cell death (AICD), dendritic cell (DC) apoptosis, DC dysfunction, and/or the enhanced expression of immune-suppressive cytokines. It has been previously demonstrated that naïve bystander CD8 T cells derived from hosts experiencing an acute virus-specific T cell response underwent AICD when polyclonally activated by anti-CD3 in vitro (Zarozinski et al., 2000). Susceptibility of naïve bystander T cells to AICD could prevent the development of a new T cell response during an ongoing immune response, and thus render infected hosts immune suppressed. Although immune suppression could result in an enhanced susceptibility to superinfections, virus-infected individuals are more commonly resistant to superinfecting pathogens. Because of these seemingly contradictory conditions, we sought to investigate how acute viral infections impact naïve bystander CD8 T cells in vivo. More specifically, we asked whether bystander CD8 T cells are susceptible to immune suppression or whether they can contribute to the resistance to superinfections. In order to address this, we examined the responses of bystander CD8 T cells activated with cognate antigen during acute viral infections in vivo. We generated several in vivomodels using P14 (LCMV glycoprotein-specific), HY (male antigen-specific), and OT-I (ovalbumin-specific) transgenic CD8 T cells, which we defined as bystander during acute infections with lymphocytic choriomeningitis virus (LCMV), Pichinde virus (PV), vaccinia virus (VV), and murine cytomegalovirus (MCMV). Consistent with the enhanced susceptibility to cell death noted in vitro, we found that bystander CD8 T cells activated with cognate antigen in vivo during acute viral infections underwent markedly reduced proliferation. Virus-induced transient T cell suppression in vivo was not exclusively mediated by Fas-FasL- or TNF-induced AICD or due to an enhanced susceptibility to apoptosis. Instead, immune suppression in vivowas associated with a delayed onset of division, which we found not to be due to a defect in antigen presentation, but rather due to a T cell intrinsic defect. Despite the suppressed proliferation of TCR-stimulated bystander CD8 T cells in vivo, we found an enhancement of the effector functions exerted by bystander CD8 T cells activated during acute viral infections. During acute viral infections or after stimulation with type 1 IFN (IFN-αβ) inducers, some bystander CD8 T cells were sensitized to immediately exert effector functions such as IFN-γ production and degranulation upon stimulation with high affinity cognate antigen. Sensitization of naïve CD8 T cells required self-MHC I and indirect effects of IFN-αβ, while IL-12, IL-18, and IFN-γ were not individually required. IL-15 was not required for the rapid expression of IFN-γ, but was required for up-regulation of granzyme B (GrzB). P14 and OT-I CD8 T cells, which are capable of homeostatic proliferation, could be sensitized by poly(I:C), but HY CD8 T cells, which are poor at homeostatic proliferation, could not, suggesting that the requirement for MHC I may be to present low affinity cryptically cross-reactive self antigens. Sensitized naive CD8 T cells up-regulated the t-box transcription factor Eomesodermin (Eomes), which can regulate these rapid effector functions. In conclusion, we demonstrate in this thesis that acute viral infections impact naïve bystander CD8 T cells such that their response to cognate antigen is altered. Prior to cognate antigen engagement, bystander CD8 T cells up-regulated Eomes, CD122, and GrzB. Following cognate antigen engagement, bystander CD8 T cells rapidly degranulated and expressed the effector cytokine IFN-γ. The ability of bystander CD8 T cells to rapidly exert effector functions may contribute to the resistance of virus-infected individuals to superinfections. Despite these rapid effector functions, the proliferation of TCR-stimulated bystander CD8 T cells was markedly inhibited. This reduced proliferation was found not to be a defect in antigen presentation, but was a T cell intrinsic defect in initiating division. Thus, bystander CD8 T cells were also susceptible to virus-induced immune suppression. It is also likely that virus-specific CD8 T cells that are not activated until later in the response, so-called latecomer CD8 T cells, may also be susceptible to immune enhancement and suppression. Thus, latecomer CD8 T cells would be able to rapidly exert effector functions at the expense of proliferation. Taken together, we propose that during an immune response, due to spatial and temporal gradients of antigen and inflammation, it is likely that a combination of heterogeneous T cells with different signal strengths and sequences of exposure from cytokines and peptide-MHC constitute the total T cell response to pathogens.
