• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 3
  • 3
  • 2
  • 1
  • Tagged with
  • 15
  • 15
  • 15
  • 9
  • 8
  • 8
  • 7
  • 7
  • 6
  • 6
  • 5
  • 5
  • 5
  • 4
  • 4
  • 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

Proliferation and lineage potential in fetal thymic epithelial progenitor cells

Cook, Alistair Martin January 2010 (has links)
The thymic stroma primarily comprises epithelial, mesenchymal and endothelial cells, interspersed with those of haematopoietic origin. Thymic epithelial cells (TECs) are highly heterogeneous, but can be divided into two broad lineages, cortical and medullary, based on phenotype, functionality and location. A population of Plet1+ TEC progenitors have been identified which, when isolated from mouse E12.5 or E15.5 fetal thymus, reaggregated, and grafted, can produce a functional thymus. However, the potential of individual progenitors to form cortex and/or medulla is undefined. The main aim of this thesis was to use retrospective clonal analysis to ascertain the point during thymus ontogeny at which the cortical and medullary lineages diverge. To this end, I used transgenic mice carrying a ubiquitous ROSA26laacZ reporter gene (where a duplication within lacZ encodes non-functional b-galactosidase). Here, rare, random laacZ-lacZ genetic recombinations result in heritable expression of functional b-gal, producing labelled clones. As this occurs at a known frequency, determination of TEC numbers would enable calculation of the expected number of TEC clones present throughout ontogeny. Due to the lack of quantitative data on all thymic cell populations, I determined the size not only of TEC (lin-EpCAM+), but also haematopoietic (CD45+), mesenchymal (lin-PDGFRa+ and/or lin-PDGFRb+) and endothelial (lin-CD31+) populations from E12.5 until E17.5. I then showed that the absolute number of Plet1+ TECs remains constant during this time, although the proportion of Plet1+ cells in cycle decreases. From these collective data, I propose a model for the role of the Plet1+ population in thymus development, in which Plet1+ cells continually give rise to Plet1- TECs in a self-renewing manner. Finally, I present a ‘dual origin coefficient’ strategy for analysis of a library of prospective TEC clones. I calculated the number of TEC lacZ+ clones expected to be present throughout thymus ontogeny, selecting an appropriate developmental stage for analysis. Although I observed several clones of apparent mesenchymal origin, supporting a single origin for intrathymic and capsular mesenchyme at E15.5, I observed no TEC clones in this extensive analysis. The CpG content of the ROSA26 promoter suggests a possibility of methylation-induced silencing brought about by de novo methylation of the lacZ reporter gene.
2

Towards understanding the signalling requirements of thymic epithelial progenitor cells

Liu, Dong January 2018 (has links)
Thymic epithelial cells (TECs) are indispensable for the development of T cells in the thymus. Two subtypes of TECs exist in the thymus, medullary mTECs and cortical cTECs. Both mTECs and cTECs originate from endodermal thymic epithelial progenitor cells (TEPCs) in the embryo, but how the differentiation of TEPCs is regulated is not well understood. The aims of this thesis were to establish the role of Notch signalling in TEPC differentiation, and how it interacts with known regulators such as FOXN1 and the NFκB pathway. Gene expression data showed that Notch is active in TEPCs and exhibits a correlation with the mTEC lineage. Loss of Notch function led to a significant reduction in the number of mTECs in the thymus, and this can be attributed to aberrant mTEC specification. Furthermore, the duration of Notch activity in determining mTEC number appears limited to the early phase of organogenesis, and precedes RANK/NFκB mediated mTEC proliferation. Gain of Notch function resulted in a considerable shift to a primitive, TEPC-like phenotype, and subsequently a latent increase in mTEC frequency. Finally, transcriptomic and functional analyses pointed to a cross-repressive mechanism between Notch and FOXN1 in TEPCs. Taken together, these results identified Notch as a novel regulator of mTEC specification, likely through maintaining the potency of fetal TEPCs, a prerequisite for mTEC lineage commitment.
3

Towards differentiation of mouse embryonic stem cells to thymic epithelial progenitor cells

