• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 10
  • 7
  • 2
  • 2
  • Tagged with
  • 23
  • 11
  • 10
  • 9
  • 5
  • 5
  • 5
  • 4
  • 4
  • 4
  • 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.
11

Le récepteur co-inhibiteur BTLA au cours du lupus érythémateux disséminé (LED) : aspects fondamentaux et implications thérapeutiques / The co-inhibitory receptor BTLA in SLE : fundamental aspects and therapeutic implications

Sawaf, Matthieu 26 April 2018 (has links)
Le lupus érythémateux disséminé (LED) est une maladie auto-immune systémique caractérisée par une inflammation provoquant des lésions dans de nombreux organes tels que les reins, les poumons ou la peau. Dans cette pathologie, une activation excessive du système immunitaire conduit à la production d’auto-anticorps dirigés, le plus souvent, contre du matériel nucléaire. La différenciation des lymphocytes B (LB) en cellules productrices d’anticorps requiert une communication entre les LT et les LB. Ce dialogue est régulé par de nombreux acteurs cellulaires et moléculaires afin de permettre la mise en place d’une réponse humorale efficace en cas d’infections, mais aussi de prévenir le développement de maladies auto-immunes. Mon projet de thèse a consisté à étudier l’implication de deux de ces acteurs, l’un favorisant la différenciation des LB en plasmocytes, à savoir, les cellules T folliculaires auxiliaires (TFH) et le second régulant négativement l’activation lymphocytaire, le récepteur co-inhibiteur BTLA (pour B and T Lymphocyte Attenuator) dans le LED chez l’Homme. Au cours de cette étude, nous avons d’une part amélioré les connaissances concernant les sous-populations de TFH circulantes humaines, en décrivant que parmi les cellules TFH CXCR3-CCR6- sont retrouvées des cellules aux propriétés suppressives. De plus, nous avons suggéré que la contraction des TFH1 (CXCR3+CCR6-) au profit des TFH2 (CXCR3-CCR6-), observées chez les patients lupiques, pourrait être le reflet d’une migration des TFH1 vers les organes inflammés. D’autre part, nous avons mis en évidence un défaut fonctionnel de BTLA dans les LT CD4+ de patients lupiques. Ce défaut, restauré en normalisant le métabolisme lipidique des LT CD4+, semble associé à la sévérité de la pathologie. En parallèle de ces observations, nous avons démontré un défaut d’expression de BTLA sur les LB et les LT CD4+ régulateurs de patients lupiques. L’ensemble de nos données sont prometteuses et ouvrent de nouvelles perspectives thérapeutiques pour le traitement du LED. / Systemic Lupus Erythematosus (SLE) is an autoimmune disease characterized by lesions in several organs such as kidneys, lungs and skin for instance. In this pathology, an excessive activation of the immune system leads to the production of autoantibodies targeting mainly nuclear antigens. B cell differentiation into antibody-secreting cells requires a close collaboration between T and B cells. This cross-talk is regulated by various cellular and molecular factors in order to mount an efficient humoral response in case of infection, but also to prevent autoimmune disease development. The aim of my thesis was to study two regulating factors of the B cell response, one promoting the B cell differentiation into plasma cells, i.e the follicular helper T cells (TFH) and the other one inhibiting lymphocyte activation, i.e a co-inhibitory receptor called BTLA (« B and T Lymphocyte Attenuator ») in human SLE. In this study, we first improved our knowledge concerning human circulating TFH cells, by describing among the CXCR3-CCR6- TFH cell subset, a population with suppressive capacities. Moreover, we suggested that the decreased frequency of TFH1 in lupus patients’ blood could be explained by the migration of these cells into inflamed tissues. We also highlighted a BTLA functional deficiency in lupus CD4+ T cells. This deficiency, which can be restored by normalizing the lipid metabolism, seems to be associated to disease severity. Furthermore, we described an altered expression of BTLA in lupus B cells and regulatory T cells. Altogether, our data show promising results and suggest new potential therapeutic strategies for lupus treatment.
12

Efeitos da sinalização purinérgica durante a infecção aguda e crônica pelo Plasmodium chabaudi AS. / Effects of purinergic signaling during acute and chronic infections by Plasmodium chabaudi AS.

