975 resultados para Alakaline phosphatase
Resumo:
Summary Acquisition of lineage-specific cell cycle duration is an important feature of metazoan development. In Caenorhabditis a/egans, differences in cell cycle duration are already apparent in two-cell stage embryos, when the larger anterior blastomere AB divides before the smaller posterior blastomere P1. This time difference is under the control of anterior-posterior (A-P) polarity cues set by the PAR proteins. The mechanism by which these cues regulate the cell cycle machinery differentially in AB and P1 are incompletely understood. Previous work established that retardation of P1 cell division is due in part to preferential activation of an ATL1/CHK-1 dependent checkpoint in P1 but how the remaining time difference is controlled was not known at the onset of my work. The principal line of work in this thesis established that differential timing relies also on a mechanism that promotes mitosis onset preferentially in AB. The polo-like kinase PLK-1, a positive regulator of mitotic entry, is distributed in an asymmetric manner in two-cell stage embryos, with more protein present in AB than in P1. We find that PLK-1 asymmetry is regulated by anterior-posterior (A-P) polarity cues through preferential protein retention in the embryo anterior. Importantly, mild inactivation of plk-1 by RNAi delays entry into mitosis in P1 but not in AB, in a manner that is independent of ATL-1/CHK-1. Together, these findings favor a model in which differential timing of mitotic entry in C. elegans embryos relies on two complementary mechanisms: ATL-1/CHK-1 dependent preferential retardation in P1 and PLK-1 dependent preferential promotion in AB, which together couple polarity cues and cell cycle progression during early development. Besides analyzing PLK-1 asymmetry and its role in differential timing of two-cells stage embryos, we also characterized t2190, a mutant that exhibits reduced differential timing between AB and P1. We found this mutant to be a new allele of par-1. Additionally, we analyzed the role of NMY-2 in regulating the asynchrony of two-cell stage embryos, which may be uncoupled from its role in A-P polarity establishment and carried out a preliminary analysis of the mechanism underlying CDC-25 asymmetry between AB and P,. Overall, our works bring new insights into the mechanism controlling cell cycle progression in early C. elegans embryos. As most of the players important in C. elegans are conserved in other organisms, analogous mechanisms may be utilized in polarized cells of other species. Résumé Au cours du développement, les processus de division cellulaire sont régulés dans l'espace et le temps afin d'aboutir à la formation d'un organisme fonctionnel. Chez les Métazoaires, l'un des mécanismes de contrôle s'effectue au niveau de la durée du cycle cellulaire, celle-ci étant specifiée selon la lignée cellulaire. L'embryon du nématode Caenorhabditis elegans apparaît comme un excellent modèle d'étude de la régulation temporelle du cycle cellulaire. En effet, suite à la première division du zygote, l'embryon est alors composé de deux cellules de taille et d'identité différentes, appelées blastomères AB et P1. Ces deux cellules vont ensuite se diviser de manière asynchrone, le grand blastomère antérieur AB se divisant plus rapidement que le petit blastomère postérieur P1. Cette asynchronie de division est sous le contrôle des protéines PAR qui sont impliquées dans l'établissement de l'axe antéro-postérieur de l'embryon. A ce jour, les mécanismes moléculaires gouvernant ce processus d'asynchronie ne sont que partiellement compris. Des études menées précédemment ont établit que le retard de division observé dans le petit blastomère postérieur P1 était dû, en partie, à l'activation préférentielle dans cette cellule de ATL-1/CHK-1, protéines contrôlant la réponse à des erreurs dans le processus de réplication de l'ADN. L'analyse des autres mécanismes responsables de la différence temporelle d'entrée en mitose des deux cellules a été entreprise au cours de cette thèse. Nous avons considéré la possibilité que l'asynchronie de division était du à l'entrée préférentielle en mitose du grand blastomère AB. Nous avons établi que la protéine kinase PLK-1 (polo-like kinase 1), impliquée dans la régulation positive de la mitose, était distribuée de manière asymétrique dans l'embryon deux cellules. PLK-1 est en effet enrichi dans le blastomère AB. Cette localisation asymétrique de PLK-1 est sous le contrôle des protéines PAR et semble établie via une rétention de PLK-1 dans la cellule AB. Par ailleurs, nous avons démontré que l'inactivation partielle de plk-7 par interférence à ARN (RNAi) conduit à un délai de l'entrée en mitose de la cellule P1 spécifiquement, indépendamment des protéines régulatrices ATL-1/CHK-1. En conclusion, nous proposons un modèle de régulation temporelle de l'entrée en mitose dans l'embryon deux cellules de C. elegans basé sur deux mécanismes complémentaires. Le premier implique l'activation préférentielle des protéines ATL-1/CHK-1, et conduit à un retard d'entrée en mitose spécifiquement dans la cellule P1. Le second est basé sur la localisation asymétrique de la protéine kinase PLK-1 