Jin, Xin January 2013 (has links)
The thymus is the major site for T-cell generation and thus is important for the adaptive immune system. Development of a properly selected, functional T-cell repertoire relies on interactions between developing T cells and a series of functionally distinct thymic stroma cell types including the cortical and medullary thymic epithelial cells (TECs). The thymus is one of the first organs to degenerate in normal healthy ageing. Related to this, there is strong interest in developing protocols for improving thymus function in patients by cell replacement or regenerative therapies. Thymic epithelial progenitor cells (TEPCs) represent a potential source of cells for thymus transplantation. However, the only source of these cells for transplantation is currently fetal thymus tissue. If TEPCs could be generated from pluripotent cells, this could provide an alternative source of cells for transplantation. The work described in this thesis therefore had two central aims (i) to test the stability of thymic epithelial progenitor cells in vivo and (ii) to investigate the possibility of generating TEPCs or TECs from mouse embryonic stem (ES) cells. The forkhead transcription factor, Foxn1, is essential for the development of a functionally mature thymic epithelium, but is not necessary for formation of the thymic primordium or for medullary thymic epithelial sub-lineage specification. By reactivating Foxn1 expression postnatally in mice carrying a revertible hypomorphic allele of Foxn1, Foxn1R, I herein demonstrate that TEPCs that can express only low levels of Foxn1 mRNA can persist postnatally in the thymic rudiment in mice until at least 6 months of age, and retain the potential to give rise to both cortical and medullary thymic epithelial cells (cTECs and mTECs). These data demonstrate that the TEPC-state is remarkably stable in vivo under conditions of low Foxn1 expression. In parallel with this work, I confirmed the possibility of generating Foxn1-expressing cells from mouse ES cells by using a Foxn1 reporter cell line. As the thymic epithelium has a single origin in the third pharyngeal pouch (3pp) endoderm, I then tested whether or not TEPCs and /or TECs were generated during ES cell differentiation via existing protocols for generating anterior definitive endoderm differentiation cells from mouse ES cells. From this work, I showed that genes expressed in the 3pp and/or TEPC,-including Plet-1, Tbx1, Hoxa3 and Pax9, were induced by differentiation of ES cells using these protocols. I further showed that cells expressing both Plet-1, a marker of foregut endoderm and 3pp, and EpCAM, a marker of proliferating epithelial cells, were induced using a novel protocol (2i ADE) for generating ES cells from ADE. However, gene expression analysis and functional testing suggested that the majority of these cells were non-thymus lineage. I subsequently developed a novel protocol which combined this 2i ADE protocol with co-culturing of the differentiating ES cells with fetal thymic lobes, and demonstrated that this further induced 3pp and TEPC related genes. Finally, I modified the culture conditions in this protocol to conditions predicted to better support TEPC/TEC, and showed that in these conditions, the TEPC-specific markers Foxn1 and IL-7 were induced more strongly than in any other conditions tested. The data presented in this thesis therefore represent an advance towards an optimized protocol for successfully generating TEPCs from ES cells in vitro.
4

Découverte de cellules souches potentielles de l’épithélium thymique

Dumont-Lagacé, Maude 05 1900 (has links)
Le thymus subit un vieillissement précoce, appelé involution thymique, qui cause une perte de fonction du thymus avec l’âge. À ce jour, les mécanismes de renouvellement des cellules épithéliales thymiques (TECs) sont encore mal compris, c’est pourquoi nous avons voulu identifier les cellules souches de l’épithélium thymique. Comme les cellules souches sont quiescentes dans plusieurs tissus, les objectifs de notre étude étaient de déterminer si l’épithélium thymique contenait des cellules quiescentes et d’étudier la cinétique de prolifération des TECs chez les souris jeunes et adultes. Pour ce faire, nous avons utilisé une souris transgénique (H2B-GFP Tet-On) nous permettant d’identifier les cellules ne se divisant pas sur une longue période de temps (LRC, label-retaining cells¬). Nous avons d’abord montré que les TECs proliféraient plus rapidement chez les femelles que les mâles. De plus, nous avons trouvé plusieurs différences entre l’épithélium thymique post-natal et adulte : (1) les TECs corticales (cTECs) et médullaires (mTECs) ont un taux de prolifération similaire chez les jeunes souris, mais chez l’adulte, les cTECs prolifèrent plus lentement que les mTECs; (2) les TECs prolifèrent plus rapidement chez les souris jeunes que adultes; (3) des LRC sont détectées chez l’adulte, mais pas chez les jeunes souris. Les LRC, retrouvées dans le compartiment cTEC, sous-expriment des gènes associés à la sénescence et surexpriment des gènes importants pour le développement et le renouvellement des TECs. Ces résultats montrent que ces cellules sont quiescentes et suggèrent qu’elles pourraient bel et bien être les progéniteurs thymiques responsables du renouvellement des TECs adultes. / The thymus undergoes a rapid degeneration with age termed thymic involution that causes a loss of function of the thymus with age. To this day, mechanisms of thymic maintenance are still unknown. This is why we aimed to identify thymic epithelial stem cells. Since stem cells are quiescent in many tissues in adults, our main objectives were to determine whether the thymic epithelium contains quiescent cells and study the proliferation kinetics of thymic epithelial cells in neonatal and adult mice. To this end, we used the transgenic mouse model H2B-GFP Tet-On, a label-retaining assay allowing us to identify cells that have not divided for a prolonged period of time, which are called label-retaining cells (LRC). First, we showed that in the adult thymus, females’ thymic epithelial cells (TECs) proliferated more actively than males’ TECs. We observed three main differences between neonatal and adult thymi: (1) cTECs and mTECs have similar proliferation rates in young, but mTECs cycled more actively in adult mice; (2) neonatal TECs have a higher turnover rate than adult’s TECs, and (3) we were able to detect LRC in adult mice, but not in neonatal mice. These LRC are contained in the cTEC compartment and express very low levels of senescence-associated proteins and show a high expression of genes important for thymic development and. These results show that the LRC identified in adult thymi are not senescent cells and therefore might represent the elusive thymic progenitor cells responsible for thymic maintenance and regeneration in adult mice.
5

