• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 71
  • 45
  • 32
  • 8
  • 6
  • 5
  • 4
  • 3
  • 2
  • 1
  • 1
  • Tagged with
  • 206
  • 206
  • 206
  • 40
  • 40
  • 39
  • 35
  • 29
  • 29
  • 28
  • 26
  • 25
  • 25
  • 21
  • 20
  • 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.
31

Regulatory T cells : molecular requirements for their selection and therapeutic use in autoimmune disease

Malpass, Katy H. January 2009 (has links)
Regulatory T cells (Tregs), expressing the transcription factor Foxp3, form a key component of peripheral immune tolerance, guarding against auto-aggressive immune responses. Multiple Sclerosis is an inflammatory and demyelinating disease of the central nervous system (CNS) which is largely believed to be mediated by immune components reacting to the self myelin antigens that insulate the nerve fibres. Recent investigations have reported that regulatory T cells are dysfunctional in MS patients; therefore enhancing the regulatory T cell responses in MS is an attractive therapeutic target. Using the mouse model of MS, experimental autoimmune encephalomyelitis (EAE) we have attempted to develop disease-relevant Treg-based therapies to prevent disease induction. This required an understanding of the antigenic-reactivity of Tregs during disease. Results described in this thesis show that a proportion of Tregs in the draining lymph nodes and CNS were reactive to the disease initiating antigen(s) and could suppress in vitro responses of naïve T cells bearing transgenic T cell receptors, recognising the same antigen. Adoptive transfer of antigen-reactive Tregs suppressed disease induced with the same antigen, but also reduced disease induced with a distinct myelin antigen. Peptide-based tolerance using a high affinity MHC binding peptide analogue expanded and maintained antigen-reactive T cells which were tolerant to antigenic restimulation, although these cells did not express Foxp3. Peptide-treated mice showed reduced incidence of disease relapses during EAE induced against a distinct myelin antigen. Thus, while EAE and MS will involve a polyclonal effector T cell response to many antigens, therapeutic targeting of Tregs reactive against one CNS component may be sufficient to reduce disease. Endogenous expression of myelin autoantigen did not grossly alter the response of antigenreactive Tregs in the periphery. However, expression of endogenously derived viral superantigen enhanced the proportion of superantigen-reactive Foxp3+ Tregs in the periphery. This observation was extended using exogenous superantigen, suggesting that prolonged exposure to low dose (super)antigen tips the balance of the immune system in favour of regulation. This has implications for the ability to successfully fight infection, as well as for the limitation of autoaggressive responses and may contribute to the understanding of the hygiene hypothesis.
32

Variations du développement thymique des lymphocytes T régulateurs : influence de l’âge et de la génétique / Age and genetic-dependent variations of thymic development of regulatory T lymphocytes

