975 resultados para Experimental cell


Relevância:

30.00% 30.00%

Publicador:

Resumo:

The circadian timing system controls cell cycle, apoptosis, drug bioactivation, and transport and detoxification mechanisms in healthy tissues. As a consequence, the tolerability of cancer chemotherapy varies up to several folds as a function of circadian timing of drug administration in experimental models. Best antitumor efficacy of single-agent or combination chemotherapy usually corresponds to the delivery of anticancer drugs near their respective times of best tolerability. Mathematical models reveal that such coincidence between chronotolerance and chronoefficacy is best explained by differences in the circadian and cell cycle dynamics of host and cancer cells, especially with regard circadian entrainment and cell cycle variability. In the clinic, a large improvement in tolerability was shown in international randomized trials where cancer patients received the same sinusoidal chronotherapy schedule over 24h as compared to constant-rate infusion or wrongly timed chronotherapy. However, sex, genetic background, and lifestyle were found to influence optimal chronotherapy scheduling. These findings support systems biology approaches to cancer chronotherapeutics. They involve the systematic experimental mapping and modeling of chronopharmacology pathways in synchronized cell cultures and their adjustment to mouse models of both sexes and distinct genetic background, as recently shown for irinotecan. Model-based personalized circadian drug delivery aims at jointly improving tolerability and efficacy of anticancer drugs based on the circadian timing system of individual patients, using dedicated circadian biomarker and drug delivery technologies.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

BACKGROUND: Regulatory T cells (Tregs) are key players in controlling the development of airway inflammation. However, their role in the mechanisms leading to tolerance in established allergic asthma is unclear. OBJECTIVE: To examine the role of Tregs in tolerance induction in a murine model of asthma. METHODS: Ovalbumin (OVA) sensitized asthmatic mice were depleted or not of CD25(+) T cells by anti-CD25 PC61 monoclonal antibody (mAb) before intranasal treatment (INT) with OVA, then challenged with OVA aerosol. To further evaluate the respective regulatory activity of CD4(+)CD25(+) and CD4(+)CD25(-) T cells, both T cell subsets were transferred from tolerized or non-tolerized animals to asthmatic recipients. Bronchoalveolar lavage fluid (BALF), T cell proliferation and cytokine secretion were examined. RESULTS: Intranasal treatment with OVA led to increased levels of IL-10, TGF-beta and IL-17 in lung homogenates, inhibition of eosinophil recruitment into the BALF and antigen specific T cell hyporesponsiveness. CD4(+)CD25(+)Foxp3(+) T cells were markedly upregulated in lungs and suppressed in vitro and in vivo OVA-specific T cell responses. Depletion of CD25(+) cells before OVA INT severely hampered tolerance induction as indicated by a strong recruitment of eosinophils into BALF and a vigorous T cell response to OVA upon challenge. However, the transfer of CD4(+)CD25(-) T cells not only suppressed antigen specific T cell responsiveness but also significantly reduced eosinophil recruitment as opposed to CD4(+)CD25(+) T cells. As compared with control mice, a significantly higher proportion of CD4(+)CD25(-) T cells from OVA treated mice expressed mTGF-beta. CONCLUSION: Both CD4(+)CD25(+) and CD4(+)CD25(-) T cells appear to be essential to tolerance induction. The relationship between both subsets and the mechanisms of their regulatory activity will have to be further analyzed.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

T helper type 17 (Th17) cells play an important pathogenic function in autoimmune diseases; their regulation, however, is not well understood. We show that the expression of a tumor necrosis factor receptor family member, death receptor 3 (DR3; also known as TNFRSF25), is selectively elevated in Th17 cells, and that TL1A, its cognate ligand, can promote the proliferation of effector Th17 cells. To further investigate the role of the TL1A-DR3 pathway in Th17 regulation, we generated a TL1A-deficient mouse and found that TL1A(-/-) dendritic cells exhibited a reduced capacity in supporting Th17 differentiation and proliferation. Consistent with these data, TL1A(-/-) animals displayed decreased clinical severity in experimental autoimmune encephalomyelitis (EAE). Finally, we demonstrated that during EAE disease progression, TL1A was required for the optimal differentiation as well as effector function of Th17 cells. These observations thus establish an important role of the TL1A-DR3 pathway in promoting Th17 cell function and Th17-mediated autoimmune disease.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