Prise en charge des thymomes chez l'homme : développement de cultures de cellules épithéliales dérivées de tumeurs pour la compréhension des dérégulations de la cellule tumorale / Management of human thymomas : development of thymic epithelial cell cultures derived from tumors for the understanding of tumoral cells dysregulation

Maury, Jean-Michel 23 May 2019 (has links)
Les tumeurs épithéliales thymiques (TET) humaines sont rares (250 - 300 cas/an en France). On distingue les thymomes de type A, AB ou B d'évolution lente avec une survie actuarielle > 95% à 5 ans pour les stades précoces et les carcinomes thymiques d'évolution plus sévère avec une survie actuarielle à 5 ans < 20% pour les stades IV. La pierre angulaire du traitement des TET est l'exérèse chirurgicale complète, facteur pronostique le plus significatif identifié à ce jour. Les récidives des TET, essentiellement pleurales pour les thymomes et générales pour les carcinomes thymiques, sont de prise en charge complexe. Les avancées thérapeutiques sont limitées notamment par l'absence de modèles d'étude de la cellule épithéliale thymique tumorale. Dans le cadre d'une prise en charge multidisciplinaire des récidives pleurales métastatiques, nous avons développé la pleurectomie de cytoréduction associée à une chimio hyperthermie (cisplatine/ mitomycine ; 42°C) intra thoracique (CHIT) pour la prise en charge des métastases pleurales de thymome. Chez des patients sélectionnés (n=19), la médiane de survie sans récidive était de 53 mois et les survies actuarielles à 1 an et 5 ans étaient respectivement de 93% et 86%. Cette technique chirurgicale innovante a permis de développer une alternative à la morbide pleuro pneumonectomie. L'efficacité de la CHIT pose des questions sur le rôle de l'hyperthermie et sur le type de chimiothérapie à associer. Avec pour objectif d'améliorer la prise en charge des patients, la connaissance de la biologie tumorale thymique et l'identification de potentielles cibles thérapeutiques sont des voies de recherche importantes pour améliorer la survie des patients. Nous avons développé des cultures de cellules épithéliales thymiques dérivées in vitro de 12 TET (11 thymomes A, AB ou B et un carcinome thymique), caractérisées par leur potentiel prolifératif et leur expression de cytokératine. La voie PI3K / Akt / mTOR joue un rôle clé dans de nombreux cancers ; plusieurs études de phases I / II ont rapporté un effet positif des inhibiteurs de mTOR pour le contrôle de l'évolution du thymome chez les patients. Nous avons mis en évidence l'expression et l'activation des effecteurs mTOR, Akt et P70S6K dans les thymomes et dans les cellules épithéliales thymiques dérivées in vitro. Nous avons montré l'efficacité de la rapamycine, inhibiteur de mTOR, à réduire la prolifération cellulaire (30%) sans induire de mort cellulaire. Nos résultats suggèrent que l'activation de la voie Akt / mTOR participe à la prolifération cellulaire associée à la croissance tumorale. Nous avons établi un nouvel outil permettant l'étude de la dérégulation cellulaire au cours des thymomes. Dans un contexte de tumeurs rares, ces cellules permettront d'aborder des études mécanistiques in vitro et de tester l'efficacité de drogues anti tumorales / Human thymic epithelial tumors (TETs) are rare (250 – 300 cases/ year in France). We distinguish thymomas (A, AB and B subtypes) with indolent evolution (5 years actuarial survival in early stages >95%) and more aggressive thymic carcinomas (5 years actuarial survival <20% in stage IV). Surgical complete resection when feasible is the corner stone of a multimodal therapy and the most significant factor on survival. Relapse of TETs principally in pleura for thymomas (75%) and general for thymic carcinomas are difficult to treat. Therapeutics advances are limited given the lack of studies models of tumoral thymic epithelial cell. In a multidisciplinary approach for the treatment of metastatic pleural relapse of thymomas we developed an innovative surgical technique: cytoreductive pleurectomy associated with hyper thermic intra pleural chemotherapy (Cysplatin/ Mitomycin; 42°C) (ITCH). In selected patient (n=19), ITCH provides an efficient alternative to the morbid pleuro pneumonectomy. The median of free disease survival was 53 months, one year and five years actuarial survival were respectively 93% and 86%. However, the effectiveness of ITCH procedure questions on the played role ok hyperthermia, on the choice of chemotherapy association. With the aim to improve TETs therapies, the knowledge of TETs biology to identify potential target therapies is currently challenging. We developed an in vitro study model of tumoral thymic epithelial cells derived from 12 TETs (11 A, AB and B thymomas and one thymic carcinoma) characterized by their proliferative abilities and the cytokeratin expression. The PIK3 / Akt / mTOR pathway is implicated in numerous cancers. Several phase I, II studies advocate the potential role of mTOR inhibitors in the control of the metastatic disease. We highlighted the expression and the activation of mTOR, Akt and P70S6K effectors in TETs and in thymic epithelial cells in vitro derived. We showed the efficacy of rapamycin (mTOR inhibitor) in the inhibition (-30%) of in vitro cell proliferation without cell death induction. Our results suggest the implication of the PIK3 / Akt / mTOR pathway in the tumoral cell growth. We established a new tool to study cell dysregulation in TETs. In the context of rare tumors, these cells could allow in vitro mechanistic studies and test the efficacy of new anti tumoral therapies
6