Darrigues, Julie 24 May 2018 (has links)
Les lymphocytes T régulateurs CD4+ Foxp3+ (Treg) sont des acteurs clés de l’homéostasie du système immunitaire. Grâce à des fonctions immunosuppressives, les Treg contrôlent la survenue et l’amplitude des réponses immunitaires. Ils sont ainsi essentiels à la prévention de pathologies auto-immunes et inflammatoires chroniques. Comme les autres lymphocytes Tαβ (Tconv), les Treg se différencient dans le thymus à partir d’un même précurseur hématopoïétique. Bien qu’encore méconnu, le développement des Treg diffère de celui des Tconv par la nature des interactions ou des signaux échangés entre les précurseurs et les cellules stromales thymiques. Dans notre équipe nous nous intéressons à la génération intra-thymique des Treg, et particulièrement aux facteurs pouvant influencer ce processus. Ainsi, au fait de l’involution du thymus avec l’âge et de la réduction de la production de Tconv qui en découle, nous avons monitoré spécifiquement le développement des Treg au cours de la vie. De façon surprenante, nous avons observé une diminution plus importante de la production de Treg, que de Tconv, avec l’âge. Cependant, nous avons constaté que la population de Treg thymique n’était pas seulement constituée de cellules en développement, mais également de cellules plus matures, présentant un phénotype activé et différencié, dont la proportion s’accroit avec l’âge. Nous avons alors démontré qu’il s’agissait de Treg ayant recirculé depuis la périphérie (tissus, organes lymphoïdes) vers leur organe d’origine ; et que ces derniers étaient capables de supprimer spécifiquement le développement de leurs précurseurs en rentrant en compétition pour un facteur de survie essentiel : l’interleukine 2 (IL-2). Ainsi, nos travaux ont révélé l’existence d’une boucle de rétrocontrôle négatif du développement thymique des Treg. Cette découverte nous a alors amené à reconsidérer les données de la littérature qui décrivaient un nombre plus élevé de Treg thymiques chez des souris NOD, enclines aux auto-immunités, par rapport aux souris B6 résistantes. Nous voulions déterminer si la variation du nombre de Treg thymiques d’une souche à l’autre résultait d’une réelle influence génétique sur le développement des Treg, ou bien sur la recirculation de ces derniers. L’analyse comparative de la proportion de Treg recirculants a révélé que celle-ci était toujours plus élevée chez les souris NOD par rapport aux souris B6 dès les premières semaines de vie. Par ailleurs, nous avons démontré que cette forte densité de Treg recirculants chez les souris NOD résultait d’une hyper-activation des Treg dans la périphérie, connue pour favoriser la migration de ces derniers vers le thymus. Enfin, en accord avec le mécanisme de rétrocontrôle précédemment décrit, la plus forte présence de Treg recirculants chez la souris NOD entraine une inhibition plus précoce du développement thymique des Treg. En conclusion, l’ensemble de nos données rapportent l’existence de variations inter-individuelles du développement des lymphocytes T régulateurs dans le thymus. Ces variations conditionnées par l’âge et/ou par la génétique de l’individu sont exercées en partie sur le processus de recirculation des Treg périphériques vers le thymus, ce qui entraine l’inhibition du développement de nouveaux Treg. Les conséquences sur l’organisme de ces variations quantitatives du développement des Treg devront être déterminées, et s’inscriront dans une démarche globale visant à une meilleure prédiction de la susceptibilité des individus vis-à-vis des immuno-pathologies. / CD4+ Foxp3+ regulatory T lymphocytes (Treg) are key players in the immune homeostasis. Thanks to their suppressive functions, Treg control the occurrence and the intensity of immune responses. They are thus essential in preventing the onset of auto-immune and chronic inflammatory diseases. Like other αβ T lymphocytes (Tconv), Treg differentiate in the thymus from a common hematopoietic precursor. Although still not completely unveiled, Treg development seems to differ from that of Tconv due to the interactions and signals exchanged between Treg precursors and thymic stromal cells. The research of our team focuses on the intra-thymic generation of Treg, and on factors that could influence this process. With age, the size of the thymus decreases and as a consequence the thymic production of new Tconv cells is reduced. Then while studying the differential requirements for Tconv and Treg cell development, we monitored their thymic production throughout life. Surprisingly, we observed a stronger decrease in Treg than Tconv production with age. However, we also found that the thymic Treg population was not only constituted by newly developing cells, but also by mature cells whose proportion increased with age, and exhibited an activated and differentiated phenotype. We then demonstrated that this population comprised Treg that have recirculated from the periphery (tissues, lymphoid organs) to the thymus. Mature thymic Treg were able to specifically suppress the development of their precursors by limiting the availability of an essential survival factor: the interleukin-2 (IL-2). Our work therefore demonstrated the existence of a negative feedback loop exerted by peripheral Treg controlling thymic Treg development. This finding led us to reconsider the data from the literature which described a higher number of thymic Treg in NOD mice, prone to autoimmunity, compared to resistant B6 mice. We determined whether the variation in the number of thymic Treg from one strain to another resulted from a genetic control on thymic Treg development, or on Treg recirculation. Strikingly, a greater proportion of recirculating thymic Treg was observed in NOD than in B6 mice at all the ages analyzed. We have subsequently demonstrated that this high amount of recirculating Treg in NOD mice was due to hyper-activation of peripheral Treg, favoring their migration to the thymus. Finally, in agreement with the negative feedback loop we previously described, the higher density of recirculating Treg in the thymus of NOD mice induced an earlier inhibition of the intra-thymic development of new Treg. To conclude, we report here the existence of inter-individual variations of regulatory T lymphocytes development in the thymus. These age-dependent and/or genetic-dependent factors seem to shape Treg recirculation from the periphery to the thymus, and as a consequence the de novo development of Treg. It will be important, in the future, to determine the impact on the organism of these quantitative variations in Treg development and on the susceptibility towards immuno-pathologies
33

Regulatory T Cells Promote Breast Cancer Progression Through Inhibiting Classical Activation of Macrophages

