• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 22
  • 12
  • 2
  • 1
  • 1
  • Tagged with
  • 42
  • 14
  • 9
  • 9
  • 8
  • 8
  • 7
  • 6
  • 6
  • 6
  • 6
  • 5
  • 5
  • 5
  • 5
  • 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

The Effect of STAT5 on Inflammation-Related Gene Expression in Diabetic Mouse Kidneys

Shaw, Samantha J. 12 June 2014 (has links)
No description available.
32

Sélection et caractérisation d’aptamères oligonucléotidiques régulateurs de la protéine STAT5B, impliquée dans les leucémies / Selection and characterization of DNA aptamers regulating STAT5B, a protein involved in leukemias

Loussouarn, Claire 20 March 2014 (has links)
Les cancers, qu’il s’agisse de leucémies ou de tumeurs solides, sont le résultat de proliférations cellulaires anormales et non contrôlées au sein des tissus. Ces proliférations anarchiques sont le reflet d’une surexpression et/ou sur-activation de protéines intracellulaires engendrées par un événement oncogénique. Aujourd’hui encore il est donc nécessaire de trouver de nouvelles molécules à usage thérapeutique ciblant spécifiquement ces protéines. C’est dans ce contexte que les facteurs de transcription STAT5 constituent de véritables cibles de choix puisque ces protéines participent activement à la leucogénèse. L’implication directe des protéines STAT5 dans la génèse des leucémies a été démontrée par l’utilisation de formes mutées constitutivement active de STAT5. Les facteurs de transcription STAT5 jouent un rôle essentiel dans la voie de signalisation JAK/STAT. Cette voie aboutit à la régulation de grandes fonctions biologiques telles que la prolifération cellulaire, la différenciation cellulaire ou encore l’apoptose. L’objectif de ce projet consiste donc à cibler spécifiquement les protéines STAT5 dans le but de rétablir le processus de mort cellulaire et empêcher la prolifération des cellules cancéreuses. Les inhibiteurs spécifiques des protéines STAT5 sont sélectionnés selon la méthode SELEX qui permet d’isoler des ligands structurés de forte affinité pour la protéine. L’affinité et la spécificité de ces inhibiteurs, appelés aptamères, sont caractérisées à partir de modèles cellulaires de leucémies dépendant de l’activité des facteurs de transcription STAT5. Les aptamères sont aujourd’hui de véritables outils thérapeutiques en pleine évolution. / Leukemias are due to abnormal cell proliferation, which is the result of intracellular over-expression or excessive activation of protein due to oncogenic event. Still today, it is necessary to find new therapeutic molecules, which specifically target these proteins. STAT5, via the JAK/STAT signaling pathway, controls fundamental cellular processes, including .cell survival, proliferation and differentiation. To struggle against tumorigenesis, JAK/STAT signaling pathway has to be inhibited. The aim of this project is to target specifically STAT5 factors to restore healthy signal transduction. We generated aptamers by an iterative in vitro selection. Aptamers are short-structured single strand DNAs or RNAs that bind with high affinity and specificity to their target. Once STAT5B recombinant proteins are produced, they are subjected to SELEX process. The number of rounds depends on various parameters. After seven rounds, two sequences are retrieved. The specificity and affinity of these aptamers are assessed by fluorescent immunoassays. Binding affinity and kinetics of interaction are characterized by SPR. Aptamer anti proliferative effects are determined by evaluation of the growth of cells depending on STAT5. Finally, we developed several .assays aiming at understanding the mechanism of an aptamer action on STAT5B such as phosphorylation measurement and EMSA. Aptamers are now emerging therapeutic tools; they exhibit significant advantages relative to protein therapeutics.
33

Régulation de l'apoptose des lymphocytes T par GIMAP5 (GTPase of Immune Associated Nucleotide Binding Protein 5) / Regulation of T Lymphocytes Apoptosis by GIMAP5 (GTPase of Immune Associated Nucleotide Binding Protein 5)