56

Étude de l’impact de la grossesse sur l’hépatite auto-immune dans un modèle expérimental murin

Bourbonnais, Sara 09 1900 (has links)
L’hépatite auto-immune (HAI) est une maladie chronique caractérisée par une destruction progressive du parenchyme hépatique par le système immunitaire. La majorité des patients atteints d’HAI sont des femmes (75% à 90% des cas). L’amélioration des traitements au cours des dernières années a permis à un grand nombre de ces femmes de devenir enceintes. Pendant la grossesse, une rémission spontanée de la maladie a pu être observée chez les femmes atteintes d’HAI. Cette rémission est temporaire et elle est généralement suivie d’une rechute suite à l’accouchement (post-partum). Les causes exactes de cette rémission associée à la grossesse et de la rechute post-partum ne sont pas connues à ce jour. Nous avons donc tenté de reproduire ces phénomènes dans un modèle murin d’HAI développé dans notre laboratoire, afin de déterminer les mécanismes possiblement impliqués. Notre modèle d’HAI consiste en une xéno-immunisation de souris C57BL/6 avec les auto-antigènes impliqués dans l’HAI de type 2 chez l’humain. Nous avons ainsi accouplées des souris préalablement xéno-immunisées, puis nous les avons sacrifiées au début de la 3e semaine de gestation ou 2 à 3 semaines post-partum, afin d’évaluer les dommages hépatiques et afin d’étudier la réponse immunitaire. Comme chez les femmes atteintes d’HAI, les souris présentent une rémission de la maladie pendant la grossesse. Nous en sommes venus à cette conclusion par l’observation d’une diminution de l’inflammation hépatique, des niveaux de transaminases sériques et des titres d’auto-anticorps circulants. À l’inverse des humains, les souris xéno-immunisées ne présentent pas de rechute post-partum. Une analyse des cellules régulatrices (cellules T régulatrices et cellules B productrices d'IL-10) suggère une implication des Tregs hépatiques dans la rémission, car ceux-ci sont augmentés pendant la gestation. Ces Tregs hépatiques sont majoritairement d’origine thymique et ne semblent pas particulièrement attirés au foie en réponse à l’inflammation. La polarisation TH2 est un phénomène connu pendant la grossesse, par contre elle ne semble pas influencer la réponse auto-immune dans nos souris. Une meilleure compréhension des mécanismes d’immunosuppression observés lors de la grossesse pourrait mener au développement d’une thérapie mieux ciblée. / Autoimmune hepatitis (AIH) is a disease of unknown aetiology, characterized by a progressive destruction of the hepatic parenchyma by the immune system. Women are predominantly affected by AIH, with a prevalence of 75% in type 1 AIH and 90% in type 2. Improvement of treatments to control liver inflammation has contributed to increase the number of pregnant patients. During pregnancy, a spontaneous, but temporary, remission of the disease has been observed in women with AIH, often followed by post-partum relapse. Currently, this phenomenon is not fully understood. Thus, we aim to study the mechanisms responsible for the pregnancy-related remission and post-partum relapses in a murine model of AIH developed in our laboratory. Xenoimmunization of C57BL/6 mice with the antigens recognized by autoreactive cells of type 2 AIH patients results in a loss of tolerance, with clinical and biochemical features of AIH similar to those observed in patients. For this study, xenoimmunized mice were mated and sacrificed at the beginning of the 3rd week of gestation or 2-3 weeks post-partum, and liver disease was evaluated at that time as well as the immune response. As it occurs in women with AIH, pregnancy induces a remission of the disease in our murin model. Pregnant mice show a decrease of liver inflammation, ALT levels and circulating autoantibodies. The mice did not show any sign of relapse during the post-partum period. Our analysis of cellular populations with regulatory properties (regulatory T cells and IL-10 secreting B cells) revealed an increase of hepatic Tregs during pregnancy, which suggests an implication of those cells in the remission. Those Tregs infiltrating the liver are mainly originated from the thymus and do not seem to be recruited to the liver in response to inflammation. Although a switch towards a TH2 response is known to occur during pregnancy, this phenomenon does not seem to be implicated in the remission of AIH in our murine model. A better understanding of the natural immunosuppression occurring during pregnancy could bring important knowledge to design new and improved therapy for AIH.