Chronic viral infections and malignant tumours induce T cells that have a reduced ability to secrete effector cytokines and have upregulated expression of the inhibitory receptor PD1 (programmed cell death protein 1). These features have so far been considered to mark terminally differentiated 'exhausted' T cells. However, several recent clinical and experimental observations indicate that phenotypically exhausted T cells can still mediate a crucial level of pathogen or tumour control. In this Opinion article, we propose that the exhausted phenotype results from a differentiation process in which T cells stably adjust their effector capacity to the needs of chronic infection. We argue that this phenotype is optimized to cause minimal tissue damage while still mediating a critical level of pathogen control. In contrast to the presently held view of functional exhaustion, this new concept better reflects the pathophysiology and clinical manifestations of persisting infections, and it provides a rationale for emerging therapies that enhance T cell activity in chronic infection and cancer by blocking inhibitory receptors.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

Mice with homologous disruption of the gene coding for the ligand-binding chain of the interferon (IFN) gamma receptor and derived from a strain genetically resistant to infection with Leishmania major have been used to study further the role of this cytokine in the differentiation of functional CD4+ T cell subsets in vivo and resistance to infection. Wild-type 129/Sv/Ev mice are resistant to infection with this parasite, developing only small lesions, which resolve spontaneously within 6 wk. In contrast, mice lacking the IFN-gamma receptor develop large, progressing lesions. After infection, lymph nodes (LN) and spleens from both wild-type and knockout mice showed an expansion of CD4+ cells producing IFN-gamma as revealed by measuring IFN-gamma in supernatants of specifically stimulated CD4+ T cells, by enumerating IFN-gamma-producing T cells, and by Northern blot analysis of IFN-gamma transcripts. No biologically active interleukin (IL) 4 was detected in supernatants of in vitro-stimulated LN or spleen cells from infected wild-type or deficient mice. Reverse transcription polymerase chain reaction analysis with primers specific for IL-4 showed similar IL-4 message levels in LN from both types of mice. The IL-4 message levels observed were comparable to those found in similarly infected C57BL/6 mice and significantly lower than the levels found in BALB/c mice. Anti-IFN-gamma treatment of both types of mice failed to alter the pattern of cytokines produced after infection. These data show that even in the absence of IFN-gamma receptors, T helper cell (Th) 1-type responses still develop in genetically resistant mice with no evidence for the expansion of Th2 cells.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

Hearing loss can be caused by a variety of insults, including acoustic trauma and exposure to ototoxins, that principally effect the viability of sensory hair cells via the MAP kinase (MAPK) cell death signaling pathway that incorporates c-Jun N-terminal kinase (JNK). We evaluated the otoprotective efficacy of D-JNKI-1, a cell permeable peptide that blocks the MAPK-JNK signal pathway. The experimental studies included organ cultures of neonatal mouse cochlea exposed to an ototoxic drug and cochleae of adult guinea pigs that were exposed to either an ototoxic drug or acoustic trauma. Results obtained from the organ of Corti explants demonstrated that the MAPK-JNK signal pathway is associated with injury and that blocking of this signal pathway prevented apoptosis in areas of aminoglycoside damage. Treatment of the neomycin-exposed organ of Corti explants with D-JNKI-1 completely prevented hair cell death initiated by this ototoxin. Results from in vivo studies showed that direct application of D-JNKI-1 into the scala tympani of the guinea pig cochlea prevented nearly all hair cell death and permanent hearing loss induced by neomycin ototoxicity. Local delivery of D-JNKI-1 also prevented acoustic trauma-induced permanent hearing loss in a dose-dependent manner. These results indicate that the MAPK-JNK signal pathway is involved in both ototoxicity and acoustic trauma-induced hair cell loss and permanent hearing loss. Blocking this signal pathway with D-JNKI-1 is of potential therapeutic value for long-term protection of both the morphological integrity and physiological function of the organ of Corti during times of oxidative stress.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