Chen, Xi Lin January 2015 (has links)
Abstract : Long-term survival of T lymphocytes in a quiescent state is essential to maintain their cell numbers in secondary lymphoid organs. Interaction of the T cell antigen receptor (TCR) with self-peptide/MHC synergizes with IL-7-induced anti-apoptotic signals to promote T cell survival. These extrinsic stimuli are also implicated in T cell metabolism and survival by regulating several signaling pathways including the phosphatidyl-inositol-3 kinase (PI3K)/Akt pathway. In mice and in rats, loss of functional GTPase of the immune associated nucleotide binding protein 5 (GIMAP5) causes peripheral T lymphopenia due to spontaneous death of T cells. The underlying mechanisms responsible for the pro-survival function of GIMAP5 in T lymphocytes remain largely unknown. Previous work from my laboratory has shown that T cells from GIMAP5-deficient rats show reduced influx of calcium (Ca[superscript 2+]) from the extracellular milieu following stimulation of the TCR complex. In this thesis, I characterized the mechanism by which GIMAP5 regulates Ca[superscript 2+] homeostasis, and elucidated the signaling pathways modulated by GIMAP5 to facilitate the survival of T cells. Firstly, I investigated if GIMAP5 prevents apoptotic death of T lymphocytes by affecting the Ca[superscript 2+] buffering capacity of mitochondria, which is required for sustained Ca[superscript 2+] influx via the plasma membrane channels. I observed that mitochondrial Ca[superscript 2+] accumulation following capacitative Ca[superscript 2+] entry is defective in T cells from Gimap5 deficient rats. Disruption of microtubules, but not the actin cytoskeleton, abrogated Ca[superscript 2+] sequestration by mitochondria in T cells from control but not Gimap5 deficient mice. Similarly, mice lacking functional GIMAP5 displayed defective T cell development and Ca[superscript 2+] influx. Furthermore, I observed that the proximal signaling events following TCR stimulation was reduced and was accompanied by defective proliferation in T cells from Gimap5 deficient mice. Additionally, IL-7-induced STAT5 phosphorylation was decreased in CD4[superscript +] T cells from Gimap5 deficient mice. I also showed that loss of functional Gimap5 results in increased basal activation of mammalian target of rapamycin (mTOR), independent of protein phosphatase 2A (PP2A) or AMP-activated protein kinase (AMPK). Instead, the constitutive activation the PI3K pathway contributed to the spontaneous high mTOR activation. Collectively, my observations suggest that the pro-survival function of GIMAP5 in T-lymphocytes may be linked to the regulation of diverse signaling pathways in a context dependent manner. GIMAP5 also facilitates microtubule-dependent mitochondrial buffering of Ca[superscript 2+] following capacitative entry. GIMAP5 is required to integrate the survival signals generated following activation through TCR and IL-7R. / Résumé : La survie à long terme des lymphocytes T en état de repos est essentielle pour maintenir leurs nombres dans les organes lymphoïdes secondaires. Le récepteur antigénique des cellules T (TCR) en contact avec les peptides du soi / CMH et en synergie avec l'IL-7 induit des signaux anti-apoptotiques pour favoriser la survie des cellules T. Ces stimuli extrinsèques sont également impliqués dans le métabolisme et la survie des cellules T grâce à la régulation de plusieurs voies de signalisation dont la voie phosphatidyl-inositol-3 kinase (PI3K) /AKT. Chez la souris et chez le rat, la perte de l’activité de GIMAP5 (GTPase of Immune Associated Nucleotide Binding Protein 5), provoque une lymphopénie T périphérique en raison de la mort spontanée des cellules T. Le mécanisme sous-jacent responsable de la fonction de survie de GIMAP5 dans les lymphocytes T reste largement inconnu. Nous avons observé que les cellules de rats déficients en GIMAP5, après stimulation par complexe TCR, montrent un afflux de calcium (Ca[indice supérieur 2+]) réduit provenant du milieu extracellulaire. Dans cette thèse, J’ai caractérisé le mécanisme d’action de GIMAP5 dans la régulation de l'homéostasie du Ca[indice supérieur 2+], ainsi que les voies de signalisation modulées par GIMAP5 pour faciliter la survie des cellules T. Tout d'abord, j’ai étudié si GIMAP5 empêche l’apoptose des lymphocytes T en affectant la capacité des mitochondries à réguler la concentration du Ca[indice supérieur 2+], ce qui est nécessaire pour soutenir l’influx de Ca[indice supérieur 2+]. J’ai trouvé que l’accumulation du Ca[indice supérieur 2+] mitochondrial après l’entrée capacitive de Ca[indice supérieur 2+] est défectueuse dans les cellules T de rat déficientes en Gimap5. La disruption des microtubules, mais pas du cytosquelette d'actine, abroge la séquestration du Ca[indice supérieur 2+] mitochondrial dans les cellules T primaires de rat, mais pas dans les cellules T déficientes en Gimap5. J’ai observé que les cellules T provenant de souris déficientes en Gimap5 démontrent une diminution de l’entrée de Ca[indice supérieur 2+]. De plus, la prolifération des cellules T déficientes en Gimap5 est diminuée suite à la stimulation du TCR. En outre, la phosphorylation de STAT5 induit par l'IL-7 est diminuée dans les cellules T CD4[indice supérieur +] de souris déficientes en Gimap5. Également, la perte de Gimap5 aboutit à une activation accrue de la cible mammalienne de la rapamycine (mTOR), indépendamment de la protéine phosphatase 2A (PP2A) ou de la protéine kinase activée par l'AMP (AMPK). Au lieu de cela, l'activation constitutive de la voie PI3K contribue à une forte activation spontanée de mTOR. Collectivement, la fonction de survie de GIMAP5 dans les lymphocytes T peut être liée à la régulation de différentes voies de signalisation. GIMAP5 facilite la fonction, microtubule dépendant, des mitochondries dans leurs actions de régulation du Ca[indice supérieur 2+] après l’entrée capacitive de Ca[indice supérieur 2+]. GIMAP5 est nécessaire pour intégrer les signaux de survie produits suite à l'activation du TCR et de l’IL-7R, qui pourrait être associée à la régulation de l'activité PI3K / AKT / mTOR.
34