57

Étude fonctionnelle et génétique d'une population de lymphocytes T CD4-CD8- impliquée dans la résistance au diabète auto-immun chez la souris

Dugas, Véronique 04 1900 (has links)
Le diabète auto-immun résulte de la destruction des cellules bêta pancréatiques sécrétrices d’insuline par les lymphocytes T du système immunitaire. Il s’ensuit une déficience hormonale qui peut être comblée par des injections quotidiennes d’insuline d’origine exogène, toutefois il demeure à ce jour impossible de guérir les patients atteints de la maladie. De façon générale, un système immunitaire sain reconnaît une multitude d’antigènes différents et assure ainsi notre défense à l’égard de différents pathogènes ou encore de cellules tumorales. Il arrive cependant que, pour des raisons génétiques et/ou environnementales, les lymphocytes T puissent s’activer de façon aberrante suite à la reconnaissance d’antigènes provenant du soi. C’est ce bris de tolérance qui mène au développement de pathologies auto-immunes telles que le diabète auto-immun. Afin de limiter l’auto-immunité, des mécanismes de sélection stricts permettent d’éliminer la majorité des lymphocytes T présentant une forte affinité envers des antigènes du soi lors de leur développement dans le thymus. Certains de ces lymphocytes réussissent toutefois à échapper à l’apoptose et migrent en périphérie afin d’y circuler en quête d’un antigène spécifiquement reconnu. Il est alors primordial que des mécanismes périphériques assurent le maintien de la tolérance immunitaire en faisant obstacle à l’activation et à la prolifération des lymphocytes T auto-réactifs. L’une des avenues afin d’inhiber le développement de réponses immunitaires aberrantes est la génération de lymphocytes T régulateurs. Ces cellules, d’origine thymique ou périphérique, peuvent arborer différents phénotypes et agissent via de multiples mécanismes afin d’inactiver et/ou éliminer les cellules impliquées dans l’apparition de pathologies auto-immunes. L’utilisation de modèles murins transgéniques a permis la mise en évidence d’une population peu caractérisée de lymphocytes T au potentiel régulateur. En effet, la proportion de ces cellules T n’exprimant pas les corécepteurs CD4 et CD8 (double négatives, DN) a été inversement corrélée à la prédisposition à l’auto-immunité chez ces ii souris. L’objectif principal de cette thèse est de démontrer la fonction immuno-régulatrice des lymphocytes T DN, tout en investiguant les facteurs génétiques responsables du maintien de cette population cellulaire. Nous avons observé que les lymphocytes T DN exercent une activité cytotoxique à l’égard des lymphocytes B de façon spécifique à l’antigène, via la libération de granules cytolytiques contenant du granzyme B et de la perforine. Par ailleurs, nous avons établi qu’un unique transfert adoptif de ces cellules est suffisant afin d’inhiber le développement du diabète auto-immun chez des hôtes transgéniques prédisposés à la maladie. Le recours à des souris déficientes pour l’expression du gène CD47 a permis de constater que la voie de signalisation CD47-Sirp est essentielle dans le maintien de la proportion des lymphocytes T DN. De plus, le locus murin de prédisposition au diabète auto-immun Idd13, qui contient le gène Sirp, a été identifié pour son rôle dans la régulation de la proportion de ces cellules. Finalement, une analyse génétique a révélé que d’autres intervalles génétiques sont impliqués dans le contrôle de la population des lymphocytes T DN. Parmi ceux-ci, un locus situé en région proximale du chromosome 12 a été validé grâce à la création de souris congéniques. Grâce aux résultats présentés dans cette thèse, notre compréhension de la biologie ainsi que de la régulation des lymphocytes T DN est approfondie. Ces connaissances constituent un pas important vers la création de thérapies cellulaires novatrices permettant de prévenir et de guérir diverses pathologies auto-immunes. / Autoimmune