Érika Machado de Salles 14 October 2016 (has links)
A malária permanece um sério problema de saúde em países subdesenvolvidos. O estágio sanguíneo da infecção é responsável por todos os sintomas associados com a malária. Recentemente, tem sido mostrado que receptores imunes inatos são capazes de detectar sinais de dano, tais como a adenosina trifosfato ATP. O receptor P2X7 detecta altas concentrações de ATP extracelular. Ao avaliarmos a parasitemia e os parâmetros clínicos da doença em camundongos C57BL/6 e P2X7-/-, observamos uma semelhança em ambos os grupos até o dia 7 p.i., mas após este período os camundongos P2X7-/- tiveram dificuldade de controlar a parasitemia e restaurar os parâmetros clínicos. O ineficiente controle da parasitemia durante o período agudo e crônico em camundongos P2X7-/- foi associado com a baixa produção de IFNγ. Além disso, o receptor P2X7 aumenta a expressão de T-bet em células Th1 e controla o número de células Tfh. Este estudo mostra que o equilíbrio mediado pelo receptor P2X7 entre os fatores de transcrição Bcl-6 e T-bet ajusta a imunidade celular e humoral na malária. / Malaria remains a serious healthcare problem in developing countries. The blood stage of infection is responsible for all symptoms associated with malaria. Recently, it has been shown that innate immune receptors are able to detect signals as adenosine triphosphate (ATP). P2X7 receptor detects high levels of extracellular ATP. Evaluating the parasitemia and clinical parameters in C57BL/6 (B6) and P2X7-/- mice, we observed a similarity in both groups to day 7 p.i., but after this period the P2X7-/- mice had difficulty in controlling the parasitemia and restoring the clinical parameters. The inefficient parasite control in acutely and chronically infected P2X7-/- mice was associated with low production of IFNγ. Furthermore, P2X7 receptor increases the expression of T-bet in Th1 cells and controls the Tfh cell number. This study provides a new insight into immunology by showing that the balance between T-bet and Bcl-6 transcriptional factors tunes the cellular and humoral immunity in malaria.
13

Quelle contribution du centre germinatif et de ses composants moléculaires et cellulaires dans la physiopathologie du lupus ? / What contribution for molecular and cellular germinal center components during lupus development?

Le Coz, Carole 19 September 2014 (has links)
Le lupus érythémateux disséminé est une maladie auto-immune systémique très invalidante dont les atteintes sont multiples, les plus fréquentes étant cutanées, articulaires et rénales. Dans ce type de maladie, le système immunitaire, hyperactif, ne se limite pas à lutter contre des agents extérieurs mais s'attaque à ses propres cellules, entre autres par le biais d'auto-anticorps. Ces anticorps délétères sont produits par des plasmocytes, cellules issus de la différenciation des lymphocytes B. Ce processus se déroule principalement au sein des centres germinatifs (GC) dans les organes lymphoïdes secondaires, et fait intervenir de nombreux acteurs moléculaires et cellulaires. Mon projet de thèse a porté sur l'étude de la contribution du GC et de ses constituants, tels que les cellules auxiliaires folliculaires (Tfh) et l'IL-21, au cours du lupus. Au cours de ce travail, nous avons mis en évidence une altération à la fois quantitative et qualitative des cellules Tfh chez des patients lupiques et dans un modèle murin, altération entre autres responsable de taux anormalement élevés d'IL-21. Nous avons également observé une sensibilité accrue des cellules B de souris lupiques à cette cytokine, dont la cause est une surexpression de molécules clés telles que STAT3, et dont la conséquence est un surcroit de différenciation plasmocytaire. Tous les éléments sont donc présents pour favoriser l'interaction "Tfh-B" et la réaction du GC, et amplifier la réponse autoimmune. Enfin, la découverte de l'existence de GC ectopiques fonctionnels dans les reins de souris lupiques permet d'envisager l'existence de réponses locales au sein même des organes cibles. Les données obtenues, fondamentales, sont prometteuses et laissent entrevoir de nouvelles perspectives de biothérapies, plus ciblées, pour le traitement de la maladie lupique. / Systemic lupus erythematosus is a disabling chronic autoimmune disease characterized by B cell hyperactivity leading to the production of autoantibodies, some of which exerting pathogenic effects. Autoantibodies are produced by plasma cells, which originate from the differentiation of B cells through a process that mainly takes place in germinal centers (GC) in secondary lymphoïd organs and involves many molecular and cellular parameters. The aim of my PhD project was to analyze the individual contribution of GC components, such as follicular helper T cells (Tfh) and IL-21, to lupus development. During this work, we have shown both a quantitative and qualitative impairment of Tfh cells in lupus patients and in a mouse model, leading, among other things, to high IL-21 levels. We also observed that B cells from lupus mice display a specific intrinsic sensitivity to this cytokine, due to over-expression of key molecules such as STAT3, which results in increased plasma cell differentiation. Thus, all elements are gathered that favor "Tfh-B" cell interactions and the GC reaction, and therefore the autoimmune response. Finally, the discovery of functional ectopic GC in the kidneys of lupus mice allows envisaging that local responses occur within the target organs and likely participate to kidney injury. The fundamental data we obtained are promising and anticipate new and better targeted biotherapies for lupus treatment.
14