The BTB/POZ Transcription Factor Miz-1 Is Required To Regulate The Commitment, Survival And Differentiation Of Early B And T Cell Lineages

Saba, Ingrid 01 1900 (has links)
Les lymphocytes B et T sont issus de cellules progénitrices lymphoïdes de la moelle osseuse qui se différencient grâce à l’action de facteurs de transcription, cytokines et voies de signalisation, dont l’interleukine-7 (IL-7)/IL-7 récepteur (IL-7R). Le facteur de transcription c-Myc est exprimé par les cellules lymphoïdes et contrôle leur croissance et leur différenciation. Cette régulation transcriptionnelle peut être coordonnée par le complexe c-Myc/Myc-Interacting Zinc finger protein-1 (Miz-1). Le but de ce projet était de comprendre les mécanismes qui impliquent Miz-1 et le complexe c-Myc/Miz-1 dans le développement des lymphocytes B et T. Pour réaliser ce projet, des souris déficientes pour le domaine de transactivation de Miz-1 (Miz-1POZ) et des souris à allèles mutantes pour c-MycV394D, mutation qui empêche l’interaction avec Miz-1, ont été générées. La caractérisation des souris Miz 1POZ a démontré que l’inactivation de Miz-1 perturbe le développement des lymphocytes B et T aux stades précoces de leur différenciation qui dépend de l’IL-7. L’analyse de la cascade de signalisation IL-7/IL-7R a montré que ces cellules surexpriment la protéine inhibitrice SOCS1 qui empêche la phosphorylation de STAT5 et perturbe la régulation à la hausse de la protéine de survie Bcl-2. De plus, Miz-1 se lie directement au promoteur de SOCS1 et contrôle son activité. En plus de contrôler l’axe IL-7/IL-7R/STAT5/Bcl-2 spécifiquement aux stades précoces du développement afin d’assurer la survie des progéniteurs B et T, Miz-1 régule l’axe EBF/Pax-5/Rag-1/2 dans les cellules B afin de coordonner les signaux nécessaires pour la différenciation des cellules immatures. La caractérisation des souris c-MycV394D a montré, quant à elle, que les fonctions de Miz-1 dans les cellules B et T semblent indépendantes de c-Myc. Les cellules T des souris Miz-1POZ ont un défaut de différenciation additionnel au niveau de la -sélection, étape où les signaux initiés par le TCR remplacent ceux induits par IL-7 pour assurer la prolifération et la différenciation des thymocytes en stades plus matures. À cette étape du développement, une forme fonctionnelle de Miz-1 semble être requise pour contrôler le niveau d’activation de la voie p53, induite lors du processus de réarrangement V(D)J du TCR. L’expression de gènes pro-apoptotiques PUMA, NOXA, Bax et du régulateur de cycle cellulaire p21CIP1 est régulée à la hausse dans les cellules des souris Miz-1POZ. Ceci provoque un débalancement pro-apoptotique qui empêche la progression du cycle cellulaire des cellules TCR-positives. La survie des cellules peut être rétablie à ce stade de différenciation en assurant une coordination adéquate entre les signaux initiés par l’introduction d’un TCR transgénique et d’un transgène codant pour la protéine Bcl-2. En conclusion, ces études ont montré que Miz-1 intervient à deux niveaux du développement lymphoïde: l’un précoce en contrôlant la signalisation induite par l’IL-7 dans les cellules B et T, en plus de l’axe EBF/Pax-5/Rag-1/2 dans les cellules B; et l’autre tardif, en coordonnant les signaux de survie issus par le TCR et p53 dans les cellules T. Étant donné que les thymocytes et lymphocytes B immatures sont sujets à plusieurs rondes de prolifération, ces études serviront à mieux comprendre l’implication des régulateurs du cycle cellulaire comme c-Myc et Miz-1 dans la génération des signaux nécessaires à la différenciation non aberrante et à la survie des ces cellules. Enfin, les modèles expérimentaux, souris déficientes ou à allèles mutantes, utilisés pour ce travail permettront de mieux définir les bases moléculaires de la transformation maligne des lymphocytes B et T et de révéler les mécanismes conduisant au lymphome. / Signaling pathways control the differentiation and proliferation of blood cells, like B and T lymphocytes. They converge into regulating the activity of transcription factors that influence ultimately gene expression patterns. The transcription factor c-Myc is a central regulator of cellular proliferation and growth, and its deregulated expression has been demonstrated to be involved in many types of cancers, in particular lymphoma. Recent studies have shown that repression by c-Myc can be mediated by a complex formed with the BTB/POZ domain transcription factor Miz-1 (Myc Interacting Zinc finger protein-1). Given that both c-Myc and Miz-1 proteins are expressed in lymphoid precursors and since c-Myc has been shown to be important for B- and T-cell development, the aim of this thesis was to investigate the role of Miz-1 and the c-Myc/Miz-1 complex in regulating B and T cell survival, commitment and differentiation. To do so, mice expressing a non-functional Miz-1 protein lacking the BTB/POZ domain (Miz-1POZ) and knock-in mice expressing a mutant c-MycV394D allele that no longer interacts with Miz-1 were generated. B- and T-cell development requires the coordinated action of transcription factors and cytokines, in particular interleukin-7 (IL-7). The studies presented in this work demonstrated that mice deficient for the BTB/POZ domain of transcription factor Miz-1 almost entirely lack follicular B cells and T cells, since their progenitors fail to activate the JAK/STAT5 pathway and to up-regulate Bcl-2 upon IL-7 stimulation. Miz-1 exerts a dual role in the IL-7 receptor (IL-7R) pathway by directly repressing the JAK inhibitor SOCS1 and by activating Bcl-2 expression. In B cells, a functional form of Miz-1 is also required for the proper expression of early B cell genes like E2A and EBF. These data suggest that Miz-1 represents a new regulatory element of early B- and T-cell differentiation required for the regulation of the IL-7/IL-7R/STAT5/Bcl-2 axis by monitoring SOCS1 for survival and by regulating the EBF/Pax-5/Rag-1/2 axis for the proper commitment and differentiation of the B-cell lineage. The regulation exerted by Miz-1 in B and T cells is mostly likely independent of its interacting partner c-Myc, and seems specifically linked to the BTB/POZ domain of Miz-1. Mice deficient for the BTB/POZ domain of Miz-1 have additionally a severe differentiation block at the pre-T cell “-selection” checkpoint. Miz-1 deficient pre-T cells are highly apoptotic and do show cell cycle defects. This concurs with enhanced expression of p53-target genes such as p21CIP1, Bax, PUMA and Noxa, most likely induced by the DNA double-strand breaks generated during the V(D)J recombination of the TCR. Only the co-expression of rearranged TCR and Bcl-2 fully rescued Miz-1-deficient cell numbers and enabled them to differentiate into TCR+ cells. These data suggest that Miz-1 is required for both the regulation of the p53 response and proper expression of the pre-TCR to support the proliferative burst of pre-T cells. In conclusion, the studies presented in this thesis revealed the so far unknown implication of Miz-1 in B- and T-cell development. More specifically, Miz-1 exerts early regulatory functions by monitoring the IL-7/IL-7R signaling in B and T cells. It regulates later stages of differentiation by controlling the EBF/Pax-5/Rag-1/2 in B cells and the TCR expression and the p53 response in T cells. These studies and the generated mice model (conditional knock-out and knock-in) will help characterize the implications of transcription factors that have been causally implicated in the altered genetic programming found in hematopoietic malignancies due to their capacities to regulate cell cycle. Ultimately the characterization of Miz-1 and c-Myc functions in B and T cells will help better understand the mechanisms responsible for the emergence of leukemia and lymphoma.
35