diabetes results from the destruction of the insulin-secreting pancreatic beta cells by the T lymphocytes of the immune system. This leads to a hormonal deficiency that can be regulated with daily injections of exogenous insulin. However, to date, there is no cure for autoimmune diabetes. A healthy immune system generally recognizes a multitude of antigens in order to ensure our defence against different pathogens and tumor cells. Yet, depending on genetic and/or environmental factors, individuals may develop T cells that are aberrantly activated following the recognition of self-antigens. This break in tolerance leads to the development of autoimmune pathologies, such as autoimmune diabetes. In order to limit autoimmunity, rigorous selection mechanisms eliminate the vast majority of the T lymphocytes that present a high affinity for self-antigens during their thymic development. However, some of these auto-reactive lymphocytes escape from the elimination processes and migrate to the periphery where they might encounter a self-antigen. It is then essential that peripheral mechanisms maintain the immune tolerance by abrogating the activation and the proliferation of these self-specific T lymphocytes. One of the means to inhibit aberrant immune responses is the generation of regulatory T lymphocytes. These cells, which can be of thymus or peripheral origin, display various phenotypes and can mediate their action through several mechanisms in order to inactivate and/or eliminate the cells that are implicated in the development of autoimmune diseases. The use of transgenic mouse models made it possible to identify a poorly characterized population of T lymphocytes that exhibit a regulatory potential, namely CD4-CD8- (double negative, DN T cells). Indeed, the proportion of DN T cells in lymphoid organs is inversely correlated to autoimmune predisposition. The main objective of this thesis is to determine the immunoregulatory function of the DN T cells, as well as to reveal the genetic factors underlying the regulation of the proportion of DN T cells. iv We observed that through the release of cytolytic granules containing granzyme B and perforin, DN T lymphocytes exert a cytotoxic activity towards B cells in an antigen-specific manner. In addition, we have established that a single injection of those DN T cells is sufficient to inhibit the development of autoimmune diabetes in highly susceptible transgenic mice. The use of CD47 deficient mice also demonstrated that the CD47-Sirp pathway is essential to maintain DN T cell proportion. Also, we identified that the autoimmune diabetes susceptibility locus Idd13, which contains Sirp participates in defining the proportion of DN T cells. Finally, a genetic analysis revealed that other loci are implicated in the control of the DN T cell population. Among those, the role of a locus situated in the proximal region of chromosome 12 has been validated through to the generation of congenic mice. The results presented in this thesis have allowed us to enhance our understanding of the biology and genetic regulation of DN T lymphocytes. This knowledge constitutes an important step towards the creation of innovative cellular therapies that may prevent and cure a diversity of autoimmune pathologies.
58

Étude de l’impact de la grossesse sur l’hépatite auto-immune dans un modèle expérimental murin

Bourbonnais, Sara 09 1900 (has links)