dans la cellule AB et induit une entrée précoce en mitose de cette cellule. Par ailleurs, nous avons étudié un mutant appelé t2190 qui réduit la différence temporelle d'entrée en mitose entre les cellules AB et P1. Nous avons démontré que ce mutant correspondait à un nouvel allèle du Bene par-1. De plus, nous avons analysé le rôle de NMY-2, une protéine myosine qui agit comme moteur moléculaire sur les filaments d'active; dans la régulation de l'asynchronie de division des blastomères AB et P1, indépendamment de sa fonction dans l'établissement de l'axe antéro-postérieur. Par ailleurs, nous avons commencé l'étude du mécanisme moléculaire régulant la localisation asymétrique entre les cellules AB et P1 de la protéine phosphatase CDC25, qui est également un important régulateur de l'entrée en mitose. En conclusion, ce travail de thèse a permis une meilleure compréhension des mécanismes gouvernant la progression du cycle cellulaire dans l'embryon précoce de C. elegans. Etant donné que la plupart des protéines impliquées dans ces processus sont conservées chez d'autres organismes multicellulaires, il apparaît probable que les mécanismes moléculaires révélés dans cette étude soit aussi utilisés chez ceux-ci.
Resumo:
BACKGROUND: The thiomethyl group of S-adenosylmethionine is often recycled as methionine from methylthioadenosine. The corresponding pathway has been unravelled in Bacillus subtilis. However methylthioadenosine is subjected to alternative degradative pathways depending on the organism. RESULTS: This work uses genome in silico analysis to propose methionine salvage pathways for Klebsiella pneumoniae, Leptospira interrogans, Thermoanaerobacter tengcongensis and Xylella fastidiosa. Experiments performed with mutants of B. subtilis and Pseudomonas aeruginosa substantiate the hypotheses proposed. The enzymes that catalyze the reactions are recruited from a variety of origins. The first, ubiquitous, enzyme of the pathway, MtnA (methylthioribose-1-phosphate isomerase), belongs to a family of proteins related to eukaryotic intiation factor 2B alpha. mtnB codes for a methylthioribulose-1-phosphate dehydratase. Two reactions follow, that of an enolase and that of a phosphatase. While in B. subtilis this is performed by two distinct polypeptides, in the other organisms analyzed here an enolase-phosphatase yields 1,2-dihydroxy-3-keto-5-methylthiopentene. In the presence of dioxygen an aci-reductone dioxygenase yields the immediate precursor of methionine, ketomethylthiobutyrate. Under some conditions this enzyme produces carbon monoxide in B. subtilis, suggesting a route for a new gaseous mediator in bacteria. Ketomethylthiobutyrate is finally transaminated by an aminotransferase that exists usually as a broad specificity enzyme (often able to transaminate aromatic aminoacid keto-acid precursors or histidinol-phosphate). CONCLUSION: A functional methionine salvage pathway was experimentally demonstrated, for the first time, in P. aeruginosa. Apparently, methionine salvage pathways are frequent in Bacteria (and in Eukarya), with recruitment of different polypeptides to perform the needed reactions (an ancestor of a translation initiation factor and RuBisCO, as an enolase, in some Firmicutes). Many are highly dependent on the presence of oxygen, suggesting that the ecological niche may play an important role for the existence and/or metabolic steps of the pathway, even in phylogenetically related bacteria. Further work is needed to uncover the corresponding steps when dioxygen is scarce or absent (this is important to explore the presence of the pathway in Archaea). The thermophile T. tengcongensis, that thrives in the absence of oxygen, appears to possess the pathway. It will be an interesting link to uncover the missing reactions in anaerobic environments.
Resumo:
Islet-brain 1 [IB1; also termed c-Jun N-terminal kinase (JNK)-interacting protein 1 (JIP-1] is involved in the apoptotic signaling cascade of JNK and functions as a scaffold protein. It organizes several MAP kinases and the microtubule-transport motor protein kinesin and relates to other signal-transducing molecules such as the amyloid precursor protein. Here we have identified IB1/JIP-1 using different antibodies that reacted with either a monomeric or a dimeric form of IB1/JIP-1. By immunoelectron microscopy, differences in the subcellular localization were observed. The monomeric form was found in the cytoplasmic compartment and is associated with the cytoskeleton and with membranes, whereas the dimeric form was found in addition in nuclei. After treatment of mouse brain homogenates with alkaline phosphatase, the dimeric form disappeared and the monomeric form decreased its molecular weight, suggesting that an IB1/JIP-1 dimerization is phosphorylation dependent and that IB1 exists in several phospho- forms. N-methyl-D-aspartate receptor activation induced a dephosphorylation of IB1/JIP-1 in primary cultures of cortical neurons and reduced homodimerization. In conclusion, these data suggest that IB1/JIP-1 monomers and dimers may differ in compartmental localization and thus function as a scaffold protein of the JNK signaling cascade in the cytoplasm or as a transcription factor in nuclei.