The BTB/POZ Transcription Factor Miz-1 Is Required To Regulate The Commitment, Survival And Differentiation Of Early B And T Cell Lineages

Saba, Ingrid 01 1900 (has links)
Les lymphocytes B et T sont issus de cellules progénitrices lymphoïdes de la moelle osseuse qui se différencient grâce à l’action de facteurs de transcription, cytokines et voies de signalisation, dont l’interleukine-7 (IL-7)/IL-7 récepteur (IL-7R). Le facteur de transcription c-Myc est exprimé par les cellules lymphoïdes et contrôle leur croissance et leur différenciation. Cette régulation transcriptionnelle peut être coordonnée par le complexe c-Myc/Myc-Interacting Zinc finger protein-1 (Miz-1). Le but de ce projet était de comprendre les mécanismes qui impliquent Miz-1 et le complexe c-Myc/Miz-1 dans le développement des lymphocytes B et T. Pour réaliser ce projet, des souris déficientes pour le domaine de transactivation de Miz-1 (Miz-1POZ) et des souris à allèles mutantes pour c-MycV394D, mutation qui empêche l’interaction avec Miz-1, ont été générées. La caractérisation des souris Miz 1POZ a démontré que l’inactivation de Miz-1 perturbe le développement des lymphocytes B et T aux stades précoces de leur différenciation qui dépend de l’IL-7. L’analyse de la cascade de signalisation IL-7/IL-7R a montré que ces cellules surexpriment la protéine inhibitrice SOCS1 qui empêche la phosphorylation de STAT5 et perturbe la régulation à la hausse de la protéine de survie Bcl-2. De plus, Miz-1 se lie directement au promoteur de SOCS1 et contrôle son activité. En plus de contrôler l’axe IL-7/IL-7R/STAT5/Bcl-2 spécifiquement aux stades précoces du développement afin d’assurer la survie des progéniteurs B et T, Miz-1 régule l’axe EBF/Pax-5/Rag-1/2 dans les cellules B afin de coordonner les signaux nécessaires pour la différenciation des cellules immatures. La caractérisation des souris c-MycV394D a montré, quant à elle, que les fonctions de Miz-1 dans les cellules B et T semblent indépendantes de c-Myc. Les cellules T des souris Miz-1POZ ont un défaut de différenciation additionnel au niveau de la -sélection, étape où les signaux initiés par le TCR remplacent ceux induits par IL-7 pour assurer la prolifération et la différenciation des thymocytes en stades plus matures. À cette étape du développement, une forme fonctionnelle de Miz-1 semble être requise pour contrôler le niveau d’activation de la voie p53, induite lors du processus de réarrangement V(D)J du TCR. L’expression de gènes pro-apoptotiques PUMA, NOXA, Bax et du régulateur de cycle cellulaire p21CIP1 est régulée à la hausse dans les cellules des souris Miz-1POZ. Ceci provoque un débalancement pro-apoptotique qui empêche la progression du cycle cellulaire des cellules TCR-positives. La survie des cellules peut être rétablie à ce stade de différenciation en assurant une coordination adéquate entre les signaux initiés par l’introduction d’un TCR transgénique et d’un transgène codant pour la protéine Bcl-2. En conclusion, ces études ont montré que Miz-1 intervient à deux niveaux du développement lymphoïde: l’un précoce en contrôlant la signalisation induite par l’IL-7 dans les cellules B et T, en plus de l’axe EBF/Pax-5/Rag-1/2 dans les cellules B; et l’autre tardif, en coordonnant les signaux de survie issus par le TCR et p53 dans les cellules T. Étant donné que les thymocytes et lymphocytes B immatures sont sujets à plusieurs rondes de prolifération, ces études serviront à mieux comprendre l’implication des régulateurs du cycle cellulaire comme c-Myc et Miz-1 dans la génération des signaux nécessaires à la différenciation non aberrante et à la survie des ces cellules. Enfin, les modèles expérimentaux, souris déficientes ou à allèles mutantes, utilisés pour ce travail permettront de mieux définir les bases moléculaires de la transformation maligne des lymphocytes B et T et de révéler les mécanismes conduisant au lymphome. / Signaling