L’hépatite auto-immune (HAI) est une maladie chronique caractérisée par une destruction progressive du parenchyme hépatique par le système immunitaire. La majorité des patients atteints d’HAI sont des femmes (75% à 90% des cas). L’amélioration des traitements au cours des dernières années a permis à un grand nombre de ces femmes de devenir enceintes. Pendant la grossesse, une rémission spontanée de la maladie a pu être observée chez les femmes atteintes d’HAI. Cette rémission est temporaire et elle est généralement suivie d’une rechute suite à l’accouchement (post-partum). Les causes exactes de cette rémission associée à la grossesse et de la rechute post-partum ne sont pas connues à ce jour. Nous avons donc tenté de reproduire ces phénomènes dans un modèle murin d’HAI développé dans notre laboratoire, afin de déterminer les mécanismes possiblement impliqués. Notre modèle d’HAI consiste en une xéno-immunisation de souris C57BL/6 avec les auto-antigènes impliqués dans l’HAI de type 2 chez l’humain. Nous avons ainsi accouplées des souris préalablement xéno-immunisées, puis nous les avons sacrifiées au début de la 3e semaine de gestation ou 2 à 3 semaines post-partum, afin d’évaluer les dommages hépatiques et afin d’étudier la réponse immunitaire. Comme chez les femmes atteintes d’HAI, les souris présentent une rémission de la maladie pendant la grossesse. Nous en sommes venus à cette conclusion par l’observation d’une diminution de l’inflammation hépatique, des niveaux de transaminases sériques et des titres d’auto-anticorps circulants. À l’inverse des humains, les souris xéno-immunisées ne présentent pas de rechute post-partum. Une analyse des cellules régulatrices (cellules T régulatrices et cellules B productrices d'IL-10) suggère une implication des Tregs hépatiques dans la rémission, car ceux-ci sont augmentés pendant la gestation. Ces Tregs hépatiques sont majoritairement d’origine thymique et ne semblent pas particulièrement attirés au foie en réponse à l’inflammation. La polarisation TH2 est un phénomène connu pendant la grossesse, par contre elle ne semble pas influencer la réponse auto-immune dans nos souris. Une meilleure compréhension des mécanismes d’immunosuppression observés lors de la grossesse pourrait mener au développement d’une thérapie mieux ciblée. / Autoimmune hepatitis (AIH) is a disease of unknown aetiology, characterized by a progressive destruction of the hepatic parenchyma by the immune system. Women are predominantly affected by AIH, with a prevalence of 75% in type 1 AIH and 90% in type 2. Improvement of treatments to control liver inflammation has contributed to increase the number of pregnant patients. During pregnancy, a spontaneous, but temporary, remission of the disease has been observed in women with AIH, often followed by post-partum relapse. Currently, this phenomenon is not fully understood. Thus, we aim to study the mechanisms responsible for the pregnancy-related remission and post-partum relapses in a murine model of AIH developed in our laboratory. Xenoimmunization of C57BL/6 mice with the antigens recognized by autoreactive cells of type 2 AIH patients results in a loss of tolerance, with clinical and biochemical features of AIH similar to those observed in patients. For this study, xenoimmunized mice were mated and sacrificed at the beginning of the 3rd week of gestation or 2-3 weeks post-partum, and liver disease was evaluated at that time as well as the immune response. As it occurs in women with AIH, pregnancy induces a remission of the disease in our murin model. Pregnant mice show a decrease of liver inflammation, ALT levels and circulating autoantibodies. The mice did not show any sign of relapse during the post-partum period. Our analysis of cellular populations with regulatory properties (regulatory T cells and IL-10 secreting B cells) revealed an increase of hepatic Tregs during pregnancy, which suggests an implication of those cells in the remission. Those Tregs infiltrating the liver are mainly originated from the thymus and do not seem to be recruited to the liver in response to inflammation. Although a switch towards a TH2 response is known to occur during pregnancy, this phenomenon does not seem to be implicated in the remission of AIH in our murine model. A better understanding of the natural immunosuppression occurring during pregnancy could bring important knowledge to design new and improved therapy for AIH.