Examining how PSMB11 orchestrates T cell development

Apavaloaei, Anca 08 1900 (has links)
No description available.
7

Reconstitution de l’architecture thymique et de la différenciation des cellules T dans les immunodéficiences génétiques : développement de stratégies thérapeutiques ciblant directement le thymus / Reconstitution of thymus architecture and T cell differentiation in genetic immunodeficiencies : development of therapeutic strategies directly targeting the thymus

Pouzolles, Marie 14 September 2018 (has links)
Les cellules souches hématopoïétiques (CSH) assurent la génération de toutes les lignées sanguines. Leur différenciation en cellules T matures se déroule dans un microenvironnement spécialisé, le thymus, orchestrée par des interactions complexes entre cytokines, chimiokines et cellules stromales. Les mutations bloquant la différenciation des cellules T ont un impact sur l'architecture du thymus, soulignant l’importance des interactions entre cellules T en développement et cellules stromales thymiques.Les déficits immunitaires combinés sévères sont généralement traités, avec succès, par transplantation de CSH allogénique par voie intraveineuse. Cependant, des complications peuvent survenir notamment en cas de greffe non compatible. Pour pallier à cela, la thérapie génique a été développée mais son efficacité et son innocuité restent à améliorer. Dans ce but, notre groupe a développé une approche par correction génique des progéniteurs T directement in vivo, via un vecteur lentiviral. Bien qu’efficace, là encore, l’efficacité de traitement reste insuffisante voire extrêmement limitée chez les macaques.Lors de ma thèse, j'ai donc évalué le potentiel de différents sérotypes de vecteur viraux adéno-associés (AAV) pour la transduction des thymocytes. L'administration IT de plusieurs sérotypes de AAV2 engendre une transduction des thymocytes >10 fois plus élevée que celle des vecteurs lentiviraux. Le sérotype AAV2/8 induit la transduction des thymocytes la plus efficace et les cellules transduites représentent jusqu'à 1% des cellules T périphériques d’une souris immunocompétente. En utilisant des souris immunodéficientes ZAP-70-/- comme paradigme, j'ai découvert que l'injection IT de l’AAV2/8-ZAP-70 entraîne une transduction et différentiation lymphocytaire T rapide, associée à la génération d’une medulla thymique. En effet, des cellules épithéliales thymiques de la médulla (mTEC) exprimant le régulateur auto-immun AIRE sont détectées en <2 semaines. Bien que cette reconstitution soit transitoire, les mTECs AIRE+ diminuant 10 semaines post-injection, les cellules T périphériques corrigées persistent >40 semaines et présentent environ 1 copie du vecteur AAV/cellule. Ces cellules T effectrices peuvent sécréter des niveaux élevés de cytokines et un nombre important de cellules T régulatrices est également généré. Ainsi, une seule vague de thymopoïèse à partir de progéniteurs transduits par l’AAV-ZAP-70, permet une restauration, rapide et transitoire de l'architecture thymique mais, à long terme de cellules T périphériques fonctionnelles.Pour évaluer les diverses populations de TEC régissant le développement et la sélection des cellules T, j'ai collaboré avec les groupes de P Jay/J Abramson/I Amit pour établir une cartographie de novo du compartiment stromal thymique. Nos analyses ont mis en évidence quatre populations majeures de mTEC (I-IV) avec des fonctions distinctes. Notamment, les mTEC-IV constituent une population unique présentant des similarités moléculaires et morphologiques avec les cellules tuft intestinales. Comme nous avions précédemment identifié la sécrétion d'IL-25 par les cellules tufts comme un régulateur des interactions entre compartiment épithélial et hématopoïétique dans l'intestin, nous avons évalué ce potentiel dans le thymus. Ainsi, des souris déficientes en cellules tuft intestinales présentent également une déficience spécifique en mTEC-IV et une homéostasie perturbée de diverses populations exprimant l'IL-25R dans le thymus. Notre recherche a donc permis d'identifier une nouvelle population de TEC tuft avec un rôle critique dans la formation de la niche immunitaire du thymus.L’ensemble de mes résultats montrent le potentiel