BACKGROUND: The central function of dendritic cells (DC) in inducing and preventing immune responses makes them ideal therapeutic targets for the induction of immunologic tolerance. In a rat in vivo model, we showed that dexamethasone-treated DC (Dex-DC) induced indirect pathway-mediated regulation and that CD4+CD25+ T cells were involved in the observed effects. The aim of the present study was to investigate the mechanisms underlying the acquired immunoregulatory properties of Dex-DC in the rat and human experimental systems. METHODS: After treatment with dexamethasone (Dex), the immunogenicity of Dex-DC was analyzed in T-cell proliferation and two-step hyporesponsiveness induction assays. After carboxyfluorescein diacetate succinimidyl ester labeling, CD4+CD25+ regulatory T-cell expansion was analyzed by flow cytometry, and cytokine secretion was measured by ELISA. RESULTS: In this study, we demonstrate in vitro that rat Dex-DC induced selective expansion of CD4+CD25+ regulatory T cells, which were responsible for alloantigen-specific hyporesponsiveness. The induction of regulatory T-cell division by rat Dex-DC was due to secretion of interleukin (IL)-2 by DC. Similarly, in human studies, monocyte-derived Dex-DC were also poorly immunogenic, were able to induce T-cell anergy in vitro, and expand a population of T cells with regulatory functions. This was accompanied by a change in the cytokine profile in DC and T cells in favor of IL-10. CONCLUSION: These data suggest that Dex-DC induced tolerance by different mechanisms in the two systems studied. Both rat and human Dex-DC were able to induce and expand regulatory T cells, which occurred in an IL-2 dependent manner in the rat system.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

The tumor necrosis factor (TNF) family member B cell activating factor (BAFF) binds B cells and enhances B cell receptor-triggered proliferation. We find that B cell maturation antigen (BCMA), a predicted member of the TNF receptor family expressed primarily in mature B cells, is a receptor for BAFF. Although BCMA was previously localized to the Golgi apparatus, BCMA was found to be expressed on the surface of transfected cells and tonsillar B cells. A soluble form of BCMA, which inhibited the binding of BAFF to a B cell line, induced a dramatic decrease in the number of peripheral B cells when administered in vivo. Moreover, culturing splenic cells in the presence of BAFF increased survival of a percentage of the B cells. These results are consistent with a role for BAFF in maintaining homeostasis of the B cell population.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

In HLA-A2 individuals, the CD8 T cell response against the differentiation Ag Melan-A is mainly directed toward the peptide Melan-A26-35. The murine Melan-A24-33 sequence encodes a peptide that is identical with the human Melan-A26-35 decamer, except for a Thr-to-Ile substitution at the penultimate position. Here, we show that the murine Melan-A24-33 is naturally processed and presented by HLA-A2 molecules. Based on these findings, we compared the CD8 T cell response to human and murine Melan-A peptide by immunizing HLA-A2 transgenic mice. Even though the magnitude of the CTL response elicited by the murine Melan-A peptide was lower than the one elicited by the human Melan-A peptide, both populations of CTL recognized the corresponding immunizing peptide with the same functional avidity. Interestingly, CTL specific for the murine Melan-A peptide were completely cross-reactive against the orthologous human peptide, whereas anti-human Melan-A CTL recognized the murine Melan-A peptide with lower avidity. Structurally, this discrepancy could be explained by the fact that Ile32 of murine Melan-A24-33 created a larger TCR contact area than Thr34 of human Melan-A26-35. These data indicate that, even if immunizations with orthologous peptides can induce strong specific T cell responses, the quality of this response against syngeneic targets might be suboptimal due to the structure of the peptide-TCR contact surface.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