59

Tolerância ao leite processado em altas temperaturas em pacientes com alergia ao leite de vaca mediada pela imunoglobulina E / Tolerance of baked milk in patients with cow\'s milk allergy mediated by immunoglobulin E

Barbosa, Claudia Plech Garcia 01 March 2016 (has links)
INTRODUÇÃO: A incidência de pacientes apresentando alergia à proteína do leite de vaca (APLV) após os 5 anos de idade vem crescendo. Definir se estes pacientes tolerariam a ingestão de alimento produzido com leite processado a altas temperaturas (LPAT) proporcionaria melhor qualidade de vida, definiria melhor prognóstico e possibilitaria avaliar a indicação de dessensibilização com muffin. OBJETIVO: (1) identificar quais pacientes com APLV persistente aos quatro anos poderiam tolerar a ingestão de LPAT, (2) descrever as características clínicas e laboratoriais dos grupos reativo e não reativo ao LPAT, e (3) compara-las entre os dois grupos. MÉTODOS: Estudo transversal, utilizando amostra de conveniência, incluindo todos os pacientes acompanhados no ambulatório de alergia alimentar do Instituto da Criança HCFMUSP que preenchiam os critérios de inclusão e que concordaram em realizar o TPO, entre janeiro/2013 e novembro/2014. Os pacientes foram admitidos em hospital-dia sob supervisão médica e submetidos à ingestão de um muffin contendo 2,8 gramas de proteína do leite de vaca. Foram definidos como tolerantes se não apresentassem nenhuma reação alérgica. Estes pacientes foram submetidos na sequência a novo TPO com leite de vaca in natura para excluir a tolerância ao leite de vaca. RESULTADOS: Foram realizados 38 TPO com LPAT, sendo que 30 pacientes (15 masculinos) preencheram todos os critérios de inclusão. A mediana da idade foi de 7 anos e 7 meses (4a10m -14a2m). 14 pacientes (46%) não apresentaram reação após a ingestão do muffin, sendo considerados como não reativos. A análise comparativa entre os grupos reativos e não reativos ao LPAT, não mostrou diferença estatisticamente significante quanto às características clínicas: idade (p=0,8), sexo (p=0,4), história pessoal de rinite (p=0,7), história pessoal de asma (p=0,7), história pessoal de outras alergias (p=0,6), história familiar de rinite (p=0,7), história familiar de asma (p=0,3), história familiar de outras alergias (p=0,1), relato de anafilaxia prévia (p=0,07), relato de ingestão de traços de leite previamente ao TPO (p=0,4), relato de reação alérgica no último ano antes da provocação (p=0,6), relato de anafilaxia no último ano antes do TPO (p=0,6). Não se observou diferença estatisticamente significante entre os dois grupos para IgE total (p=0,1) e eosinófilos (p=0,6). O teste de puntura para leite de vaca e frações mostrou diferença estatisticamente significante para ?-lactoalbumina (p= 0,01) e para a caseína (p = 0,004); em relação ao ImmunoCAP® apenas para a caseína (p= 0,05) essa diferença foi significante. Ao avaliar estes pacientes 1 ano após o TPO, nenhum dos 16 pacientes que foram reativos ao LPAT estava ingerindo leite de vaca, enquanto 28% dos pacientes que foram tolerantes ao LPAT estavam consumindo leite de vaca in natura sem reação (p=0,037). CONCLUSÃO: O estudo mostrou que os pacientes com APLV desta amostra brasileira apresentaram 2 diferentes fenótipos, sendo que aproximadamente metade tolerou o LPAT. Sendo assim, o TPO para LPAT deve ser considerado para pacientes com APLV, sempre sob supervisão médica e estrutura segura e adequada, pois pode contribuir para uma mudança no paradigma do seguimento destes pacientes. Teste de puntura e ImmunoCAP® para caseína podem sugerir quais pacientes estariam tolerantes ao TPO com LPAT, reforçando dados da literatura internacional / INTRODUCTION: The incidence of patients with cow\'s milk allergy (CMA) after the age of 5 has been growing. Defining whether these patients can tolerate the ingestion of food produced with baked milk without allergy reaction could provide a better quality of life, a better prognosis and would make it possible to evaluate indication of desensitization with baked milk. OBJECTIVE: (1) To identify which patients with persistent CMA at the age of four could tolerate the baked milk, (2) to describe the clinical and laboratory characteristics of the baked milk reactive group and the baked milk non-reactive group, and (3) to compare those two groups. METHODS: A cross-sectional study was conducted between January/2013 and November/2014. A convenience sample was applied, including all the patients followed in the Food Allergy Center of the Instituto da Criança HCFMUSP, who met inclusion criteria and agreed to carry out the oral food challenge (OFC). The patients were admitted to a day-hospital under medical supervision. They were submitted to a muffin intake containing 2.8 grams of cow\'s milk protein, and then classified as tolerant if they did not present any allergic reaction. To exclude cow\'s milk tolerance these patients were submitted to a new OFC with cow\'s milk in natura. RESULTS: 38 OFC with baked milk were performed, 30 patients (15 male) met all of the inclusion criteria. The median of age was 7 years and 7 months (4y10m -14y2m). 