Clark, Nicholas M 01 January 2019 (has links)
Transient ablation of regulatory T cells has been shown to be effective at hindering tumor growth and metastasis in murine breast cancer model. Based on our lab’s previous work, we have demonstrated that NK cells and CD8+ cytotoxic T cells were not required for the protective effect of Treg cell ablation. However, we also reported that CD4+ helper T cells and IFN-γ were required for the protective effect of Treg cell ablation. Furthermore, we observed that CD11B+ cells responded to Treg ablation therapy by up-regulating target genes of IFN-γ. Therefore, this study aimed to investigate the connection between the myeloid cell compartment and IFN-γ signaling after regulatory T cell ablation therapy. Through a combination of conditional knockout mouse models, cellular fate mapping experiments, adoptive transfers, and co-injection experiments, we demonstrated that tumor-associated macrophages (TAMs), derived from the bone marrow via CCR2/CCL2 axis, were responsible for the therapeutic effect of regulatory T cell ablation. In addition, we determined that IFN-γ signaling was required for the TAMs to mediate the protective phenotype seen after regulatory T cell ablation. Furthermore, based on our findings, we developed a genetic signature based on TAMs from treated or untreated tumors that had a predictive value for patient survival. Thus, our findings indicated a strong connection between IFN-γ release, classical activation of TAMs, and depletion of regulatory T cells, and taken together, our data could offer potential clinical strategies to mimic regulatory T cell ablation.
34

Achievement of Transplantation Tolerance: Novel Approaches and Mechanistic Insights

Pidala, Joseph 17 March 2014 (has links)
Current immune suppressive strategies fail to induce donor-recipient immune tolerance after allogeneic hematopoietic cell transplantation. Accordingly, patients suffer morbidity and mortality from graft vs. host disease (GVHD) and prolonged immune suppressive therapy. Biologic insight into transplantation tolerance is needed, and translation of such insight to novel clinical strategies may improve clinical outcomes. We report original investigation at seminal phases of this process including initial prophylactic immune suppression, onset of acute graft vs. host disease, and ultimate immune suppression discontinuation: In a controlled randomized clinical trial, we demonstrate that sirolimus-based immune suppression reduces risk for acute GVHD, ameliorates the severity of subsequent chronic GVHD, and supports reconstitution of functional regulatory T cells. Study of tissue-infiltrating CD4+ T cell subsets in acute GVHD target organs supports a pathogenic role for Th17 cells. Finally, we demonstrate that peripheral blood transcriptional biomarkers provide mechanistic insight into human transplantation tolerance. These data signal progress, and suggest rational translational efforts to achieve transplantation tolerance.
35

Contribution à l'étude de la reconstitution immunitaire après miniallogreffe de cellules souches hématopoïétiques.