Cell death is essential for a plethora of physiological processes, and its deregulation characterizes numerous human diseases. Thus, the in-depth investigation of cell death and its mechanisms constitutes a formidable challenge for fundamental and applied biomedical research, and has tremendous implications for the development of novel therapeutic strategies. It is, therefore, of utmost importance to standardize the experimental procedures that identify dying and dead cells in cell cultures and/or in tissues, from model organisms and/or humans, in healthy and/or pathological scenarios. Thus far, dozens of methods have been proposed to quantify cell death-related parameters. However, no guidelines exist regarding their use and interpretation, and nobody has thoroughly annotated the experimental settings for which each of these techniques is most appropriate. Here, we provide a nonexhaustive comparison of methods to detect cell death with apoptotic or nonapoptotic morphologies, their advantages and pitfalls. These guidelines are intended for investigators who study cell death, as well as for reviewers who need to constructively critique scientific reports that deal with cellular demise. Given the difficulties in determining the exact number of cells that have passed the point-of-no-return of the signaling cascades leading to cell death, we emphasize the importance of performing multiple, methodologically unrelated assays to quantify dying and dead cells.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

Background: The G1-to-S transition of the cell cycle in the yeast Saccharomyces cerevisiae involves an extensive transcriptional program driven by transcription factors SBF (Swi4-Swi6) and MBF (Mbp1-Swi6). Activation of these factors ultimately depends on the G1 cyclin Cln3. Results: To determine the transcriptional targets of Cln3 and their dependence on SBF or MBF, we first have used DNA microarrays to interrogate gene expression upon Cln3 overexpression in synchronized cultures of strains lacking components of SBF and/or MBF. Secondly, we have integrated this expression dataset together with other heterogeneous data sources into a single probabilistic model based on Bayesian statistics. Our analysis has produced more than 200 transcription factor-target assignments, validated by ChIP assays and by functional enrichment. Our predictions show higher internal coherence and predictive power than previous classifications. Our results support a model whereby SBF and MBF may be differentially activated by Cln3. Conclusions: Integration of heterogeneous genome-wide datasets is key to building accurate transcriptional networks. By such integration, we provide here a reliable transcriptional network at the G1-to-S transition in the budding yeast cell cycle. Our results suggest that to improve the reliability of predictions we need to feed our models with more informative experimental data.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

A role for the NADPH oxidases NOX1 and NOX2 in liver fibrosis has been proposed, but the implication of NOX4 is poorly understood yet. The aim of this work was to study the functional role of NOX4 in different cell populations implicated in liver fibrosis: hepatic stellate cells (HSC), myofibroblats (MFBs) and hepatocytes. Two different mice models that develop spontaneous fibrosis (Mdr2−/−/p19ARF−/−, Stat3Δhc/Mdr2−/−) and a model of experimental induced fibrosis (CCl4) were used. In addition, gene expression in biopsies from chronic hepatitis C virus (HCV) patients or non-fibrotic liver samples was analyzed. Results have indicated that NOX4 expression was increased in the livers of all animal models, concomitantly with fibrosis development and TGF-β pathway activation. In vitro TGF-β-treated HSC increased NOX4 expression correlating with transdifferentiation to MFBs. Knockdown experiments revealed that NOX4 downstream TGF-β is necessary for HSC activation as well as for the maintenance of the MFB phenotype. NOX4 was not necessary for TGF-β-induced epithelial-mesenchymal transition (EMT), but was required for TGF-β-induced apoptosis in hepatocytes. Finally, NOX4 expression was elevated in patients with hepatitis C virus (HCV)-derived fibrosis, increasing along the fibrosis degree. In summary, fibrosis progression both in vitro and in vivo (animal models and patients) is accompanied by increased NOX4 expression, which mediates acquisition and maintenance of the MFB phenotype, as well as TGF-β-induced death of hepatocytes.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