Molecular Insights of CD4+ T Cell Differentiation, Effector Formation and Helper Function

Liu, Siqi January 2016 (has links)
<p>CD4+ T cells play a crucial in the adaptive immune system. They function as the central hub to orchestrate the rest of immunity: CD4+ T cells are essential governing machinery in antibacterial and antiviral responses by facilitating B cell affinity maturation and coordinating the innate and adaptive immune systems to boost the overall immune outcome; on the contrary, hyperactivation of the inflammatory lineages of CD4+ T cells, as well as the impairments of suppressive CD4+ regulatory T cells, are the etiology of various autoimmunity and inflammatory diseases. The broad role of CD4+ T cells in both physiological and pathological contexts prompted me to explore the modulation of CD4+ T cells on the molecular level.</p><p>microRNAs (miRNAs) are small RNA molecules capable of regulating gene expression post-transcriptionally. miRNAs have been shown to exert substantial regulatory effects on CD4+ T cell activation, differentiation and helper function. Specifically, my lab has previously established the function of the miR-17-92 cluster in Th1 differentiation and anti-tumor responses. Here, I further analyzed the role of this miRNA cluster in Th17 differentiation, specifically, in the context of autoimmune diseases. Using both gain- and loss-of-function approaches, I demonstrated that miRNAs in miR-17-92, specifically, miR-17 and miR-19b in this cluster, is a crucial promoter of Th17 differentiation. Consequently, loss of miR-17-92 expression in T cells mitigated the progression of experimental autoimmune encephalomyelitis and T cell-induced colitis. In combination with my previous data, the molecular dissection of this cluster establishes that miR-19b and miR-17 play a comprehensive role in promoting multiple aspects of inflammatory T cell responses, which underscore them as potential targets for oligonucleotide-based therapy in treating autoimmune diseases. </p><p>To systematically study miRNA regulation in effector CD4+ T cells, I devised a large-scale miRNAome profiling to track in vivo miRNA changes in antigen-specific CD4+ T cells activated by Listeria challenge. From this screening, I identified that miR-23a expression tightly correlates with CD4+ effector expansion. Ectopic expression and genetic deletion strategies validated that miR-23a was required for antigen-stimulated effector CD4+ T cell survival in vitro and in vivo. I further determined that miR-23a targets Ppif, a gatekeeper of mitochondrial reactive oxygen species (ROS) release that protects CD4+ T cells from necrosis. Necrosis is a type of cell death that provokes inflammation, and it is prominently triggered by ROS release and its consequent oxidative stress. My finding that miR-23a curbs ROS-mediated necrosis highlights the essential role of this miRNA in maintaining immune homeostasis. </p><p>A key feature of miRNAs is their ability to modulate different biological aspects in different cell populations. Previously, my lab found that miR-23a potently suppresses CD8+ T cell cytotoxicity by restricting BLIMP1 expression. Since BLIMP1 has been found to inhibit T follicular helper (Tfh) differentiation by antagonizing the master transcription factor BCL6, I investigated whether miR-23a is also involved in Tfh differentiation. However, I found that miR-23a does not target BLIMP1 in CD4+ T cells and loss of miR-23a even fostered Tfh differentiation. This data indicate that miR-23a may target other pathways in CD4+ T cells regarding the Tfh differentiation pathway.</p><p>Although the lineage identity and regulatory networks for Tfh cells have been defined, the differentiation path of Tfh cells remains elusive. Two models have been proposed to explain the differentiation process of Tfh cells: in the parallel differentiation model, the Tfh lineage is segregated from other effector lineages at the early stage of antigen activation; alternatively, the sequential differentiation model suggests that naïve CD4+ T cells first differentiate into various effector lineages, then further program into Tfh cells. To address this question, I developed a novel in vitro co-culture system that employed antigen-specific CD4+ T cells, naïve B cells presenting cognate T cell antigen and BAFF-producing feeder cells to mimic germinal center. Using this system, I were able to robustly generate GC-like B cells. Notably, well-differentiated Th1 or Th2 effector cells also quickly acquired Tfh phenotype and function during in vitro co-culture, which suggested a sequential differentiation path for Tfh cells. To examine this path in vivo, under conditions of classical Th1- or Th2-type immunizations, I employed a TCRβ repertoire sequencing technique to track the clonotype origin of Tfh cells. Under both Th1- and Th2- immunization conditions, I observed profound repertoire overlaps between the Teff and Tfh populations, which strongly supports the proposed sequential differentiation model. Therefore, my studies establish a new platform to conveniently study Tfh-GC B cell interactions and provide insights into Tfh differentiation processes.</p> / Dissertation
15