thérapeutique de stratégie intrathymique de thérapie génique pour des patients ayant besoin d’une reconstitution rapide en cellules T et fournissent de nouvelles perspectives sur les populations stromales thymique et leur rôle dans l’équilibre de la niche immunitaire. / Hematopoietic stem cells (HSC) ensure the generation of all blood lineages. Their differentiation to mature T lymphocytes occurs in the specialized microenvironment of the thymus, orchestrated by complex interactions between cytokines, chemokines, and stromal cells. Mutations resulting in a block in T cell differentiation impact on the architecture of the thymus, pointing to the critical crosstalk between developing T cells and thymic stromal components.Genetic severe combined immunodeficiencies (SCID) are generally treated by the intravenous transplantation of healthy allogeneic HSCs. Although this therapy is often successful, complications can occur, especially for patients receiving non-histocompatible HSC transplants. To circumvent these problems , significant efforts have gone into developing gene therapy strategies but adverse events indicate the necessity of exploring other avenues. Our group hypothesized that in situ gene correction of T lymphoid progenitors in the thymus itself may overcome some of the drawbacks of ex vivo gene therapy. While intrathymic (IT) lentiviral vector administration corrected immunodeficient thymocyte precursors in mice, thymus transduction was inefficient and efficacy in macaques was limited.During my PhD, I assessed the in vivo potential of adeno-associated vectors (AAV) to transduce thymocyte precursors. Intrathymic administration of several different scAAV2 serotypes resulted in a >10-fold higher transduction of thymocytes (3-5%) as compared to lentiviral vectors. scAAV2/8 promoted the highest level of gene transfer and strikingly, transduced cells represented up to 1% of peripheral T lymphocytes in immunocompetent mice. Using ZAP-70-/- immunodeficient mice as a paradigm, I found that IT injection of an AAV2/8-ZAP-70 vector resulted in a rapid transduction and T cell differentiation, correlating with a dramatic generation of the thymus medulla. Indeed, medullary thymic epithelial cells (mTEC) expressing the AIRE autoimmune regulator were detected within <2 weeks. While this reconstitution was transient––AIRE+ mTECs decreased by 10 weeks post gene transfer––gene-corrected peripheral T cells, harboring approximately 1 AAV genome/ cell, persisted for >40 weeks. Effector T cells had the potential to secrete high levels of cytokines and significant numbers of gene-corrected regulatory T cells were also generated. Thus, a single wave of thymopoiesis, from intrathymic AAV-ZAP-70-transduced progenitors, allows for a rapid but transient restoration of the thymic architecture and long-term peripheral T cell function.To better assess the diverse TEC populations that orchestrate T cell development and selection, I collaborated with the groups of P. Jay/J. Abramson/I. Amit to combine single cell analysis and in-vivo fate-mapping to de novo characterize the entire stromal compartment of the thymus. Our analyses highlighted four major medullary TEC (mTEC I-IV) populations with distinct lineage regulator function and specifically, we found that mTEC-IV constitutes a highly divergent TEC subset that bears strong molecular and morphological characteristics to intestinal tuft cells. As we previously identified tuft cell secretion of IL-25 as a regulator of the crosstalk between the epithelial and hematopoietic compartments in the gut, we assessed the potential immune-modulatory function of mTEC-IV. Notably, mice deficient in intestinal tuft cells exhibited a specific depletion of mTEC-IV and a perturbed homeostasis of various IL25-R-expressing populations in the thymus. Taken together, our data identify a new tuft TEC population critical for shaping the thymus immune niche.In conclusion, the data generated during my PhD advance the therapeutic potential of intrathymic-based vector strategies for the treatment of patients requiring a rapid T cell reconstitution and provide new insights into thymic stromal subsets that are critical for shaping the thymus immune niche.
8