Castermans, Emilie 23 June 2008 (has links)
Les principaux échecs des greffes de cellules souches allogéniques (HSCT) sont la rechute et les infections, accompagnées ou non de GvHD (Graft versus Host Disease, maladie du greffon contre lhôte), manifestations qui pourraient être partiellement attribuées à un déficit immunitaire (rechute, infections) ou à une réaction immunitaire exacerbée envers le receveur (GvHD) (Baron, Storer et al. 2006). Létude de la reconstitution du système immunitaire, et particulièrement, lymphocytaire, savère dès lors capitale dans le développement des HSCT. Nous avons investigué la reconstitution immune de 50 patients traités par HSCT nonmyéloablative classique vs HSCT nonmyéloablative déplétée en lymphocytes T CD8+. 50 patients ont été randomisés : greffon déplété en CD8 (n=22) vs non manipulé (n=28). Lâge médian était de 57 ans au moment de la greffe (range 36-69). Le régime de conditionnement consistait en une irradiation corporelle totale de 2 Gy avec ou sans ajout de Fludarabine. 20 patients ont reçu une greffe de donneur familial, 14 de donneurs non familiaux HLA identiques, et 16 de donneurs non familiaux présentant une disparité HLA. La reconstitution immunitaire la première année après HCT a été monitorée par cytométrie en flux, analyse de la diversité du répertoire du TCR (spectratyping), quantification de sjTREC (signal joint T cell receptor excision circle, marqueur de la thymopoïèse). La déplétion des CD8 a réduit la reconstitution des taux de CD8 durant les 6 premiers mois postgreffe (P<0.0001) mais na pas présenté dimpact significatif sur la récupération des autres populations cellulaires. Les concentrations de sjTREC et des taux de CD3 ont augmenté parallèlement entre le jour 100 et le jour 365 après greffe (P=0.006 et P=0.022, respectivement), suggérant ainsi la néoproduction de lymphocytes T par le thymus, même chez ces patients âgés. Les facteurs associés à une concentration conséquente de TREC un an après greffe incluent 1° le choix dun donneur non familial HLA-matched (P=0.029), 2° de hautes concentrations de lymphocytes T dans le greffon (P=0.002), et 3° labsence de GVHD chronique (P<0.0001). Nos données suggèrent un modèle biphasique de reconstitution du pool lymphocytaire T: 1) une expansion des T matures du greffon en périphérie durant les 3 premiers mois ;2) une néoproduction active par voie intrathymique assurant la reconstitution du système immunitaire à plus long terme. Combien de temps cette néosynthèse intrathymique perdure-t-elle ? Quels facteurs laffectent ? Est-elle associée à une diversité accrue du répertoire lymphocytaire ? Ces questions nous ont amenés à étudier la reconstitution immunitaire à long terme (entre 1 et 6.5 ans) de 73 patients après minigreffe (211 points au total). Nous avons observé un maintien de la thymopoïèse réenclenchée au cours de la première année postgreffe chez les patients âgés de moins de 50 ans et de 50-60 ans. Cette reprise de la thymopoïèse na pas été mise en évidence au sein du groupe des plus de 60 ans. Ainsi, une application clinique concrète à cette observation pourrait être ladministration de greffons particulièrement riches en lymphocytes T à ce type de patients, puisquils seront plus susceptibles de développer une lymphopénie persistante postgreffe par absence de réenclenchement de la voie thymodépendante. Les facteurs associés à une reconstitution thymodépendante à long terme après minigreffe étaient : 1°labsence de cGvHD (P<0.0001); 2°lâge du receveur (P<0.0001); 3° la concentration en lymphocytes T dans le greffon (P=0. 0.0038) ; 4°laugmentation de la diversité HLA (P=0.0001). Enfin, une tendance non significative à une augmentation parallèle de la diversité du répertoire TCR et des concentrations en sjTREC a été mise en évidence. Cette analyse doit être confirmée par létude dun plus grand nombre de sujets. Afin déliminer au maximum les influences extrathymiques sur les taux de TREC périphériques, nous comptons mesurer pour chaque patient des TREC précoces (BTREC) et un TREC tardif (sjTREC), et calculer un ratio reflétant exactement le nombre de divisions intrathymiques. Cette méthode a été validée dans notre centre au cours dune expansion de lymphocytes T in vitro au moyen de billes anti-CD3 anti-CD28, expansion durant laquelle nous avons pu observer une diminution des sjTREC et des BTREC, mais pas du ratio sjTREC/BTREC. Lanalyse des ratios de TREC des patients greffés est actuellement à létude. Une troisième étude a été également menée afin déclaircir le lien entre GvHD, thymopoïèse et présence de lymphocytes T régulateurs (TRegs) chez 64 patients après HCT. Lémergence dun nouveau marqueur spécifique des TRegs, le CD127, a permis pour la première fois une isolation sans équivoque des TRegs (Liu, Putnam et al. 2006). Nous navons pas pu mettre en évidence de différence significative entre lapparition dune cGvHD chez les patients présentant après greffe des taux de TRegs supérieurs ou inférieurs à la médiane (P=0.13). Inversement, loccurrence de cGvHD na pas paru significativement affecter les concentrations en TRegs après greffe (P=0.1). Nous avons également mis en évidence une corrélation positive significative entre le taux de sjTREC/ml et le taux de TReg/ul au J100 (R=0.46, P=0.007) et à 1 an R=0.47, P=0.001). Afin de déterminer lorigine précise de ces TReg après greffe (thymus du donneur?), nous réalisons actuellement des mesures de sjTREC et de chimérisme sur les populations cellulaires triées au moyen du triple marquage CD4+CD25+CD127- (cellules T classiques vs régulatrices).
36

Therapeutic immunomodulation of allergic lung disease using regulatory dendritic cells in a mouse model of asthma