Influenza A virus induces regulated T cell-driven B cell responses

Boyden, Alexander Wiser 01 December 2012 (has links)
Protection from influenza A virus (IAV) challenge requires switched, high affinity Abs derived from long-lived memory B cells and plasma cells. These subsets are generated in germinal centers (GCs), hallmark structures of T helper cell-driven B cell immunity. A full understanding of the acute and persistent GC B cell reaction following respiratory IAV infection is lacking, as is the characterization of IAV-induced T follicular helper (TFH) cells that support GCs. Additionally, it remains unclear as to whether IAV-induced GC B cells are subject to control by regulatory T cells (Tregs). To address this, GC B cell and TFH cell responses were analyzed in mice following pulmonary challenge with IAV. Studies demonstrated that marked GC reactions were induced in lung-draining lymph nodes (dLNs), lung, spleen and nasal-associated lymphoid tissue (NALT), although the magnitude, kinetics, and isotype switching patterns of the response was site-specific, and largely depended on the magnitude of IAV-induced TFH cell populations. TFH cell magnitude peaked prior to that of GC B cells in all tissues, and TFH cells purified from dLNs generated IL-21 and IFN-gamma upon activation, although CD4+CXCR5- T effector cells produced higher levels of all cytokines. IgA+ GC B cells were infrequent in most sites, but composed a significant subset of the switched GC population in NALT. Further, splenectomized mice withstood a lethal recall challenge, suggesting the spleen to be unnecessary for long-term protection. Additionally, GC B cell populations were analyzed at distal time points to assess the understudied, persistent GC B cell response after IAV infection. Our analysis demonstrated that persistent GC B cell populations in mouse lungs directly correlated with infectious dose, pathogenicity of the virus, as well as the presence of long-term CD4+ T cell help. Finally, experiments showed that Tregs contribute to the control of GCs induced in the spleen by IAV challenge. This was demonstrated by a marked increase in the number of total and switched GC B cell numbers when Tregs were either depleted or disrupted in vivo proximal to IAV exposure.
16

The impact of age and gut microbiota on Th17 and Tfh cells in K/BxN autoimmune arthritis