Role of EFNBs and EphB4 in T cell development and function

Jin, Wei 08 1900 (has links)
Eph kinases are the largest family of cell surface receptor tyrosine kinases. The ligands of Ephs, ephrins (EFNs), are also cell surface molecules. Ephs interact with EFNs and the receptors and ligands transmit signals in both directions, i.e., from Ephs to EFNs and from EFNs to Ephs. Ephs and EFNs are widely involved in various developmental, physiological pathophysiological processes. Our group and others have reported the roles of Ephs/EFNs in the immune system. To further investigate the function of EphBs/EFNBs in T cell development and responses, we generated EFNB1, EFNB2, EphB4 conditional gene knockout (KO) mice and EFNB1/2 double KO mice. In the projects using EFNB1 and EFNB2 knockout mice, we specifically deleted EFNB1 or EFNB2 in T cells. The mice had normal size and cellularity of the thymus and spleen as well as normal T cell subpopulations in these organs. The bone marrow progenitors from KO mice and WT mice repopulated the host lymphoid organs to similar extents. The activation and proliferation of KO T cells was comparable to that of control mice. Naïve KO CD4 cells differentiated into Th1, Th2, Th17 and Treg cells similar to naïve control CD4 cells. In EFNB2 KO mice, we observed a significant relative increase of CD4CD8 double negative thymocytes in the thymus. Flowcytometry analysis revealed that there was a moderate increase in the DN3 subpopulation in the thymus. This suggests that EFNB2 is involved in thymocyte development. Our results indicate that the functions of EFNB1 and EFNB2 in the T cell compartment could be compensated by each other or by other members of the EFN family, and that such redundancy safeguards the pivotal roles of EFNB1 and EFNB2 in T cell development and function. In the project using EFNB1/B2 double knockout (dKO) model, we revealed a novel regulatory function of EFNb1 and EFNb2 in stabilizing IL-7Rα expression on the T cell surface. IL-7 plays important roles in thymocyte development, T cell homeostasis and survival. IL-7Rα undergoes internalization upon IL-7 binding. In the dKO mice, we observed reduced IL-7Rα expression in thymocytes and T cells. Moreover, the IL-7Rα internalization was accelerated in dKO CD4 cells upon IL-7 stimulation. In T cell lymphoma cell line, EL4, over-expression of either EFNB1 or EFNB2 retarded the internalization of IL-7Rα. We further demonstrated compromised IL-7 signaling and homeostatic proliferation of dKO T cells. Mechanism study using fluorescence resonance energy transfer and immunoprecipitation demonstrated that physical interaction of EFNB1 and EFNB2 with IL-7Rα was likely responsible for the retarded IL-7Rα internalization. In the last project, using medullary thymic epithelial cell (mTEC)-specific EphB4 knockout mice, we investigated T cell development and function after EphB4 deletion in mTEC. EphB4 KO mice demonstrated normal thymic weight and cellularity. T cell development and function were not influenced by the EphB4 deletion. Lastly, the KO mice developed normal delayed type hypersensitivity. Overall, our results suggest that comprehensive cross interaction between Eph and EFN family members could compensate function of a given deleted member in the T cell development, and only simultaneous deletion of multiple EFNBs will reveal their true function in the immune system. In fact, such redundancy signifies vital roles of Ephs and EFNs in the immune system. / Kinases Eph est la plus grande famille de tyrosines kinases récepteurs Éphrines (EFN) est un ligand de Ephs. Eph et EFN sont toutes les molécules de surface cellulaire. L’interaction entre Ephs et EFNs permet de transmettre des signaux dans les deux directions (c.-à-d. partir de Ephs à EFNs, et de EFNs à Ephs.) Eph et EFNs sont largement impliqués dans divers processus développementaux, physiologiques et physiopathologiques. Notre groupe et d'autres groupes ont rapporté les rôles de Ephs / EFNs dans le système immunitaire. Pour approfondir la fonction de EphBs / EFNBs dans le développement des lymphocytes T et des réponses immunitaires, nous avons généré des souris EFNB1, EFNB2, et EphB4 knock-out conditionnel (KO) et des souris EFNB1 / 2 doubles KO. Dans les projets qui utilisent EFNB1 et EFNB2 comme souris knock-out, nous avons spécifiquement supprimé EFNB1 ou EFNB2 dans les cellules T. Les souris présentaient une taille normale, la cellularité du thymus et de la rate, ainsi que des sous-populations de cellules T étaient normales dans ces organes. Les progéniteurs de la moelle osseuse de souris KO et les souris WT ont repeuplé les organes lymphoïdes de l’hôte à des degrés similaires. L'activation et la prolifération des cellules KO T étaient comparables à celles des souris témoins. Les cellules CD4 naïves KO différenciées en Th1, Th2, Th17 et Treg étaient similaires aux cellules CD4 naïves de souris contrôle. Chez les souris KO EFNB2, nous avons observé une augmentation relative importante des thymocytes CD4CD8 : les double négatifs dans le thymus. L'analyse par cytométrie en flux a révélé qu'il y avait une augmentation modérée de la sous-population DN3 dans le thymus. Les résultats suggèrent qu’EFNB2 est impliqué dans le développement des thymocytes. Nos résultats indiquent que les fonctions de EFNB1 et EFNB2 dans le compartiment des cellules T pourraient être compensées entre eux ou par d'autres EFNB. La redondance des fonctions suggèrent le contrôle critique d’EFNB1 et EFNB2 dans le développement des cellules T. Dans le projet, en utilisant EFNB1/B2 (modèle double KO) (dKO), nous avons observé une fonction de régulation de EFNB1 et EFNB2. dans la stabilisation de l’expression l'IL-7R α , à la surface des cellules T, IL-7 joue un rôle important dans le développement des thymocytes, l'homéostasie des lymphocytes T , et leur survie. IL-7R α subit une internalisation i contraignante de IL-7. Chez les souris DKO, nous avons observé une perte d’expression de l’ IL-7Rα dans les thymocytes et les cellules T. En outre, l’ internalisation IL-7Rα a été accélérée dans les cellules CD4 dKO, suite à la stimulation IL-7. Dans la lignée cellulaire de lymphome T, EL4, la surexpression de EFNB1 ou EFNB2 retarde l'internalisation de l'IL-7Rα. Nous avons aussi démontré les signalisations compromises de l’ IL-7 et de la prolifération homéostatique des cellules T dKO. Les études du méchanisme qui utilisent la fluorescence de transfert d'énergie par résonance et immunoprécipitation ont montré que l'interaction physique de EFNB1 et EFNB2 avec IL-7R était probablement responsable du retard de l’ internalisation IL-7Rα. Dans le dernier projet, nous avons étudié le développement des cellules T et la fonction des cellules épithéliales médullaires du thymus (mTEC), chez les souris knock-out EphB4. Les souris KO EphB4 ont démontré un poids et une cellularité qui sont normaux. La fonction et le développement de cellules T ne sont pas influencés par la suppression de l’ EphB4. Enfin, les souris KO ont développé une hypersensibilité de type retardée normale. Dans l'ensemble, nos résultats suggèrent que l'interaction globale de croisement entre Eph et les membres de la famille EFN pourrir compenser la fonction d'un membre supprimé. Seule la suppression simultanée de plusieurs EFNBs va révéler leur vraie fonction dans le système immunitaire. En fait, une telle redondance montre les rôles vitaux d’Ephs et EFNS dans le système immunitaire.
9