Resumo:
Calcineurin is a key protein phosphatase required for hyphal growth and virulence in Aspergillus fumigatus, making it an attractive antifungal target. However, currently available calcineurin inhibitors, FK506 and cyclosporine A, are immunosuppressive, limiting usage in the treatment of patients with invasive aspergillosis. Therefore, the identification of endogenous inhibitors of calcineurin belonging to the calcipressin family is an important parallel strategy. We previously identified the gene cbpA as the A. fumigatus calcipressin member and showed its involvement in hyphal growth and calcium homeostasis. However, the mechanism of its activation/inhibition through phosphorylation and its interaction with calcineurin remains unknown. Here we show that A. fumigatus CbpA is phosphorylated at three distinct domains, including the conserved SP repeat motif (phosphorylated domain-I; PD-I), a filamentous fungal-specific domain (PD-II), and the C-terminal CIC motif (Calcipressin Inhibitor of Calcineurin; PD-III). While mutation of three phosphorylated residues (Ser208, Ser217, Ser223) in the PD-II did not affect CbpA function in vivo, mutation of the two phosphorylated serines (Ser156, Ser160) in the SP repeat motif caused reduced hyphal growth and sensitivity to oxidative stress. Mutational analysis in the key domains in calcineurin A (CnaA) and proteomic interaction studies confirmed the requirement of PxIxIT motif-binding residues (352-NIR-354) and the calcineurin B (CnaB)-binding helix residue (V371) for the binding of CbpA to CnaA. Additionally, while the calmodulin-binding residues (442-RVF-444) did not affect CbpA binding to CnaA, three mutations (T359P, H361L, and L365S) clustered between the CnaA catalytic and the CnaB-binding helix were also required for CbpA binding. This is the first study to analyze the phosphorylation status of calcipressin in filamentous fungi and identify the domains required for binding to calcineurin.
Resumo:
Protophloem is a specialized vascular tissue in growing plant organs, such as root meristems. In Arabidopsis mutants with impaired primary root protophloem differentiation, brevis radix (brx) and octopus (ops), meristematic activity and consequently overall root growth are strongly reduced. Second site mutation in the protophloem-specific presumed phosphoinositide 5-phosphatase COTYLEDON VASCULAR PATTERN 2 (CVP2), but not in its homolog CVP2-LIKE 1 (CVL1), partially rescues brx defects. Consistent with this finding, CVP2 hyperactivity in a wild-type background recreates a brx phenotype. Paradoxically, however, while cvp2 or cvl1 single mutants display no apparent root defects, the root phenotype of cvp2 cvl1 double mutants is similar to brx or ops, although, as expected, cvp2 cvl1 seedlings contain more phosphatidylinositol-4,5-biphosphate. Thus, tightly balanced phosphatidylinositol-4,5-biphosphate levels appear essential for proper protophloem differentiation. Genetically, OPS acts downstream of phosphatidylinositol-4,5-biphosphate levels, as cvp2 mutation cannot rescue ops defects, whereas increased OPS dose rescues cvp2 cvl1 defects. Finally, all three mutants display higher density and accelerated emergence of lateral roots, which correlates with increased auxin response in the root differentiation zone. This phenotype is also created by application of peptides that suppress protophloem differentiation, CLAVATA3/EMBRYO SURROUNDING REGION 26 (CLE26) and CLE45. Thus, local changes in the primary root protophloem systemically shape overall root system architecture.
Resumo:
The AMPK/Snf1 kinase has a central role in carbon metabolism homeostasis in Saccharomyces cerevisiae. In this study, we show that Snf1 activity, which requires phosphorylation of the Thr210 residue, is needed for protection against selenite toxicity. Such protection involves the Elm1 kinase, which acts upstream of Snf1 to activate it. Basal Snf1 activity is sufficient for the defense against selenite, although Snf1 Thr210 phosphorylation levels become increased at advanced treatment times, probably by inhibition of the Snf1 dephosphorylation function of the Reg1 phosphatase. Contrary to glucose deprivation, Snf1 remains cytosolic during selenite treatment, and the protective function of the kinase does not require its known nuclear effectors. Upon selenite treatment, a null snf1 mutant displays higher levels of oxidized versus reduced glutathione compared to wild type cells, and its hypersensitivity to the agent is rescued by overexpression of the glutathione reductase gene GLR1. In the presence of agents such as diethyl maleate or diamide, which cause alterations in glutathione redox homeostasis by increasing the levels of oxidized glutathione, yeast cells also require Snf1 in an Elm1-dependent manner for growth. These observations demonstrate a role of Snf1 to protect yeast cells in situations where glutathione-dependent redox homeostasis is altered to a more oxidant intracellular environment and associates AMPK to responses against oxidative stress.