Teng, Fei, Felix, Krysta M., Bradley, C. Pierce, Naskar, Debdut, Ma, Heqing, Raslan, Walid A., Wu, Hsin-Jung Joyce 15 August 2017 (has links)
Background: Age is an important risk factor for rheumatoid arthritis (RA), which often develops in middle age. However, how age-associated changes in immunity impact RA is poorly understood. Gut microbiota are known to be involved in the pathogenesis of RA, but the effects of microbiota in older subjects remain mostly unknown. Methods: We used segmented filamentous bacteria (SFB), a gut commensal species with immunomodulatory effects, and K/BxN mice, a T cell receptor (TCR) transgenic model, to study the effect of age and microbiota on autoimmune arthritis. Comparing young and middle-aged K/BxN T cells of the same TCR specificity allows us to study T cells with an age focus eliminating a key variable: TCR repertoire alteration with age. In addition to joints, we also studied pathological changes in the lung, an important extra-articular RA manifestation. We used flow cytometry to evaluate T follicular helper (Tfh) and T helper 17 (Th17) cells, as they both contribute to autoantibody production, a key disease index in both RA and K/BxN arthritis. Results: Middle-aged K/BxN mice had aggravated arthritis and pathological changes in the lung compared to young mice. Middle-aged mice displayed a strong accumulation of Tfh but not Th17 cells, and had defective Th17 differentiation and low expression of interleukin-23, a critical cytokine for Th17 maintenance. Although a soaring Tfh cell population accompanied by robust germinal center B cell responses were found in middle-aged mice, there was decreased cycling of Tfh cells, and SFB only induced the non-Tfh cells to upregulate Bcl-6, the Tfh master transcription factor, in the young but not the middle-aged group. Finally, the accumulated Tfh cells in middle-aged mice had an effector phenotype (CD62LloCD44hi). Conclusion: Age-dependent Tfh cell accumulation may play a crucial role in the increased autoimmune disease phenotype in middle-age. SFB, a potent stimulus for inducing Tfh differentiation, fails to promote Tfh differentiation in middle-aged K/BxN mice, suggesting that most of the middle-aged Tfh cells with an effector phenotype are Tfh effector memory cells induced at an earlier age. Our results also indicate that exposure to immunomodulatory commensals may allow the young host to develop an overactive immune system reminiscent of that found in the middle-aged host.
17

Au coeur du contrôle de l’immunité humorale : (re)définition, mode d’action et répertoire des lymphocytes T folliculaires régulateurs / The immune humoral response offers the organism an efficient protection through the production of antibodies following an immune stimulation

Ritvo, Paul-Gydéon 26 September 2017 (has links)
La réponse immunitaire humorale désigne l’ensemble des processus menant à la production d’anticorps suite à une stimulation du système immunitaire. Les lymphocytes T folliculaires régulateurs (Tfr) forment une population essentielle dans le contrôle de l’immunité humorale. Dotés de fonctions régulatrices comme les lymphocytes T régulateurs conventionnels (Treg), les Tfr joueraient un rôle majeur dans les mécanismes de régulation de la production d’anticorps suite à une stimulation. Ils pourraient, par exemple, contrôler l’aide apportée par les lymphocytes T folliculaires helpers (Tfh) aux cellules B leur permettant de se différencier en plasmocytes producteurs d’anticorps dans une structure hautement spécialisée appelée centre germinatif (CG). Suite au constat de la non-réponse des Tfr à l’interleukine (IL)-2, nous avons observé l’absence d’expression de la sous-unité du récepteur à l’IL-2 (CD25) sur ces cellules et ainsi donné de nouvelles bases à la caractérisation phénotypique des Tfr. Cette redéfinition restrictive de la population cellulaire a permis une description plus approfondie de ces cellules et la découverte d’un axe de régulation du CG dépendant de l’IL-1ß. La dualité de régulations observée entre d’une part l’axe Treg - lymphocytes T effecteurs contrôlé par l’IL-2 en dehors du CG et d’autre part l’axe Tfr - Tfh contrôlé par l’IL-1ß dans le CG nous a amenés à nous poser la question de l’origine et de la spécificité des Tfr en partie élucidée par une analyse du répertoire de ces populations. Nous avons mis en évidence une similarité en matière de distribution et caractéristiques globales des répertoires des cellules folliculaires qu’elles soient régulatrices (Tfr) ou non (Tfh). Il est également apparu une proximité de spécificités entre le répertoire des Treg et des Tfr confortant l’hypothèse d’une origine commune de ces deux populations. Ce travail de thèse a permis la découverte d’éléments importants de la biologie des Tfr, population impliquée dans le contrôle des régulations humorales. Il ouvre des perspectives relatives au contrôle de la production d’anticorps tant sa limitation - dans le contexte de maladies auto-immunes - que son exploitation dans le développement de stratégies vaccinales. / The immune humoral response offers the organism an efficient protection through the production of antibodies following an immune stimulation. Follicular regulatory T cell (Tfr) is an essential subset in the control of humoral immunity. These cells share with conventional regulatory T (Treg) cells regulatory functions and should play a major role in the control of antibody production following stimulation. As T follicular helper (Tfh) cells help is essential in the differentiation of B cell into antibody-producing plasma cells, one of the possible mechanisms of Tfr’s control could be the limitation of the Tfh cells’ help to the B cells. As a consequence of the non-response of Tfr cells to interleukin (IL)-2, we thoroughly revealed the CD25- phenotype of Tfr cells thus redefining the subset. This stringently-selected population allowed a fine-tuned characterization of Tfr cells and the discovery of an IL-1β axis regulating the germinal center responses. The dual regulation of T cells in secondary lymphoid organs, one between Treg and Teff cells regulated by IL-2 outside germinal centers and the other between Tfh and Tfr cells regulated by IL-1 inside GCs brought us to question the origin and specificity of Tfr cells. We partially answered this with a high-resolution analysis of these populations’ repertoires. We highlighted a similarity in the distributions and global characteristics of the follicular cells’ repertoires regardless their regulatory (Tfr) or not (Tfh) phenotype. We also brought out the major sharing between Treg and Tfr repertoires underpinning the hypothesis of a common origin for these populations. This work has disclosed important aspects of Tfr cells’ biology, a fundamental subset in the control of humoral immune responses. It opens many perspectives including the control of antibody production, negatively in the context of autoimmune diseases or its positive exploitation to enhance vaccine efficacy
18