O gene Aire pode controlar mRNAs bem como os lncRNAs em células tímicas epiteliais medulares como evidenciado pela edição do genoma por CRISPR-Cas9 / Aire gene can control mRNAs as well as lncRNAs in medullary thymic epitelial cells as evidentiated by genome editing by CRISPR-Cas9

Duarte, Max Jordan de Souza 26 November 2018 (has links)
O timo é um órgão linfoide primário essencial para a manutenção da tolerância central através da seleção e eliminação de células T autoreativas. Precursores de células T, oriundas da medula óssea, chegam ao timo e migram do córtex para região da medula. As células epiteliais medulares tímicas (mTECs) expressam em sua superfície antígenos de tecidos periféricos (em inglês tissue-restricted antigens ou TRAs) que representam autoantígenos de todos os tecidos do corpo. Atuando como um fator de transcrição não clássico em células mTEC, o gene Autoimmune Regulator (Aire) desempenha um papel na expressão dos TRAs, cuja proteína codificada libera a RNA polimerase II (RNA Pol II) ancorada na cromatina e regula a expressão de mRNAs na glândula timo. A função biológica deste gene está ligada à indução de tolerância imunológica central impedindo o aparecimento de doenças autoimunes. Isso é resultado da seleção negativa de timócitos (precursores de células T) autoreativos que interagem fisicamente com as mTECs. Os timócitos autoreativos que reconhecem os TRAs como elementos estranhos são eliminados por apoptose. O co-cultivo de mTECs com timócitos representa um sistema-modelo in vitro adequado para se aproximar da interação celular que ocorre dentro do timo. Os resultados anteriores do nosso laboratório demonstraram que além do controle de mRNA de TRAs, o gene Aire também participa da modulação de miRNAs em mTECs uma vez que estas espécies de RNA são transcritas pela RNA Pol II. Continuando com essa linha de estudos, neste trabalho nós demonstramos pela primeira vez que Aire também modula a expressão de long noncoding RNAs (lncRNAs) em mTECs. Para isto fizemos uso da estratégia da perda de função analisando a expressão dessa espécie de RNA, assim como de mRNAs, em células mTEC Aire +/+ e mTEC Aire nocautes (KO Aire -/-) obtidas pela edição gênica por Crispr-Cas9. O transcriptoma dessas células que passaram ou não por adesão com timócitos, foi então analisado por hibridizações com microarrays. Isso evidenciou que Aire e adesão celular influenciam a expressão tanto de mRNAs como de lncRNAs. A reconstrução de redes de interação lncRNAs-mRNAs possibilitou evidenciar uma nova via de regulação pós-transcricional em células mTEC. / The thymus is a primary lymphoid organ essential for the maintenance of central tolerance through the selection and elimination of autoreactive T cells. Precursors of T cells, originating from the bone marrow, reach the thymus and migrate from the thymic cortex to the medullary region. Thymic medullary epithelial cells (mTECs) express on their surface tissue-restricted antigens (TRAs) that represent autoantigens of all tissues in the body. Acting as a non-classical transcription factor in mTEC cells, the Autoimmune regulator (Aire) gene plays a role in the expression of TRAs, whose encoded protein releases the RNA polymerase II (RNA Pol II) anchored in the chromatin and regulates the expression of mRNAs in the thymus gland. The biological function of this gene is associated to the induction of central immune tolerance preventing the onset of autoimmune diseases. This is a result of negative selection of autoreactive thymocytes (T cell precursors) that interact physically with mTECs. Self-reactive thymocytes that recognize TRAs as foreign elements are eliminated by apoptosis. The co-culture of mTECs with thymocytes represents an appropriate in vitro model system to approximate the cellular interaction that occurs within the thymus. Previous results from our laboratory demonstrated that in addition to the control of TRA mRNAs, Aire also participates in the modulation of miRNAs in mTECs since these RNA species are transcribed by RNA Pol II. Continuing with this line of studies, in this study we demonstrate for the first time that Aire also modulates the expression of long non-coding RNAs (lncRNAs) in mTECs. For this, we used the loss-of-function strategy to analyze the expression of this RNA species, as well as mRNAs in mTEC Aire + / + or Aire knockout mTEC cells (KO Aire - / -) obtained by the gene editing by Crispr-Cas9. The transcriptome of these cells, whether or not adhered to thymocytes, was then analyzed by microarray hybridizations. This demonstrated that Aire and cell adhesion influence the expression of both mRNAs and lncRNAs. The reconstruction of lncRNAs-mRNAs interaction networks made possible to evidence a new post-transcriptional regulation pathway in mTEC cells.
10