Antigenic recognition by naive CD4+ T cells induces their proliferation and differentiation into functionally distinct T helper (Th) cell. Each CD4+ Th cell subset expresses specific transcription factors and produces signature cytokines that coordinate immune responses against encountered pathogens. Among the factors influencing CD4+ Th cell differentiation, Notch signaling pathway has been reported to play a role in the differentiation and function of multiple CD4+Thcell subsets. Notch signaling is an evolutionarily conserved cell-to-cell signaling cascade involved in many cell fate decision processes. How Notch signaling modulates the differentiation of CD4+ Th cell subsets and whether Notch signaling alone is sufficient or not for the differentiation of CD4+ Th cells is still a matter of debate. Th17 cells are a distinct subset of CD4+ Th cells. They play a role in the control of extracellular bacterial and fungal infections and may lead to inflammatory and autoimmune diseases if not properly regulated. Th17 cells are defined by the expression of RAR-related orphan receptor (ROR)a and RORyT transcription factors and their secretion of IL-17A, IL-17F cytokines. The involvement of Notch signaling in Th17 cell differentiation has mostly been studied in vitro. However, neither the experimental conditions when Notch signaling might be involved in Th17 cell differentiation in vitro and in vivo nor the precise role of Notch in this process remain clear. To better define how Notch signaling impacts Th17 differentiation, we used mice with T cell specific ablation of Notchl and Notch2 (N1 N2ACD4Cre) or of Notch transcriptional repressor RBP- JK (RBP-J ACD4Cre). We show that impaired Notch signaling in T cells, when TCR activating signal were reduced, increased RORyT and IL-17 mRNA levels during in vitro Th17 cell differentiation. Following immunization with OVA in CFA, an adjuvant that induces mostly Th17 cell response, increased IL-17A mRNA and intracellular IL-17A levels were observed in draining lymph nodes of Notch-deficient CD4+T cells. Our data suggest that Notch limited Th17 cell differentiation. Despite high levels of IL-17 mRNA and intracellular IL-17 proteins observed in Notch-deficient T cells, their release of Th17 cytokines ex vivo was markedly decreased, indicating a role for Notch signaling. During the second part of this thesis, we observed that the impact of Notch on Th17 cell differentiation and effector functions was context-dependent using different in vivo experimental models, in which Th17 cells and IL-17A were reported to contribute in the disease development. Collectively, our data reveal that Notch signaling controls the fine-tuning of Th17 cell differentiation and effector functions by limiting their differentiation but promoting selectively cytokine release through Notch-dependent mechanisms that still need to be defined. -- Lors d'une réponse immunitaire et grâce à la reconnaissance antigénique, les lymphocytes CD4+ T naïfs prolifèrent, puis se différencient en CD4+ T auxiliaires ("T helper" ou Th) fonctionnellement distincts. Chaque sous-population de lymphocytes CD4+ T auxiliaires exprime des facteurs de transcription et des cytokines spécifiques qui coordonnent la réponse immunitaire contre les pathogènes rencontrés. Parmi les facteurs influençant la différenciation des lymphocytes CD4+ T auxiliaires, la voie de signalisation Notch a été identifiée comme ayant un rôle dans la différenciation et la fonction des différents sous-types de cellules CD4+ T auxiliaires. La voie de signalisation Notch est une voie évolutivement conservée, qui est impliquée dans la signalisation entre les cellules et dans de nombreux processus de décisions cellulaires. La manière dont la voie de signalisation Notch régule la différenciation des lymphocytes CD4+ T en sous-types de cellules CD4+ auxiliaires, mais également la question de savoir si la voie de signalisation Notch est capable ou non d'induire la différenciation des cellules CD4+T auxiliaires, restent à débattre. Les cellules T auxiliaires 17 (Th17) sont un sous-type distinct de cellules CD4+T. Elles jouent un rôle important dans la défense immunitaire contre des pathogènes tels que les bactéries extracellulaires et les champignons. Une dérégulation de la réponse des cellules Th17 peut conduire à des inflammations mais également à des maladies auto-immunes. Les cellules Th17 sont définies par l'expression de leurs facteurs de transcription RAR-related orphan receptor (ROR)a, RORyT et par la sécrétion de cytokines comme IL-17A, IL-17F. Le rôle de la voie de signalisation Notch dans la différenciation des cellules Th17 a principalement été démontré in vitro. Malgré tout, ni les conditions expérimentales dans lesquelles cette voie pourrait être impliquée dans la différenciation des cellules Th17 in vitro et in vivo, mais également ni la fonction exacte de Notch dans ces processus, ne sont des questions résolues. Afin de mieux définir comment la voie de signalisation Notch est impliquée dans la différenciation des cellules Th17, nous avons utilisé des souris avec une déficience spécifique dans les cellules T des récepteurs Notchl et Notch2 (N1N2ACD4Cre) ou du répresseur transcriptionnel de Notch RBP-JK (RBP-J ACD4Cre). Nous avons montré que lorsque la voie de signalisation Notch est déficiente, les niveaux d'ARN messager (ARNm) de RORyT et de IL-17A sont augmentés dans les cellules Th17 pendant la différenciation in vitro, en présence de niveaux réduits des signaux activant les cellules T CD4+. Une augmentation dans les niveaux d'ARNm de IL-17A et de IL-17A intracellulaire au niveau protéinique a été observée dans les cellules T CD4+ Notch déficientes, au