Régulation des réponses immunes humorales par la voie de signalisation IL-6 / STAT3

Hercor, Mélanie 18 December 2015 (has links) (PDF)
L’aide fournie aux lymphocytes B par les lymphocytes T CD4+ est cruciale pour une réponse humorale de qualité. Les lymphocytes T helper folliculaires (Tfh) jouent un rôle majeur dans l’aide apportée aux lymphocytes B au sein des centres germinatifs et peu de choses sont connues sur la capacité d’aide des autres populations de cellules T CD4+. Le but de notre étude est d’évaluer la capacité d’aide aux lymphocytes B des lymphocytes T helper de type 2 (Th2), une population considérée, à l’origine, comme responsable de l’aide apportée aux lymphocytes B in vivo ainsi que d’analyser le rôle de l’axe IL6 / STAT3 dans la différenciation des lymphocytes Tfh et leur plasticité phénotypique. Nous montrons que les lymphocytes Th2 co-expriment des formes actives des facteurs de transcription STAT6 et STAT3 et que l’expression de STAT3 est requise pour la fonction d’aide aux lymphocytes B des lymphocytes Th2. Nous avons également pu montrer que la voie de signalisation IL-6 / STAT3 durant le développement des lymphocytes Tfh s’oppose à l’expression des gènes associés au programme de différenciation Th2. / Option Biologie moléculaire du Doctorat en Sciences / info:eu-repo/semantics/nonPublished
19

T cell factor-1 regulates CD4+ and CD8+ T cell responses in a stage-specific manner