A função do gene Autoimmune Regulator (Aire) no controle da adesão de células tímicas epiteliais medulares com timócitos / The fuction of Autoimmune Regulator (Aire) gene in the control of adhesion between medullary thymic epithelial cells with thymocytes

Pezzi, Nicole 26 February 2016 (has links)
O crosstalk entre timócitos e células epiteliais tímicas é crucial para o desenvolvimento das células T e estabelecimento da tolerância central. Células tímicas epiteliais medulares (mTECs) contribuem para a autotolerância por meio da expressão ectópica de antígenos restritos aos tecidos (TRAs). A expressão de TRAs em mTECs é altamente dependente do gene Autoimmune Regulator (Aire). Por meio do reconhecimento de TRAs com alta afinidade, células T autoreativas são selecionadas negativamente do pool de timócitos em desenvolvimento. Apesar do papel de Aire na indução da tolerância central ser bem conhecido, os mecanismos celulares e moleculares precisos do processo permanecem obscuros. Nesse estudo, hipotetizamos que perturbações na expressão do gene Aire influenciam a adesão entre mTECs e timócitos, o que poderia resultar em um desequilíbrio na imunotolerância a antígenos próprios. Um ensaio funcional realizado com timócitos frescos, extraídos de um timo normal de camundongo e cocultivados com células epiteliais tímicas medulares da linhagem mTEC 3.10, demonstrou que a inibição do gene Aire por meio de RNA de interferência reduziu significativamente a capacidade das mTECs de promover a adesão dos timócitos. Análises por microarray revelaram que o silenciamento do gene Aire nas células mTEC 3.10 causou a modulação de mais de 1000 genes, alguns que codificam TRAs, outros que codificam proteínas envolvidas na adesão celular, como VCAM-1, e também outros que codificam moléculas coestimuladoras como CD80. Esses resultados contribuem para uma melhor compreensão do papel de Aire no controle da adesão mTEC-timócitos, a qual constitui um processo essencial para a seleção negativa de timócitos autoreativos / The crosstalk between thymocytes and thymic epithelial cells is critical for T cell development and the establishment of central tolerance. Medullary thymic epithelial cells (mTECs) contribute to self-tolerance through the ectopic expression of tissuerestricted antigens (TRAs) in the thymus. TRAs expression in mTECs is largely dependent on Autoimmune Regulator (Aire) gene. Through the recognition of TRAs with high affinity, developing autoreactive T cells are negatively select from the pool of developing thymocytes. Although the role of Aire in the induction of central tolerance is well known, the precise cellular and molecular mechanisms remain unclear. In this study, we hypothesize that disturbance in Aire gene expression influences adhesion between mTECs and thymocytes, which could result in an imbalance in immune-tolerance to self-antigens. A functional assay performed with fresh thymocytes dissociated from a normal mouse thymus and co-cultured with a medullary thymic epithelial cell line named mTEC 3.10, demonstrated that Aire RNAi knockdown significantly decreased the ability of mTECs to promote thymocyte adhesion. Microarray analysis revealed that Aire knockdown of the murine mTEC 3.10 cell line led to the modulation of more than 1000 genes, some of them coding for TRAs, others for proteins involved in cell adhesion like VCAM-1 and also for costimulatory molecules like CD80. These results contribute to a better understanding of the role of Aire in the control of mTEC-thymocyte adhesion, which is an essential process for negative selection of autoreactive thymocytes

Page generated in 0.0877 seconds