pathways control the differentiation and proliferation of blood cells, like B and T lymphocytes. They converge into regulating the activity of transcription factors that influence ultimately gene expression patterns. The transcription factor c-Myc is a central regulator of cellular proliferation and growth, and its deregulated expression has been demonstrated to be involved in many types of cancers, in particular lymphoma. Recent studies have shown that repression by c-Myc can be mediated by a complex formed with the BTB/POZ domain transcription factor Miz-1 (Myc Interacting Zinc finger protein-1). Given that both c-Myc and Miz-1 proteins are expressed in lymphoid precursors and since c-Myc has been shown to be important for B- and T-cell development, the aim of this thesis was to investigate the role of Miz-1 and the c-Myc/Miz-1 complex in regulating B and T cell survival, commitment and differentiation. To do so, mice expressing a non-functional Miz-1 protein lacking the BTB/POZ domain (Miz-1POZ) and knock-in mice expressing a mutant c-MycV394D allele that no longer interacts with Miz-1 were generated. B- and T-cell development requires the coordinated action of transcription factors and cytokines, in particular interleukin-7 (IL-7). The studies presented in this work demonstrated that mice deficient for the BTB/POZ domain of transcription factor Miz-1 almost entirely lack follicular B cells and T cells, since their progenitors fail to activate the JAK/STAT5 pathway and to up-regulate Bcl-2 upon IL-7 stimulation. Miz-1 exerts a dual role in the IL-7 receptor (IL-7R) pathway by directly repressing the JAK inhibitor SOCS1 and by activating Bcl-2 expression. In B cells, a functional form of Miz-1 is also required for the proper expression of early B cell genes like E2A and EBF. These data suggest that Miz-1 represents a new regulatory element of early B- and T-cell differentiation required for the regulation of the IL-7/IL-7R/STAT5/Bcl-2 axis by monitoring SOCS1 for survival and by regulating the EBF/Pax-5/Rag-1/2 axis for the proper commitment and differentiation of the B-cell lineage. The regulation exerted by Miz-1 in B and T cells is mostly likely independent of its interacting partner c-Myc, and seems specifically linked to the BTB/POZ domain of Miz-1. Mice deficient for the BTB/POZ domain of Miz-1 have additionally a severe differentiation block at the pre-T cell “-selection” checkpoint. Miz-1 deficient pre-T cells are highly apoptotic and do show cell cycle defects. This concurs with enhanced expression of p53-target genes such as p21CIP1, Bax, PUMA and Noxa, most likely induced by the DNA double-strand breaks generated during the V(D)J recombination of the TCR. Only the co-expression of rearranged TCR and Bcl-2 fully rescued Miz-1-deficient cell numbers and enabled them to differentiate into TCR+ cells. These data suggest that Miz-1 is required for both the regulation of the p53 response and proper expression of the pre-TCR to support the proliferative burst of pre-T cells. In conclusion, the studies presented in this thesis revealed the so far unknown implication of Miz-1 in B- and T-cell development. More specifically, Miz-1 exerts early regulatory functions by monitoring the IL-7/IL-7R signaling in B and T cells. It regulates later stages of differentiation by controlling the EBF/Pax-5/Rag-1/2 in B cells and the TCR expression and the p53 response in T cells. These studies and the generated mice model (conditional knock-out and knock-in) will help characterize the implications of transcription factors that have been causally implicated in the altered genetic programming found in hematopoietic malignancies due to their capacities to regulate cell cycle. Ultimately the characterization of Miz-1 and c-Myc functions in B and T cells will help better understand the mechanisms responsible for the emergence of leukemia and lymphoma.
36