niveau des ganglions drainants après immunisation avec l'OVA dans le CFA, un adjuvant induisant une réponse des cellules Th17. Nos résultats suggèrent que Notch pourrait réguler négativement l'expression de IL-17A au niveau transcriptionnel mais également protéinique. Malgré une augmentation de IL-17A au niveau de l'ARNm et protéinique dans les cellules CD4+ T Notch déficientes, paradoxalement la sécrétion de IL-17A mais également de cytokines associées aux fonctions effectrices des cellules Th17 sont profondément diminuées 6X vivo, suggérant un rôle de la voie de signalisation Notch dans ce processus. Dans la deuxième partie de ce travail de thèse, nous avons observé que l'impact de Notch dans la différenciation des cellules Th17 et dans leurs fonctions effectrices était dépendant du contexte dans d'autres modèles expérimentaux in vivo, où les cellules Th17 et l'IL-17A ont été identifiées comme ar-.riCociêSM dans le développement ds la pathologie. En résumé, nous avons montré que la voie de la signalisation Notch contrôle la régulation précise de la différenciation des cellules Th17 en limitant leur différenciation, mais en promouvant sélectivement leur relâchement en cytokines associés aux cellules Th17 par l'intermédiaire de mécanismes dépendant de Notch, qui restent toujours à déterminer. -- Lors d'une réponse immunitaire et grâce à la reconnaissance antigénique, les lymphocytes CD4+ T naïfs prolifèrent, puis se différencient en CD4+ T auxiliaires ("T helper" ou Th) fonctionnellement distincts. Chaque sous-population de lymphocytes T auxiliaires exprime des facteurs de transcription et des cytokines spécifiques qui coordonnent une réponse immunitaire contre différents pathogènes. Les mécanismes liés à la différenciation des lymphocytes CD4+ T auxiliaires sont complexes et régulés. Une mauvaise régulation de la différenciation des lymphocytes CD4+ T auxiliaires peut conduire à des maladies auto-immunes, mais également à des processus inflammatoires. Parmi les facteurs influençant la différenciation des lymphocytes T auxiliaires, la voie de signalisation Notch a été identifiée comme ayant un rôle dans la différenciation et la fonction des différents sous-types de cellules CD4+ T auxiliaires. La voie de signalisation Notch est une voie évolutivement conservée, qui est impliquée dans la signalisation entre les cellules, mais également dans de nombreux processus de décisions cellulaires. Quelle est l'implication de la voie de signalisation Notch dans la différenciation des lymphocytes CD4+ en sous-types de cellules CD4+T auxiliaires et comment cette voie agit dans ce processus, sont des questions débattues. Les cellules T auxiliaires 17 (Th17) sont une sous-population distincte de lymphocytes CD4+. Elles jouent un rôle important dans la défense immunitaire contre les bactéries extracellulaires et les champignons. Une dérégulation de la réponse des cellules Th17 a été associée à des maladies auto-immunes et à l'inflammation. Les cellules Th17 sont définies par l'expression du facteur de transcription RAR-related orphan receptor (ROR)yT et des cytokines comme IL-17A, IL-17F. Le rôle de la voie de signalisation Notch dans la différenciation des cellules Th17 a été principalement démontré dans des études expérimentales in vitro. Malgré tout, les conditions expérimentales exactes dans lesquelles la voie de signalisation de Notch pourrait être impliquée dans la différenciation des cellules Th17, mais également le rôle de Notch dans ce processus ne sont pas encore clairement élucidés. Afin de mieux définir comment la voie de signalisation Notch est impliquée dans la différenciation des cellules Th17, nous avons utilisé des souris avec une déficience spécifique dans les cellules T des récepteurs Notchl et Notch2 (N1 N2ACD4Cre) ou du répresseur transcriptionnel de Notch RBP-JK (RBP-JACD4CRE). Nous avons montré que lorsque la voie de signalisation Notch est déficiente, les niveaux d'ARN messager (ARNm) de RORyT et de IL-17 sont augmentés dans les cellules Th17 pendant leur différenciation in vitro. Cet effet de Notch sur la transcription apparaît être facultatif lorsque les conditions environnementales sont en excès in vitro. Après immunisation avec un adjuvant qui induit principalement une réponse des cellules Th17, nous avons observé que les niveaux de ARNm de IL-17A et aussi de IL-17A intracellulaire au niveau protéinique étaient augmentés dans les ganglions drainants dans les cellules CD4+ Notch déficientes. Ces résultats suggèrent que Notch pourrait réguler négativement l'expression de IL- 17 au niveau transcriptionnel mais également protéinique. Malgré des niveaux plus élevés de IL- 17 ARNm et aussi IL-17A intracellulaire dans les cellules T Notch déficientes, le relâchement en cytokines Th17 est profondément diminué indiquant un rôle de la voie de signalisation Notch dans ces processus de sécrétion. Dans la deuxième partie de cette thèse, nous avons observé que le rôle de Notch dans ia différenciation dss cellules Ti,17 et dans leurs fonctions effectrices était dépendant du contexte dans d'autres modèles expérimentaux, qui ont été rapportés comme une réponse induisant des cellules Th17. En résumé, nos données montrent que la voie de la signalisation Notch contrôle la régulation précise de la différenciation des cellules Th17 en limitant leur différenciation mais en promouvant sélectivement le relâchement en cytokines associées aux cellules Th17 par des mécanismes dépendant de Notch qui restent toujours à déterminer. Par conséquent, l'inhibition de la voie de signalisation Notch pourrait être utilisée dans des situations inflammatoires ou d'auto-immunité où la réponse des cellules Th17 est exacerbée.