Nayyar, Aarti 24 February 2009
We report herein that IL-10-treated dendritic cells (DC) can be used effectively to reverse established severe asthma-like disease in a mouse model. Our lab had shown previously that allergen-presenting splenic CD8¦Á+ DCs could ¡Ö50% reduce airway hyper responsiveness (AHR), eosinophilia, and Th2 responses in asthma-phenotype mice, but only marginally reduce IgE/IgG1 levels. We now show that bone marrow-derived DCs that have been differentiated in the presence of IL-10 (DCIL-10) are effective in reversing the asthma phenotype. Co-culture of DCIL-10 with T memory (TM) cells from asthma-phenotype mice was associated with lack of Th2 responses, and this was partially reversed by IL-2. Immunostimulatory DC activated these Th2 cells. <i>In vivo</i>, delivery of allergen-pulsed DCIL-10, either into the airway or intraperitoneally abrogated AHR from weeks 3-10 post-treatment, and ameliorated lung eosinophilia and Th2 (IL-4, -5, -9, & -13, IgE) responses, as well as circulating allergen-specific IgE responses for at least 32 weeks following treatment. Repeated OVADCIL-10 treatments kept AHR normalized for 8 weeks as well as Th2 responses significantly low. In vivo, delivery of anti-IL-10R, but not anti-TGF-¦Â from day 12-21 after treatment had moderate effects on DCIL-10-driven tolerance, but 1-methyl tryptophan (inhibitor of indoleamine-2,3-dioxygenase) treatment had significant effects on Th2 responses. The mechanisms mediating tolerance in vivo are likely complex, but we speculate that infectious tolerance sustains this regulatory activity during the 32-week period in which we have observed tolerance to be in place.
37

Tolerogenic CD4-8- Dendritic Cells and their Conversion into Immunogenic Ones via TLR9 Signaling

Zhang, Xueshu 07 November 2008
It is clear that dendritic cells (DCs) are essential for priming of T cell responses against tumors. However, the distinct roles DC subsets play in regulation of T cell responses in vivo are largely undefined. In this study, we investigated the capacity of ovalbumin (OVA)-presenting CD48, CD4+8, or CD48+ DCs (OVA-pulsed DC (DCOVA)) from mouse spleen in stimulation of OVA-specific T cell responses. Our data show that each DC subset stimulated proliferation of allogeneic and autologous OVA-specific CD4+ and CD8+ T cells in vitro, but that the CD48 DCs did so only weakly. Both CD4+8 and CD48+ DCOVA induced strong tumor-specific CD4+ Th1 responses and fully protective CD8+ cytotoxic T lymphocyte (CTL)-mediated antitumor immunity, whereas CD48 DCOVA, which were less mature and secreted substantial transforming growth factor (TGF- ) upon coculture with T cell receptor (TCR)-transgenic OT II CD4+ T cells, induced the development of interleukin-10 (IL-10)-secreting CD4+ T regulatory 1 (Tr1) cells. Transfer of these Tr1 cells, but not T cells from cocultures of CD48 DCOVA and IL-10/ OT II CD4+ T cells, into CD48+ DCOVA-immunized animals abrogated otherwise inevitable development of antitumor immunity. Taken together, CD48 DCs stimulate development of IL-10-secreting CD4+ Tr1 cells that mediated immune suppression, whereas both CD4+8 and CD48+ DCs effectively primed animals for protective CD8+ CTL-mediated antitumor immunity. <p> Different DC subsets play distinct roles in immune responses. CD4-8- DCs secreting TGF-â stimulate CD4+ regulatory T type 1 (Trl) cell responses leading to inhibition of CD8 CTL responses and antitumor immunity. In this study, we explored the potential effect of three stimuli CpG, lipopolysaccharide (LPS) and anti-CD40 antibody in conversion of CD4-8- DC-induced tolerance. We demonstrated that when CD4-8- DCs were isolated from overnight culture and cultured for another 8 hrs in AIM-V plus recombinant mouse granulocyte-macrophage colony-stimulating factor (rmGM-CSF) (15-20 ng/ml) and OVA (0.1 mg/ml) with CpG (5 ug/ml), LPS (2 ug/ml) and anti-CD40 antibody (10 ug/ml), their phenotype became more mature compared with the freshly isolated ones. CpG is the only agent that stimulates the DCs to secrete significant level of interleukin-6 (IL-6) and interleukin-15 (IL-15); DNA array analyses also indicate that CpG stimulates higher expression of IL-6 and IL-15 mRNA. CpG treatment most efficiently converts the tolerogenic DCs into immunogenic ones which stimulated the OTII CD4+ T cell to become T helper type 1 (Th1) and T helper type 17 (Th17) rather Tr1, while the other two stimulator-treated DCs could not induce Th17 response. Their vaccination also induced the strongest antitumor CTL responses and protective immunity against tumor cell challenge. When CD4-8- DCs were isolated from IL-6 knock out (IL-6-/-) mice, CpG-treated DCOVA vaccination almost completely lost their animal protection capacity. Wild type B6 DCOVA-vaccinated IL-15 receptor knock out (IL-15R-/-) mice can only provide up to 30% protection against tumor challenge. Those results indicate that IL-6/ IL-l5-induced Th17 plays a critical role in their conversion. Taken together, our findings indicate that CpG treatment is the most efficient agent that can convert tolerogenic DCs into immunogenic ones and induce long-lasting antitumor immunity. We previously demonstrated that the nonspecific CD4+ T cells can acquire antigen-specific DC-released exosomes (EXO) and these CD4+ T cells with acquired exosomal MHC I peptide complex (pMHC I) can stimulate antigen-specific CD8+ CTL responses. In my project we have found that CD4-8-DCs could induce regulatory T cell type 1(Tr1) response, thus it would be very necessary to know whether regulatory T cells would change their antigen specificity if they got the membrane complex from DC through coculture or DC-derived exosome pulsing. During the beginning of my regulatory T cell project, we found that CD8+CD25+ Tr were much more easily expanded, while CD4+CD25+ Tr usually began to die just after 3 days in vitro culture and its very hard to get enough cells for further research. Therefore, CD8+CD25+ were used as a model Tr cells in the following project. To assess whether the nonspecific CD8+CD25+ Tr cells can acquire antigen-specificity via acquired exosomal pMHC I, we purified CD8+CD25+ Tr cells from wild-type C57BL/6 mice and OVA-pulsed DCOVA-released EXOOVA expressing pMHC I complexes. We demonstrated that the nonspecific CD8+CD25+ Tr cells expressing forkhead box P3 (Foxp3), cytotoxic T-Lymphocyte Antigen 4 (CTLA-4), glucocorticoid-induced tumor necrosis factor receptor (GITR), perforin and granzyme B inhibited in vitro T cell proliferation and in vivo OVA-specific CD4+ T cell-dependent and independent CD8+ CTL responses and antitumor immunity. CD8+CD25+ Tr cells suppressive effect is possibly mediated through its inhibition of DC maturation, down-regulation of secretion of Th1 polarization cytokines by DCs and its induction of T cell anergy via cell-to-cell contact. The nonspecific CD8+CD25+ Tr cells acquired antigen specificity by uptake of DCOVA-released EXOOVA expressing pMHC I and enhanced its effect on inhibition of OVA-specific CD8+ T cell responses and antitumor immunity by 10-folds. The principles elucidated in this study may have significant implications not only in antitumor immunity, but also in other sectors of immunology (e.g, autoimmunity and transplantation).
38