14 patients (46.6%) were considered as non-reactive because they did not present any reaction after the muffin intake. The comparative analysis between baked milk reactive group and baked milk non-reactive group did not show statistically significant difference in the clinical characteristics: age (p=0.8), gender (p=0.4), personal history of rhinitis (p=0.7), personal history of asthma (p=0.7), personal history of others allergies (p=0.6), family history of rhinitis (p=0.7), family history of asthma (p=0.3) family history of others allergies (p=0.1), previous anaphylaxis report (p=0.07), report of milk traits intake prior to OFC (p=0.4), allergic reaction in the last year before the OFC (p=0.6), anaphylaxis in the last year before the OFC (p=0.6). There was no statistically significant difference between the two groups for total IgE (p=0.1) and eosinophils (p=0.6). The Prick test for cow\'s milk and fractions showed statistically significant difference for ?-lactalbumin (p = 0.01) and for casein (p =0.004); in relation to the ImmunoCAP® only for casein (p=0.05) this difference was significant. After 1 year of the OFC, none of the patients which have been reactive to the baked milk were ingesting cow\'s milk, while 28% of the baked milk tolerant patients were consuming cow\'s milk in natura without reaction (p=0.037). CONCLUSION: The present study showed that patients with CMA of this brazilian sample presented 2 different phenotypes. Approximately half of them tolerated baked milk at age four. In conclusion, OFC for baked milk should be considered for patients with CMA, always under medical supervision and appropriate structure, so it could contribute for a change in these patients follow-up. Prick test and ImmunoCAP® for casein can suggest which patients would tolerate the OFC with baked milk, strengthening data of international literature
60

Caractérisation in vivo de la réponse des lymphocytes T CD4+ naïfs spécifiques d'un néoantigène à différents stades du développement tumoral / Characterization of the in vivo response of naive CD4+ T cells upon tumor neoantigen recognition at different stages of tumor development

Alonso Ramirez, Ruby 23 November 2016 (has links)
Au cours du développement tumoral, le système immunitaire est constamment exposé aux antigènes tumoraux, mais le plus souvent dans un contexte non-inflammatoire qui favorise l'induction d'une tolérance envers ces antigènes. La tolérance peut être médiée par des mécanismes passifs (ignorance, anergie ou délétion des clones spécifiques de la tumeur) ou actifs, pour lesquels les lymphocytes T régulateurs (Tregs) jouent un rôle prépondérant. Les lymphocytes T (LT) CD4+ sont la source principale des Tregs mais présentent également des fonctions antitumorales directes et indirectes. Les connaissances actuelles sur le rôle des LT CD4+ au cours du développement tumoral proviennent en grande partie d'études de modèles murins de tumeurs transplantées. Cependant, l'inflammation initiée lors de l'inoculation de ces tumeurs due à une mort cellulaire importante favorise la présentation persistante par le MHC-II des antigènes tumoraux dans un environnement inflammatoire artificiel. Nous tentons ici de contourner ce problème en utilisant deux modèles murins différents: le premier est un modèle de tumeur transplantée dans lequel un néoantigène de classe II (DBY) est induit à distance du moment de l'inoculation de la tumeur. Le deuxième est un modèle d'adénocarcinome pulmonaire induit génétiquement exprimant l’epitope DBY, dans lequel la tumorigenèse est initiée par l'expression d’un oncogène associée à la délétion d’un gène suppresseur de tumeurs. La réponse antitumorale des LT CD4+ est suivie par le transfert de LT CD4+ spécifiques de DBY "Marilyn". Dans le modèle de tumeur transplantée, nous montrons que l'apparition d'un néoantigène dans une tumeur bien établie n'est pas ignorée par le système immunitaire. Bien au contraire, le néoantigène arrive au ganglion drainant la tumeur et induit une activation efficace des cellules Marilyn, qui prolifèrent, produisent de l'IFN-γ et recirculent jusqu'à la tumeur. En