Resumo:
Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is considered a multifunctional protein with defined functions in numerous mammalian cellular processes. GAPDH functional diversity depends on various factors such as covalent modifications, subcellular localization, oligomeric state and intracellular concentration of substrates or ligands, as well as protein-protein interactions. In bacteria, alternative GAPDH functions have been associated with its extracellular location in pathogens or probiotics. In this study, new intracellular functions of E. coli GAPDH were investigated following a proteomic approach aimed at identifying interacting partners using in vivo formaldehyde cross-linking followed by mass spectrometry. The identified proteins were involved in metabolic processes, protein synthesis and folding or DNA repair. Some interacting proteins were also identified in immunopurification experiments in the absence of cross-linking. Pull-down experiments and overlay immunoblotting were performed to further characterize the interaction with phosphoglycolate phosphatase (Gph). This enzyme is involved in the metabolism of 2-phosphoglycolate formed in the DNA repair of 3"-phosphoglycolate ends generated by bleomycin damage. We show that interaction between Gph and GAPDH increases in cells challenged with bleomycin, suggesting involvement of GAPDH in cellular processes linked to DNA repair mechanisms.
Resumo:
Concentration gradients provide spatial information for tissue patterning and cell organization, and their robustness under natural fluctuations is an evolutionary advantage. In rod-shaped Schizosaccharomyces pombe cells, the DYRK-family kinase Pom1 gradients control cell division timing and placement. Upon dephosphorylation by a Tea4-phosphatase complex, Pom1 associates with the plasma membrane at cell poles, where it diffuses and detaches upon auto-phosphorylation. Here, we demonstrate that Pom1 auto-phosphorylates intermolecularly, both in vitro and in vivo, which confers robustness to the gradient. Quantitative imaging reveals this robustness through two system's properties: The Pom1 gradient amplitude is inversely correlated with its decay length and is buffered against fluctuations in Tea4 levels. A theoretical model of Pom1 gradient formation through intermolecular auto-phosphorylation predicts both properties qualitatively and quantitatively. This provides a telling example where gradient robustness through super-linear decay, a principle hypothesized a decade ago, is achieved through autocatalysis. Concentration-dependent autocatalysis may be a widely used simple feedback to buffer biological activities.
Resumo:
Fibroblast growth factor receptors (FGFRs) are involved in proliferative and differentiation physiological responses. Deregulation of FGFR-mediated signaling involving the Ras/PI3K/Akt and the Ras/Raf/ERK MAPK pathways is causally involved in the development of several cancers. The caspase-3/p120 RasGAP module is a stress sensor switch. Under mild stress conditions, RasGAP is cleaved by caspase-3 at position 455. The resulting N-terminal fragment, called fragment N, stimulates anti-death signaling. When caspase-3 activity further increases, fragment N is cleaved at position 157. This generates a fragment, called N2, that no longer protects cells. Here, we investigated in Xenopus oocytes the impact of RasGAP and its fragments on FGF1-mediated signaling during G2/M cell cycle transition. RasGAP used its N-terminal Src homology 2 domain to bind FGFR once stimulated by FGF1, and this was necessary for the recruitment of Akt to the FGFR complex. Fragment N, which did not associate with the FGFR complex, favored FGF1-induced ERK stimulation, leading to accelerated G2/M transition. In contrast, fragment N2 bound the FGFR, and this inhibited mTORC2-dependent Akt Ser-473 phosphorylation and ERK2 phosphorylation but not phosphorylation of Akt on Thr-308. This also blocked cell cycle progression. Inhibition of Akt Ser-473 phosphorylation and entry into G2/M was relieved by PHLPP phosphatase inhibition. Hence, full-length RasGAP favors Akt activity by shielding it from deactivating phosphatases. This shielding was abrogated by fragment N2. These results highlight the role played by RasGAP in FGFR signaling and how graded stress intensities, by generating different RasGAP fragments, can positively or negatively impact this signaling.