Therapeutic immunomodulation of allergic lung disease using regulatory dendritic cells in a mouse model of asthma

Nayyar, Aarti 24 February 2009 (has links)
We report herein that IL-10-treated dendritic cells (DC) can be used effectively to reverse established severe asthma-like disease in a mouse model. Our lab had shown previously that allergen-presenting splenic CD8¦Á+ DCs could ¡Ö50% reduce airway hyper responsiveness (AHR), eosinophilia, and Th2 responses in asthma-phenotype mice, but only marginally reduce IgE/IgG1 levels. We now show that bone marrow-derived DCs that have been differentiated in the presence of IL-10 (DCIL-10) are effective in reversing the asthma phenotype. Co-culture of DCIL-10 with T memory (TM) cells from asthma-phenotype mice was associated with lack of Th2 responses, and this was partially reversed by IL-2. Immunostimulatory DC activated these Th2 cells. <i>In vivo</i>, delivery of allergen-pulsed DCIL-10, either into the airway or intraperitoneally abrogated AHR from weeks 3-10 post-treatment, and ameliorated lung eosinophilia and Th2 (IL-4, -5, -9, & -13, IgE) responses, as well as circulating allergen-specific IgE responses for at least 32 weeks following treatment. Repeated OVADCIL-10 treatments kept AHR normalized for 8 weeks as well as Th2 responses significantly low. In vivo, delivery of anti-IL-10R, but not anti-TGF-¦Â from day 12-21 after treatment had moderate effects on DCIL-10-driven tolerance, but 1-methyl tryptophan (inhibitor of indoleamine-2,3-dioxygenase) treatment had significant effects on Th2 responses. The mechanisms mediating tolerance in vivo are likely complex, but we speculate that infectious tolerance sustains this regulatory activity during the 32-week period in which we have observed tolerance to be in place.
39