revanche, malgré une activation efficace des LT CD4+, les tumeurs ne sont pas rejetées. Dans le modèle génétiquement induit, nous montrons que des néoantigènes exprimés dès le début du développement tumoral, arrivent jusqu'au ganglion drainant la tumeur en quantité suffisante pour induire l'activation et la prolifération des LT CD4+, mais que cette activation est non-optimale et ne permet qu'une faible migration vers le site de la tumeur. En revanche, une partie des cellules Marilyn acquièrent l'expression de FOXP3 ainsi qu'une signature transcriptomique de Tregs et ce dès les stades précoces du développement tumoral, tandis que le reste des cellules Marilyn présentent un phénotype anergique (CD44hiCD73hiFR4hi). L'administration de CpG n'empêche pas la conversion en Treg des cellules Marilyn, malgré l'augmentation de la maturation des cellules dendritiques dans le poumon et le ganglion drainant la tumeur. La déplétion des Tregs de l'hôte en revanche inhibe cette conversion et favorise l'activation des cellules Marilyn en cellules effectrices compétentes, capables de migrer jusqu'au site tumoral. Enfin, les cellules Marilyn, lorsqu'elles sont activées hors du ganglion drainant la tumeur échappent à l'inhibition induite par la tumeur et deviennent des cellules effectrices compétentes. Ainsi, dans un modèle tumoral reproduisant le développement naturel progressif des tumeurs humaines, un état de tolérance est induit par la tumeur. Cette tolérance est dépendante des Tregs présents dans le ganglion drainant la tumeur, qui confèrent une tolérance aux LT CD4+ naïfs arrivant dans le ganglion. / During tumor development, the immune system is persistently exposed to tumor-associated antigens, frequently in a non-inflammatory context, favoring the establishment of tolerance. Passive (ignorance, anergy or deletion of tumor-specific T cells) or active mechanisms mediated by regulatory T cells (Tregs) may be involved in tolerance. CD4+ T cells are the main source of Tregs but they also display indirect and direct antitumor activity. So far, the contribution of CD4+ T cells during tumor development has been mainly addressed in murine transplanted tumor models. However, in these models the artificial inflammation associated with the presence of dying tumor cells at the time of tumor inoculation favors a long-lasting MHC-II-restricted tumor antigen presentation in an artificial inflammatory context. Here, we addressed this issue using two different models: a transplanted one in which the MHC-II neoantigen (DBY) is induced long after tumor implantation and a genetically engineered mouse (GEM) model of lung adenocarcinoma also expressing the DBY epitope, in which malignant transformation results from both the expression of an oncogene and the deletion of a tumor suppressor gene. Tumor-specific CD4+ T cell response was followed by transfer of naive DBY-specific Marilyn CD4+ T cells. In the transplanted tumor model, we found that the appearance of a neoantigen in established tumors was not ignored by the immune system. On the contrary, the neoantigen reached the tumor-draining lymph node (TdLN) and induced efficient priming of Marilyn cells that proliferated, produced IFN-γ, and recirculated to the tumor site. However, despite efficient induction of a tumor-specific CD4+ T cell response, tumors were not rejected. In the GEM model, we found that starting at the early tumor stages, neoantigens were expressed and reached the TdLN in sufficient amount to induce activation and proliferation of naive Marilyn T cells. However, this priming was suboptimal and resulted in a weak migration to the tumor site. Instead, some of the activated Marilyn cells acquired the expression of FOXP3 and a Treg gene signature while the remaining FOXP3- cells displayed a CD44hiCD73hiFR4hi anergic phenotype. CpG administration did not revert the Marilyn Treg conversion despite reinforcing dendritic cell maturation in the lung and the TdLN. Depletion of the host Treg compartment however, inhibited this conversion and favored Marilyn cell activation into full-blown effector cells able to migrate to the tumor site. Finally, Marilyn cells that were primed at distance of the TdLN, escaped tumor induced inhibition and became full effectors. Thus, in a tumor model reproducing the natural development of slowly growing human tumors, a tumor-associated dominant tolerance is established in the lymph node draining the tumor. This state of unresponsiveness is highly dependent on the presence of Treg cells in the TdLN, conferring tolerance to incoming tumor-specific naive CD4+ T cells.

Page generated in 0.0685 seconds