Gullicksrud, Jodi Ann 01 August 2017 (has links)
CD4+ and CD8+ T cells are critical components of the adaptive arm of immune responses. During viral infection, CD8+ T cells utilize their cytotoxic function to kill infected cells and clear the infection. In addition, CD4+ T cells differentiate into either T helper 1 (Th1) or T follicular helper (Tfh) cells, which provide essential help to enhance the efficacy of other response immune cells, including macrophages, CD8+ T cells, and B cells. The transcription factor, T cell factor-1 (TCF1), and its homologue, Lymphoid enhancer-binding factor-1 (LEF1), have critical roles in the development, differentiation, and persistence of both CD4+ and CD8+ T cells. However, the influence of TCF1 and LEF1 on Th1 and Tfh differentiation remains to be examined. Furthermore, due to alternative promoter usage, TCF1 and LEF1 are expressed as both long and short isoforms. The distinct roles of the long and short isoforms of TCF1 in the context of CD4+ and CD8+ T cell responses have not been defined. My studies utilized multiple novel mouse strains to examine the roles of TCF1 and LEF1 in Tfh and Th1 differentiation during viral infection, and the unique requirements of TCF1 long isoforms in CD4+ and CD8+ T cell responses. Specifically, my initial studies characterized a new TCF1 reporter construct (referred to as p45GFP reporter) and used this reporter to address the specific contributions of TCF1 long isoforms to the CD8+ T cell response. Previous studies have abrogated all TCF1 isoforms and shown that in the absence of TCF1, the memory CD8+ T cell population is dramatically impaired and exhibits defective persistence over time. Here, I showed that TCF1 short isoforms are sufficient for the generation of memory CD8+ T cells, however TCF1 long isoforms are important for the maturation of memory CD8+ T cells. Another critical component of pathogen clearance and long-term protection is a productive humoral response, which is optimized by the B cell help provided by Tfh cells. Using the p45GFP reporter, I showed that TCF1 is specifically retained in Tfh cells, but downregulated in Th1 cells. I utilized a huCd2-Cre system to conditionally delete TCF1 and LEF1 in mature T cells. In response to viral infection, TCF1 and LEF1 double-deficient mice showed normal Th1 responses, but severely defective Tfh differentiation and a concomitant impaired B cell response. I further demonstrated that TCF1 promotes Tfh differentiation by directly regulating many Tfh-associated genes. Furthermore, I used the p45GFP reporter to I identified distinct, but critical, roles for both long and short isoforms of TCF1 in driving Tfh differentiation and repressing differentiation toward Th1 or germinal center Tfh cells. Finally, while TCF1 is known to be critical in the formation of memory CD8+ T cells, its impact on memory CD4+ T cell generation has not been assessed. Once again utilizing the p45GFP reporter, my studies identified an important role for TCF1 long isoforms in the survival of both Th1 and Tfh cells through contraction. In the absence of TCF1 long isoforms, the memory CD4+ T cell population is severely reduced. Taken together, my work has demonstrated critical roles for TCF1 during both effector and memory phases of the CD4+ T cell response to viral infection. In summary, TCF1 is crucial for CD4+ T cells to effectively differentiate and provide important help to B cells during viral infection. Moreover, my studies have identified critical and unique roles for long and short isoforms of TCF1. Finally, TCF1 is necessary for optimal formation of memory CD4+ and CD8+ T cells, and thus is an essential component in achieving protective immunological memory after viral infection.
20

Rétrocontrôle des réponses Th2 par l'interleukine-6 et identification d'un nouveau facteur de transcription exprimé par les lymphocytes T helper folliculaires / Restriction of Th2 responses by interleukin-6 and identification of a new transcription factor expressed in follicular helper T cells

Debuisson, Delphine 05 December 2014 (has links)
L’objectif de notre travail a été de caractériser le rôle de l’IL-6 dans la différenciation des lymphocytes Tfh et des lymphocytes Th2. Les lymphocytes Tfh ont pour fonction d’aider les lymphocytes B à produire des anticorps indispensables pour nous protéger contre divers pathogènes. Les lymphocytes Th2, quant à eux, sont spécialisés dans l’élimination de parasites extracellulaires tels que les helminthes.<p>Dans un premier temps, nous avons voulu identifier les gènes dont l’expression est induite par l’IL-6, avec comme objectif une meilleure compréhension des mécanismes permettant aux lymphocytes T de se différencier en cellules Tfh.<p>Au cours de notre travail, nous avons identifié le facteur de transcription, MyoR (Myogenic Repressor) comme étant exprimé au sein des lymphocytes T helper et dont l’expression est induite par l’IL-6. Nos observations expérimentales ont démontré que le facteur MyoR n’est pas indispensable pour la différenciation des lymphocytes Tfh, ni pour leur fonction. Cependant, l’expression de l’ARNm codant pour MyoR pourrait être utilisée comme un biomarqueur des cellules Tfh in vitro ou in vivo.<p>Nous avons ensuite caractérisé la réponse immune induite in vivo par des cellules présentatrices d’antigènes issues de souris déficientes pour l’IL-6. Cette approche nous a permis de mettre en évidence le rôle immunosuppresseur de l’IL-6 sur le développement des réponses de type Th2. En effet, nous avons montré que l’injection de BMDCs (Bone Marrow derived dendritic cells) IL-6-/- dans des souris receveuses de type sauvage induisent une réponse Th2 augmentée in vivo.<p>Nos résultats suggèrent que l’inhibition de la réponse Th2 par l’IL-6 in vivo et in vitro pourrait impliquer la présence d’un ou de plusieurs miRNAs.<p>Cette inhibition pourrait être un mécanisme de rétrocontrôle afin d’éviter une exacerbation de la réponse immune Th2. / Doctorat en Sciences / info:eu-repo/semantics/nonPublished

Page generated in 0.041 seconds