Tolerogenic CD4-8- Dendritic Cells and their Conversion into Immunogenic Ones via TLR9 Signaling

Zhang, Xueshu 07 November 2008 (has links)
It is clear that dendritic cells (DCs) are essential for priming of T cell responses against tumors. However, the distinct roles DC subsets play in regulation of T cell responses in vivo are largely undefined. In this study, we investigated the capacity of ovalbumin (OVA)-presenting CD48, CD4+8, or CD48+ DCs (OVA-pulsed DC (DCOVA)) from mouse spleen in stimulation of OVA-specific T cell responses. Our data show that each DC subset stimulated proliferation of allogeneic and autologous OVA-specific CD4+ and CD8+ T cells in vitro, but that the CD48 DCs did so only weakly. Both CD4+8 and CD48+ DCOVA induced strong tumor-specific CD4+ Th1 responses and fully protective CD8+ cytotoxic T lymphocyte (CTL)-mediated antitumor immunity, whereas CD48 DCOVA, which were less mature and secreted substantial transforming growth factor (TGF- ) upon coculture with T cell receptor (TCR)-transgenic OT II CD4+ T cells, induced the development of interleukin-10 (IL-10)-secreting CD4+ T regulatory 1 (Tr1) cells. Transfer of these Tr1 cells, but not T cells from cocultures of CD48 DCOVA and IL-10/ OT II CD4+ T cells, into CD48+ DCOVA-immunized animals abrogated otherwise inevitable development of antitumor immunity. Taken together, CD48 DCs stimulate development of IL-10-secreting CD4+ Tr1 cells that mediated immune suppression, whereas both CD4+8 and CD48+ DCs effectively primed animals for protective CD8+ CTL-mediated antitumor immunity. <p> Different DC subsets play distinct roles in immune responses. CD4-8- DCs secreting TGF-â stimulate CD4+ regulatory T type 1 (Trl) cell responses leading to inhibition of CD8 CTL responses and antitumor immunity. In this study, we explored the potential effect of three stimuli CpG, lipopolysaccharide (LPS) and anti-CD40 antibody in conversion of CD4-8- DC-induced tolerance. We demonstrated that when CD4-8- DCs were isolated from overnight culture and cultured for another 8 hrs in AIM-V plus recombinant mouse granulocyte-macrophage colony-stimulating factor (rmGM-CSF) (15-20 ng/ml) and OVA (0.1 mg/ml) with CpG (5 ug/ml), LPS (2 ug/ml) and anti-CD40 antibody (10 ug/ml), their phenotype became more mature compared with the freshly isolated ones. CpG is the only agent that stimulates the DCs to secrete significant level of interleukin-6 (IL-6) and interleukin-15 (IL-15); DNA array analyses also indicate that CpG stimulates higher expression of IL-6 and IL-15 mRNA. CpG treatment most efficiently converts the tolerogenic DCs into immunogenic ones which stimulated the OTII CD4+ T cell to become T helper type 1 (Th1) and T helper type 17 (Th17) rather Tr1, while the other two stimulator-treated DCs could not induce Th17 response. Their vaccination also induced the strongest antitumor CTL responses and protective immunity against tumor cell challenge. When CD4-8- DCs were isolated from IL-6 knock out (IL-6-/-) mice, CpG-treated DCOVA vaccination almost completely lost their animal protection capacity. Wild type B6 DCOVA-vaccinated IL-15 receptor knock out (IL-15R-/-) mice can only provide up to 30% protection against tumor challenge. Those results indicate that IL-6/ IL-l5-induced Th17 plays a critical role in their conversion. Taken together, our findings indicate that CpG treatment is the most efficient agent that can convert tolerogenic DCs into immunogenic ones and induce long-lasting antitumor immunity. We previously demonstrated that the nonspecific CD4+ T cells can acquire antigen-specific DC-released exosomes (EXO) and these CD4+ T cells with acquired exosomal MHC I peptide complex (pMHC I) can stimulate antigen-specific CD8+ CTL responses. In my project we have found that CD4-8-DCs could induce regulatory T cell type 1(Tr1) response, thus it would be very necessary to know whether regulatory T cells would change their antigen specificity if they got the membrane complex from DC through coculture or DC-derived exosome pulsing. During the beginning of my regulatory T cell project, we found that CD8+CD25+ Tr were much more easily expanded, while CD4+CD25+ Tr usually began to die just after 3 days in vitro culture and its very hard to get enough cells for further research. Therefore, CD8+CD25+ were used as a model Tr cells in the following project. To assess whether the nonspecific CD8+CD25+ Tr cells can acquire antigen-specificity via acquired exosomal pMHC I, we purified CD8+CD25+ Tr cells from wild-type C57BL/6 mice and OVA-pulsed DCOVA-released EXOOVA expressing pMHC I complexes. We demonstrated that the nonspecific CD8+CD25+ Tr cells expressing forkhead box P3 (Foxp3), cytotoxic T-Lymphocyte Antigen 4 (CTLA-4), glucocorticoid-induced tumor necrosis factor receptor (GITR), perforin and granzyme B inhibited in vitro T cell proliferation and in vivo OVA-specific CD4+ T cell-dependent and independent CD8+ CTL responses and antitumor immunity. CD8+CD25+ Tr cells suppressive effect is possibly mediated through its inhibition of DC maturation, down-regulation of secretion of Th1 polarization cytokines by DCs and its induction of T cell anergy via cell-to-cell contact. The nonspecific CD8+CD25+ Tr cells acquired antigen specificity by uptake of DCOVA-released EXOOVA expressing pMHC I and enhanced its effect on inhibition of OVA-specific CD8+ T cell responses and antitumor immunity by 10-folds. The principles elucidated in this study may have significant implications not only in antitumor immunity, but also in other sectors of immunology (e.g, autoimmunity and transplantation).
40