Relevância:

30.00% 30.00%

Publicador:

Resumo:

Experimental models demonstrated that therapeutic induction of CD8 T cell responses may offer protection against tumors or infectious diseases providing that T cells have sufficiently high TCR/CD8:pMHC avidity for efficient Ag recognition and consequently strong immune functions. However, comprehensive characterization of TCR/CD8:pMHC avidity in clinically relevant situations has remained elusive. In this study, using the novel NTA-His tag-containing multimer technology, we quantified the TCR:pMHC dissociation rates (koff) of tumor-specific vaccine-induced CD8 T cell clones (n = 139) derived from seven melanoma patients vaccinated with IFA, CpG, and the native/EAA or analog/ELA Melan-A(MART-1)(26-35) peptide, binding with low or high affinity to MHC, respectively. We observed substantial correlations between koff and Ca(2+) mobilization (p = 0.016) and target cell recognition (p < 0.0001), with the latter independently of the T cell differentiation state. Our strategy was successful in demonstrating that the type of peptide impacted on TCR/CD8:pMHC avidity, as tumor-reactive T cell clones derived from patients vaccinated with the low-affinity (native) peptide expressed slower koff rates than those derived from patients vaccinated with the high-affinity (analog) peptide (p < 0.0001). Furthermore, we observed that the low-affinity peptide promoted the selective differentiation of tumor-specific T cells bearing TCRs with high TCR/CD8:pMHC avidity (p < 0.0001). Altogether, TCR:pMHC interaction kinetics correlated strongly with T cell functions. Our study demonstrates the feasibility and usefulness of TCR/CD8:pMHC avidity assessment by NTA-His tag-containing multimers of naturally occurring polyclonal T cell responses, which represents a strong asset for the development of immunotherapy.