Resumo:
BACKGROUND: Immune checkpoint inhibitors targeting programmed cell death 1 (PD1) or its ligand (PD-L1) showed activity in several cancer types. METHODS: We performed immunohistochemistry for CD3, CD8, CD20, HLA-DR, phosphatase and tensin homolog (PTEN), PD-1, and PD-L1 and pyrosequencing for assessment of the O6-methylguanine-methyltransferase (MGMT) promoter methylation status in 135 glioblastoma specimens (117 initial resection, 18 first local recurrence). PD-L1 gene expression was analyzed in 446 cases from The Cancer Genome Atlas. RESULTS: Diffuse/fibrillary PD-L1 expression of variable extent, with or without interspersed epithelioid tumor cells with membranous PD-L1 expression, was observed in 103 of 117 (88.0%) newly diagnosed and 13 of 18 (72.2%) recurrent glioblastoma specimens. Sparse-to-moderate density of tumor-infiltrating lymphocytes (TILs) was found in 85 of 117 (72.6%) specimens (CD3+ 78/117, 66.7%; CD8+ 52/117, 44.4%; CD20+ 27/117, 23.1%; PD1+ 34/117, 29.1%). PD1+ TIL density correlated positively with CD3+ (P < .001), CD8+ (P < .001), CD20+ TIL density (P < .001), and PTEN expression (P = .035). Enrichment of specimens with low PD-L1 gene expression levels was observed in the proneural and G-CIMP glioblastoma subtypes and in specimens with high PD-L1 gene expression in the mesenchymal subtype (P = 5.966e-10). No significant differences in PD-L1 expression or TIL density between initial and recurrent glioblastoma specimens or correlation of PD-L1 expression or TIL density with patient age or outcome were evident. CONCLUSION: TILs and PD-L1 expression are detectable in the majority of glioblastoma samples but are not related to outcome. Because the target is present, a clinical study with specific immune checkpoint inhibitors seems to be warranted in glioblastoma.
Resumo:
Proteasome inhibitors, used in cancer treatment for their proapoptotic effects, have anti-inflammatory and antifibrotic effects on animal models of various inflammatory and fibrotic diseases. Their effects in cells from patients affected by either inflammatory or fibrotic diseases have been poorly investigated. Nasal polyposis is a chronic inflammatory disease of the sinus mucosa characterized by tissue inflammation and remodeling. We tested the hypothesis that proteasome inhibition of nasal polyp fibroblasts might reduce their proliferation and inflammatory and fibrotic response. Accordingly, we investigated the effect of the proteasome inhibitor Z-Leu-Leu-Leu-B(OH)2 (MG262) on cell viability and proliferation and on the production of collagen and inflammatory cytokines in nasal polyp and nasal mucosa fibroblasts obtained from surgery specimens. MG262 reduced the viability of nasal mucosa and polyp fibroblasts concentration- and time-dependently, with marked effects after 48 h of treatment. The proteasome inhibitor bortezomib provoked a similar effect. MG262-induced cell death involved loss of mitochondrial membrane potential, caspase-3 and poly(ADP-ribose) polymerase activation, induction of c-Jun phosphorylation, and mitogen-activated protein kinase phosphatase-1 expression. Low concentrations of MG262 provoked growth arrest, inhibited DNA replication and retinoblastoma phosphorylation, and increased expression of the cell cycle inhibitors p21 and p27. MG262 concentration-dependently inhibited basal and transforming growth factor-β-induced collagen mRNA expression and interleukin (IL)-1β-induced production of IL-6, IL-8, monocyte chemoattractant protein-1, regulated on activation normal T cell expressed and secreted, and granulocyte/macrophage colony-stimulating factor in both fibroblast types. MG262 inhibited IL-1β/tumor necrosis factor-α-induced activation of nuclear factor-κB. We conclude that noncytotoxic treatment with MG262 reduces the proliferative, fibrotic, and inflammatory response of nasal fibroblasts, whereas high MG262 concentrations induce apoptosis.