Regulační úlohy proteinů PAG a CSK v FcɛRI signalizaci žírných buněk / Regulatory roles of PAG and CSK in FcɛRI signaling of mast cells

Potůčková, Lucie January 2017 (has links)
8 1 ABSTRACT (EN) This thesis is focused mainly on understanding mechanisms of regulatory roles of C-terminal Src kinase (CSK) and phosphoprotein associated with glycosphingolipid- enriched microdomains (PAG) in the high-affinity IgE receptor (FcɛRI)-mediated signaling of murine mast cells. FcɛRI activation is initiated by aggregation of the receptor by complexes of multivalent antigen with IgE, followed by activation and enhanced activities of protein tyrosine kinases, phosphatases, adaptor proteins and number of other signal transduction molecules. The signaling events result in mast cell degranulation and release of variety of proinflammatory mediators, responsible for initiation of allergy and other inflammatory diseases. Understanding the function of key regulatory molecules controlling FcεRI-mediated mast cell activation, degranulation, and cytokines production could have therapeutic impact. CSK is a major negative regulator of Src family tyrosine kinases (SFKs) that play a critical role in various immunoreceptor signaling events. However, its function in mast cell activation has not been completely understood. Because of its cytoplasmic localization, CSK was assumed to be brought to the vicinity of the plasma membrane- bound SFKs via binding to membrane-bound adaptors and PAG was a major candidate....
37

Digital and Analog STAT5 Signaling in Erythropoiesis: A Dissertation

Porpiglia, Ermelinda 16 August 2011 (has links)
Erythropoietin (Epo) modulates red blood cell production (erythropoiesis) by binding to its receptor and activating STAT5, a Signal Transducer and Activator of Transcription (STAT) protein implicated in both basal and stress erythropoiesis. Epo concentration in serum changes over three orders of magnitude, as it regulates basal erythropoiesis and its acceleration during hypoxic stress. However, it is not known how STAT5 translates the changes in Epo concentration into the required erythropoietic rates. We addressed this question by studying STAT5 phosphorylation, at the single cell level, in developing erythroblasts. We divided erythroid progenitors in tissue into several flow-cytometric subsets and found that each of them exhibited distinct modes of Stat5 activation, based on their developmental stage. STAT5 activation is bistable in mature erythroblasts, resulting in a binary (or digital), low-intensity STAT5 phosphorylation signal (p-Stat5). In early erythroblasts, and in response to stress levels of Epo, the low intensity bistable p-Stat5 signal is superseded by a high-intensity graded, or analog, signal. The gradual shift from high-intensity graded signaling in early erythroblasts to low intensity binary signaling in mature erythroblasts is due to a decline in STAT5 expression with maturation. We were able to convert mature, digital transducing erythroblasts into analog transducers simply by expressing high levels of exogenous STAT5. We found that EpoR-HM mice, expressing a mutant EpoR that lacks STAT5 docking sites, generate the binary, but not the analog, STAT5 signal. Unlike Stat5-null mice, which die perinatally, the EpoR-HM mice are viable but deficient in their response to stress, demonstrating that while binary STAT5 signaling is sufficient to support basal erythropoiesis, analog signaling is required for the stress response. Bistable systems contain a positive loop, which is important for flipping the switch between the two stable ‘on’ or ‘off’ states. We show that bistable activation is the result of an autocatalytic loop in which active STAT5 promotes further STAT5 activation. The isolated STAT5 N-terminal domain, which is not required for STAT5 phosphorylation, enhanced autocatalysis, converting a high intensity graded signal into a high intensity binary response. The N-terminal domain is known to participate in a radical conformational reorientation of STAT5 dimers inherent in STAT5 activation. We propose that the N-terminal domains of active STAT5 dimers facilitate the conformational reorientation of inactive dimers, in a prion-like autocatalytic interaction that underlies bistability and binary signaling. Together, bistable STAT5 activation, combined with a graded response allow erythropoietic rate to faithfully reflect a wide Epo concentration range, while preventing aberrant signaling.
38