The interactions of tolerogenic dendritic cells, induced regulatory T cells and antigen-specific IgG1-secreting plasma cells in asthma

2015 June 1900 (has links)
Allergic asthma is a chronic inflammatory airway disease that is dominated by Th2 immune responses, with accumulation of eosinophils, IgE and IgG1 production, and airway hyperresponsiveness. We reported previously that treatment of OVA-asthmatic mice with allergen-presenting IL-10-differentiated dendritic cells (DC) (DC10) leads to progressive and long-lasting full-spectrum asthma tolerance. However, little has been done in investigating a role for antigen-specific B cells in DC10-induced tolerance. In this study, we characterized the surface markers of DC10 and found that these cells expressed lower levels of CD40, CD80, MHC II, PD-L1 and PD-L2 relative to immunostimulatory LPS-differentiated DCs (DCLPS). Co-culturing DC10 or DC10-induced regulatory T cells (iTreg) with CD4+ Th2 effector T cells from asthmatic mice led to a marked suppression of DCLPS-induced T effector cell proliferation. Moreover, DC10 treatment of asthma phenotype mice down-regulated airway eosinophilic inflammation as determined 48 h after a recall allergen challenge, and reduced pulmonary parenchymal tissue OVA-specific IgG1-secreting (OVA-IgG1) plasma cell numbers. The number of lung OVA-specific IgG1 plasma cells decreased by 46.7% over a 2 week period in the absence of repeated allergen challenge, while the numbers of bone marrow OVA-specific IgG1 plasma cells stayed relatively stable over a 6 week period, as determined 48 h after a single allergen challenge of asthmatic mice. DC10 treatment had a significant impact on the serum of IgG1/IgE response. To address the question of how DC10 influence OVA-IgG1 plasma cells responses, we co-cultured enzymatically-dispersed lung total cells from asthmatic mice with or without DC10, and found that the DC10 significantly suppressed OVA-IgG1 plasma cell antibody production. To determine whether DC10 required input from T cells to accomplish this, we co-cultured CD4 T cell-depleted, B cell-enriched populations from the lungs of asthmatic mice with or without DC10, and found that DC10 strongly (65.4+/-3.5%) suppressed OVA-IgG1 plasma cells in CD4 T cell-depleted lung cell cultures. To assess whether DC10-induced Treg also suppress IgG1-secretion, we co-cultured lung CD4+ T cells from untreated or DC10-tolerized asthmatic mice with total lung cells from asthmatic donors, and found that the DC10-induced Tregs effectively (52.2+/-8.7%) suppressed OVA-IgG1 plasma cell responses. In summary, DC10 treatment strongly down-regulate OVA-specific IgG1 plasma cell responses of asthmatic mice, both in vivo and in vitro by at least two mechanisms: directly via DC10 as well as indirectly through DC10-induced Tregs.

Page generated in 0.0836 seconds