Resumo:
Proteasome inhibitors, used in cancer treatment for their proapoptotic effects, have anti-inflammatory and antifibrotic effects on animal models of various inflammatory and fibrotic diseases. Their effects in cells from patients affected by either inflammatory or fibrotic diseases have been poorly investigated. Nasal polyposis is a chronic inflammatory disease of the sinus mucosa characterized by tissue inflammation and remodeling. We tested the hypothesis that proteasome inhibition of nasal polyp fibroblasts might reduce their proliferation and inflammatory and fibrotic response. Accordingly, we investigated the effect of the proteasome inhibitor Z-Leu-Leu-Leu-B(OH)2 (MG262) on cell viability and proliferation and on the production of collagen and inflammatory cytokines in nasal polyp and nasal mucosa fibroblasts obtained from surgery specimens. MG262 reduced the viability of nasal mucosa and polyp fibroblasts concentration- and time-dependently, with marked effects after 48 h of treatment. The proteasome inhibitor bortezomib provoked a similar effect. MG262-induced cell death involved loss of mitochondrial membrane potential, caspase-3 and poly(ADP-ribose) polymerase activation, induction of c-Jun phosphorylation, and mitogen-activated protein kinase phosphatase-1 expression. Low concentrations of MG262 provoked growth arrest, inhibited DNA replication and retinoblastoma phosphorylation, and increased expression of the cell cycle inhibitors p21 and p27. MG262 concentration-dependently inhibited basal and transforming growth factor-β-induced collagen mRNA expression and interleukin (IL)-1β-induced production of IL-6, IL-8, monocyte chemoattractant protein-1, regulated on activation normal T cell expressed and secreted, and granulocyte/macrophage colony-stimulating factor in both fibroblast types. MG262 inhibited IL-1β/tumor necrosis factor-α-induced activation of nuclear factor-κB. We conclude that noncytotoxic treatment with MG262 reduces the proliferative, fibrotic, and inflammatory response of nasal fibroblasts, whereas high MG262 concentrations induce apoptosis.
Resumo:
Inhibitors of the HIV aspartyl protease [HIV protease inhibitors (HIV-PIs)] are the cornerstone of treatment for HIV. Beyond their well-defined antiretroviral activity, these drugs have additional effects that modulate cell viability and homeostasis. However, little is known about the virus-independent pathways engaged by these molecules. Here we show that the HIV-PI Nelfinavir decreases translation rates and promotes a transcriptional program characteristic of the integrated stress response (ISR). Mice treated with Nelfinavir display hallmarks of this stress response in the liver, including α subunit of translation initiation factor 2 (eIF2α) phosphorylation, activating transcription factor-4 (ATF4) induction, and increased expression of known downstream targets. Mechanistically, Nelfinavir-mediated ISR bypassed direct activation of the eIF2α stress kinases and instead relied on the inhibition of the constitutive eIF2α dephosphorylation and down-regulation of the phophatase cofactor CReP (Constitutive Repressor of eIF2α Phosphorylation; also known as PPP1R15B). These findings demonstrate that the modulation of eIF2α-specific phosphatase cofactor activity can be a rheostat of cellular homeostasis that initiates a functional ISR and suggest that the HIV-PIs could be repositioned as therapeutics in human diseases to modulate translation rates and stress responses.
Resumo:
Les antipsychotiques atypiques, de deuxième génération, ont largement contribué à améliorer le traitement des patients souffrant de schizophrénie. Cependant, leur mécanisme d'action reste mal compris et leurs effets secondaires sont importants, notamment la prise de poids. CRTC1 (CREB-regulated transcription coactivator 1), aussi appelé TORC1 (transducers of regulated CREB activity 1), est un coactivateur de CREB. Il régule la transcription de Bdnf1 qui joue un rôle essentiel dans le contrôle de la balance énergétique dépendant du VMH 2, 3, 4. Nous pensons que CRTC1 est impliqué dans la prise de poids induite par certains antipsychotiques. En effet, il a été démontré que les souris Crtc1-/- devenaient hyperphagiques et obèses 5, 6, que la régulation de l'activité de CRTC1 se faisait par l'AMPK et que les antipsychotiques atypiques activaient cette kinase dans l'hypothalamus.7 L'AMPK de l'hypothalamus est liée à la régulation de la prise alimentaire, elle inverse l'action de la leptine, hormone anorexigène. Suite à ces constatations, nous proposons de suivre l'hypothèse de travail suivante : l'activation de l'AMPK par les antipsychotiques atypiques dans l'hypothalamus peut maintenir la phosphorylation de CRTC1 et le bloquer dans le cytoplasme, l'empêchant ainsi d'activer les gènes anorexigènes, comme Bdnf par exemple. En effet, la forme phosphorylée inactive de CRTC1 est séquestrée dans le cytoplasme et sa migration dans le noyau nécessite en même temps l'activation de la phosphatase calcineurine et l'inactivation des kinases de la famille de l'AMPK. Dans le travail que nous avons entamé, nous cherchons donc, par western blot et par immunohistochimie, à comprendre si les antipsychotiques atypiques inactivent CRTC1 en induisant sa phosphorylation par l'AMPK et sa rétention dans le cytoplasme.