Molecular Mechanisms of FLT3-ITD-Induced Leukemogenesis

Nabinger, Sarah Cassidy 07 August 2012 (has links)
Indiana University-Purdue University Indianapolis (IUPUI) / Internal tandem duplications in FMS-like receptor tyrosine kinase (FLT3-ITDs) are seen in approximately 25% of all acute myeloid leukemia (AML) patients. FLT3-ITDs induce FLT3 ligand (FL)-independent cellular hyperproliferation, promiscuous and aberrant activation of STAT5, and confer a poor prognosis in patients; however, the molecular mechanisms contributing to FLT3-ITD-induced malignancy remain largely unknown. The protein tyrosine phosphatase, Shp2, is important for normal hematopoiesis as well as hematopoietic stem cell (HSC) differentiation, engraftment, and self-renewal. Furthermore, FLT3-ITD- or constitutive active STAT5-expressing CD34+ cells demonstrate enhanced hematopoietic stem cell self-renewal. Together with the previous findings that Shp2 is critical for normal hematopoiesis, that dysregulated Shp2 function contributes to myeloid malignancies, and that Shp2 has been shown to interact with WT-FLT3 tyrosine 599, which is commonly duplicated in FLT3-ITDs, a positive role for Shp2 in FLT3-ITD-induced signaling and leukemogenesis is implied. I demonstrated that Shp2 is constitutively associated with the reported FLT3-ITDs, N51-FLT3 and N73-FLT3, compared to WT-FLT3; therefore, I hypothesized that increased Shp2 recruitment to N51-FLT3 or N73-FLT3 contributes to hyperproliferation and hyperactivation of STAT5. I also hypothesized that Shp2 cooperates with STAT5 to activate STAT5 transcriptional targets contributing to the up-regulation of pro-leukemic proteins. Finally, I hypothesized that reduction of Shp2 would result in diminished N51-FLT3-induced hyperproliferation and activation of STAT5 in vitro, and prevent FLT3-ITD-induced malignancy in vivo. I found that genetic disruption of Ptpn11, the gene encoding Shp2, or pharmacologic inhibition of Shp2 with the novel Shp2 inhibitor, II-B08, resulted in significantly reduced FLT3-ITD-induced hematopoietic cell hyperproliferation and STAT5 hyperphosphorylation. I also demonstrated a novel role of Shp2 in the nucleus of FLT3-ITD-expressing hematopoietic cells where Shp2 and STAT5 co-localized at the promoter region of STAT5-transcriptional target and pro-survival protein, Bcl-XL. Furthermore, using a Shp2flox/flox;Mx1Cre+ mouse model, I demonstrated that reduced Shp2 expression in hematopoietic cells resulted in an increased latency to and reduced severity of FLT3-ITD-induced malignancy. Collectively, these findings demonstrate that Shp2 plays an integral role in FLT3-ITD-induced malignancy and suggest that targeting Shp2 may be a future therapeutic option for treating FLT3-ITD-positive AML patients.
39