Resumo:
Notre système immunitaire joue un rôle important pour la protection envers les maladies infectieuses. Au cours d'une réponse à une infection primaire, des cellules B et des cellules T spécifiques, dirigées contre le pathogène en question, sont générées et certaines d'entre elles deviennent des cellules dites mémoires. Leur fonction est de nous protéger contre une nouvelle infection avec le même pathogène, une infection secondaire. Dans certaines situations, comme c'est par exemple le cas avec la grippe, les pathogènes ne sont pas toujours complètement identiques et les cellules mémoires ne sont pas à même d'assurer leur rôle protecteur et d'empêcher une réinfection. Pourtant, on ne sait à l'heure actuelle que très peu comment une immunité acquise, mais non protectrice, influence le développement d'une réponse immunitaire ultérieure. Dans la première partie de cette thèse, nous avons étudié comment les cellules T mémoires cytotoxiques altèrent la réponse de cellules T cytotoxiques nouvellement induites. Au cours d'une réaction immunitaire dirigée contre une infection primaire, un vaste répertoire de lymphocytes T est créé, constitué de cellules T possédant divers degrés d'affinité pour le pathogène. Lors d'une infection secondaire, seules les cellules T ayant une forte affinité pour le pathogène participent à la réponse. Nous avons pu démontrer que ce phénomène de restriction du répertoire des cellules T est principalement causé par les cellules T mémoires qui sont à même de reconnaître un antigène pathogénique présent dans les deux infections. Dans un deuxième projet, nous avons étudié comment l'absence de PTPN2 influence la réponse des cellules T. Chez l'homme, une mutation dans le gène de PTPN2 est associée à des maladies auto-immunes et résulte en une activité réduite de cette phosphatase dans les lymphocytes T. Nous avons montré que la baisse d'activité de la phosphatase PTNP2 conduit à une meilleure expansion des cellules T ayant une qualité comparable à des cellules T auto-antigène spécifiques. De plus, nous avons observé que la survie de ces cellules T effectues ayant une phosphatase diminuée est nettement améliorée. Cela peut conduire à une réponse immunitaire plus efficace ou, éventuellement, à une pathologie auto-immune plus grave. En outre, nos résultats montrent qu'en manipulant l'activité de cette phosphatase, il est possible d'augmenter l'efficacité du transfert des cellules T dans un hôte receveur. Un tel transfert de cellules T est pratiqué chez des patients atteints de tumeurs. Nos travaux suggèrent que la manipulation de la phosphatase PTPN2 pourrait donc représenter une approche thérapeutique novatrice et prometteuse. -- Notre système immunitaire joue un rôle important pour la protection contre les maladies. Les cellules T CD8+ ont une importance primordiale pour le contrôle d'infections primaires causées par des virus ou bactéries, mais également contre certaines tumeurs. Par conséquent, mieux comprendre les exigences nécessaires à l'induction de bonnes réponses des cellules T CD8 pourrait nous permettre de construire des vaccins contre les pathogènes contre lesquels nous n'avons pour l'instant pas de vaccins mais aussi d'améliorer les réactions immunitaires dirigées anti-tumorales. Dans la première partie de cette thèse, nous avons étudié l'influence qu'une immunité préexistante a sur la réponse des cellules T CD8. Nous sommes souvent exposés à des pathogènes qui sont similaires mais pas identiques à ceux que nous avons rencontrés auparavant. De telles infections hétérologues ne sont pas l'objet de beaucoup d'études et certains exemples indiquent même qu'une immunité préexistante partielle peut mener à une aggravation de la maladie. Nous avons étudié le répertoire des lymphocytes T CD8 qui sont générés lors d'une rencontre avec un nouvel antigène, et ce en comparant infection primaire et secondaire. En utilisant le modèle expérimental d'infections à Listeria monocytogenes, nous avons pu montrer que lors d'une infection primaire, un répertoire diversifié comprenant des cellules T CD8 de forte et faible affinité est constitué. Au contraire, dans le cas d'une infection secondaire, le répertoire des cellules T est fortement limité et seulement les lymphocytes T de forte affinité sont impliqués dans la réponse immunitaire. Nous avons pu démontrer que ces Rangements sont provoqués par des cellules T CD8 mémoires capables de reconnaître un antigène présent dans les deux infections. Cette augmentation du seuil d'activation des cellules effectrices est majoritairement causée par les lymphocytes T CD8 mémoires non transférables. Ces observations indiquent que les vaccins visant à induire des cellules T anti-tumorales de faible affinité seraient inefficaces si le vaccin contient des épitopes contre lesquels il existe une mémoire immunologique. Les réponses immunitaires conduites par les cellules T contre les antigènes tumoraux dépendent des cellules T CD8 de faible réactivité contre les antigènes tumoraux puisque les cellules à forte réactivité sont éliminées par les mécanismes de tolérance. Nous basant sur l'existence dans la littérature de preuves indiquant que PTPN2 influence la réponse des cellules T de faible affinité, nous nous sommes intéressés à comprendre comment PTPN2 impacte les réponses des cellules T CD8 en général. Nous avons remarqué que des cellules T CD8 déficientes en PTPN2 exhibent une meilleure capacité à proliférer suite à une faible ou courte stimulation du récepteur des lymphocytes T. La phase effectrice est prolongée et la contraction retardée résultant ainsi à globalement plus de cellules effectrices. Ce phénomène est également accompagné d'une meilleure survie des cellules effectrices de différentiation terminale. Une fois transférées dans un nouvel hôte receveur, les cellules effectrices terminales KLRG1+CD127- déficientes en phosphatase PTPN2 peuvent survivre et se transformer en cellules mémoires CD127+ fonctionnelles. De façon inattendue, nous avons découvert que l'élimination de PTPN2 améliore l'efficacité du transfert et la formation des cellules mémoires ainsi que leur capacité protectrice. Manipuler l'activité de cette phosphatase apparaît donc comme une approche intéressante et prometteuse pour la thérapie cellulaire par transfert adoptif de lymphocytes T. Nos observations montrent que la manipulation d'un facteur intrinsèque, l'absence de PTPN2, peut, dans certaines circonstances, améliorer la réponse des cellules T. Une meilleure connaissance des mécanismes contrôlant la réponse des lymphocytes T CD8 pourrait donc permettre la manipulation de ces derniers et conduire à des réponses immunitaires plus vigoureuses. Si ces réponses sont déclenchées par l'utilisation de vaccins, il est nécessaire de considérer l'historique d'une exposition préalable à des agents pathogènes ou à des vaccins puisque celle-ci peut, comme nous l'avons démontré, influencer le répertoire des cellules T recrutées dans la réponse immunitaire et, par conséquent, modifier l'aptitude de notre système immunitaire à faire face à une infection. -- Our immune system plays an important role in the protection from disease. CD8 T cells are critical for the control of primary infections with most viruses and certain bacteria as well as against some tumors. Therefore, better knowledge of CD8 T cell responses might enable us to generate vaccines against pathogens for which currently no vaccines are available or to improve anti-tumor immune responses. In the first part of this thesis we addressed the issue how previously acquired immunity impacts on the response of CD8 T cells. We are often exposed to pathogens that are related but not identical to the previously encountered ones. Such heterologous infections are not well studied and there are some indications that partial pre-existing immunity may in some cases even lead to an enhancement of disease. We specifically studied the T cell repertoire of CD8 T cells that are responding to a newly encountered antigen in secondary compared to primary infections. Using the experimental model of Listeria monocytogenes infections, we showed that in primary infections a wide repertoire including high and low affinity CD8 T cells is recruited into the immune response. In contrast to this, in secondary infections, the T cell repertoire is severely restricted and only T cells of high affinity are responding. We were able to pinpoint this difference to the presence of memory CD8 T cells that recognize an antigen that is shared between the two subsequent infections. This increase in the activation threshold was most effectively mediated via non-transferable memory CD8 T cells. This would argue that vaccines targeting low affinity tumor-specific T cells would fail if the vaccine contains previously encountered CD8 T cell epitopes. T cell mediated immune responses to tumor antigen rely often on T cells which weakly react to tumor antigen as high affinity T cells are eliminated by tolerance mechanisms. Following indication in the literature that PTPN2 impacts on the response of such weakly antigen-reactive T cells, we investigated how PTPN2 impacts in general the response of CD8 T cells. We observed that CD8 T cells lacking PTPN2 show an enhanced expansion following weak or short-term T cell receptor stimulation. The effector phase is prolonged and contraction delayed thus resulting in overall more effector cells. This is accompanied by a better survival of terminal effector cells. When transferred into new recipients, KLRG1+CD127- terminal effector cells lacking PTPN2 can survive and convert into CD127+ functional memory cells. Surprisingly, we discovered that elimination of PTPN2 enhances the transfer efficacy and formation of memory cells as well as the protective capacity. Targeting PTPN2 might thus be a promising approach for adoptive T cell therapy. Our observations show how the manipulation of an intrinsic factor, the absence of PTPN2, can enhance T cell responses under certain circumstances. A better understanding of underlying mechanisms for the control of CDS T cell responses might enable the manipulation of these and allow for more powerful responses. If these responses are induced through vaccines it is imperative that the previous history of exposure to pathogens or vaccines is considered as it can, as we have shown in this thesis, influence the recruited T cell repertoire and thus possibly the ability to handle the infection.