TRANSCRIPTIONAL CONTROL OF T HELPER CELL DIFFERENTIATION

Daniel Alejandro Canaria Gonzalez (15334258) 24 April 2023 (has links)
<p>  </p> <p>IL-9-producing CD4+ T helper (Th9) cells contribute to inflammatory responses during infection, anti-cancer responses and autoimmune disease. Thus, elucidating the signals that regulate their differentiation is critical for understanding the roles of Th9 cells in protective immunity and disease. Th9 cells differentiate in response to IL-4, TGF-β and IL-2, where IL-2 signaling through STAT5 is crucial for transactivating <em>Il9</em> locus. While the roles of IL-4 and TGF- β-mediated signaling are relatively well understood, how IL-2 signaling contributes to Th9 cell differentiation outside of directly inducing the <em>Il9</em> locus remains less clear. I found that human allergen-induced Th9 cells exhibited a strong signature of STAT5-mediated gene repression that was associated with inhibition of a Th17-like transcriptional signature. Likewise, blockade of IL-2/STAT5 signaling increased IL-17 and RORγt expression in murine Th9 cells <em>in vitro</em>. Interestingly, development of this Th17-like phenotype was independent of STAT3. While STAT3 was not required for IL-17 expression, it was required for their long-term persistence. These results suggest that IL-2/STAT5 signaling controls the balance between Th9 and Th17-like cell differentiation in vitro and during allergy. Additionally, I found that murine Th9 cells cultured in a low IL-2 environment had reduced IL-9 production and a diminished NF-kB-associated transcriptional signature, suggesting that IL-2 signaling is associated with NF-kB activation in Th9 cells. Interestingly, NF-kB activation via IL-1β stimulation enhanced Th9 differentiation under IL-2 limiting conditions and promoted their inflammatory potential in a mouse model of Lung inflammation. Mechanistically, we found that IL-2- limiting conditions enhanced IL-1β receptor expression and that IL-1β/NF-kB signaling increased the sensitivity to IL-2 and silenced the expression of the anti-Th9 transcription factor BCL6. Together, these findings indicate that IL-1β /NF-kB signaling can promote Th9 cell differentiation in IL-2-limiting conditions and that this pathway may be targeted to enhance Th9 differentiation and their inflammatory function.  Collectively, these data revealed two novel roles for the IL-2/STAT5 axis in Th9 cells.</p> <p>The Thymocyte associated High Mobility Group (HMG) box, known as TOX has been previously described to have paramount functions in the development of all the lineages of CD4+ T cells during thymic selection, during CD8+ T cell exhaustion and in Tfh cell differentiation and function. However, the role of TOX in non-Tfh CD4+ T cells in the periphery has not been addressed. In these studies, I found that CD4+ T cells express TOX in the steady state in secondary lymphoid organs like spleen, lymph nodes, and Peyer’s patches. Specifically, TOX was expressed remarkably in Tfh, Th1, Treg cells, and other non-Tfh unidentified Th cells, as well as Th2 cells in the lungs. Transcriptomics analyses using bulk RNA-seq revealed that TOX minimally alters s gene expression, however it revealed for the first time, that TOX induced genes associated with cell migration i.e., <em>Xcl1</em> <em>Ccl3</em>, <em>Ccl4</em> and also the inhibitory cytokine <em>Il10</em>. The induction of IL-10 and CCL3 was validated at the protein levels, and mechanistic studies revealed that the induction of these molecules required the transcription factor BATF, indicating for the first time a mechanism of TOX-mediated functions. Together, these data shed light in novel roles of TOX in CD4+ T cell function and opens the door for future functional and mechanistic studies that may be relevant during health and disease.</p>
40

Flow cytometric measurement of STAT5 phosphorylation in cytomegalovirus-stimulated T cells

Bitar, Michael, Boettcher, Marcus, Boldt, Andreas, Hauck, Fabian, Köhl, Ulrike, Liebert, Uwe G., Magg, Thomas, Schulz, Marian S., Sack, Ulrich 02 June 2023 (has links)
Cytomegalovirus (CMV)-specific T cells expand with CMV reactivation and are probably prerequisite for control and protection. Given the critical role STAT5A phosphorylation (pSTAT5A) in T cell proliferation, this study presents a simple and sensitive flow cytometric-based pSTAT5A assay to quickly identify CMV-specific T cell proliferation. We determined pSTAT5A in T cells treated with CMV-specific peptide mix (pp65 + IE1 peptides) from 20 healthy adult subjects and three immunodeficient patients with CARMIL-2 mutation. After stimulation, the percentage of pSTAT5A+ T cells in CMV-seropositive (CMV+) subjects significantly increased from 3.0% ± 1.9% (unstimulated) to 11.4% ± 5.9% (stimulated) for 24 h. After 7 days of stimulation, the percentage of expanded T cells amounted to 26% ± 17.2%. Conversely, the percentage of pSTAT5A+ T cells and T cell proliferation from CMV-seronegative (CMV−) subjects hardly changed (from 3.0% ± 1.3% to 3.7% ± 1.8% and from 4.3% ± 2.1% to 5.7% ± 1.7%, respectively). We analyzed the correlation between the percentage of pSTAT5A+ T cells versus (1) CMV-IgG concentrations versus (2) the percentage of expanded T cells and versus (3) the percentage of initial CMV-specific T cells. In immunodeficient patients with CARMIL-2 mutation, CMV-specific pSTAT5A and T cell proliferation were completely deficient. In conclusion, flow cytometric-based pSTAT5A assay represents an appropriate tool to quickly identify CMV-specific T cell proliferation and helps to understand dysfunctions in controlling other pathogens. Flow cytometric-based pSTAT5A assay may be a useful test in clinical practice and merits further validation in large studies.

Page generated in 0.4074 seconds