• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 2
  • 2
  • 1
  • Tagged with
  • 8
  • 4
  • 4
  • 3
  • 3
  • 3
  • 3
  • 3
  • 2
  • 2
  • 2
  • 2
  • 2
  • 1
  • 1
  • 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.
1

Evaluation of the actin architecture in dysplastic megakaryocytes expressing the NUP98-HOXD13 leukemic fusion gene

Okyere, Benjamin 30 August 2013 (has links)
Some myelodysplastic syndrome (MDS) patients present with macrothrombocytopenia due to impaired megakaryocyte (MK) differentiation. Transgenic mice that express the NUP98-HOXD13 (NHD13) fusion gene is a model for MDS and recapitulates the key features of MDS. The study investigated the hypothesis that expression of NHD13 disrupts actin architecture during MK differentiation leading to macrothrombocytopenia. To test the hypothesis, sternums were stained with hematoxylin and eosin, and evaluated by light microscopy to analyze MK morphology in vivo. NHD13 bone marrow (BM) contained many dysplastic MK. BM from wild type (WT) and NHD13 mice were also flushed, cultured in media supplemented with thrombopoietin only or with estrogen to induce proplatelet formation, and MK harvested after 5 days. Harvested MK and BM cores were processed and analyzed by transmission electron microscopy (TEM) to detail the ultrastructural features. TEM of MK revealed that NHD13 leads to formation of an irregular demarcation membrane system and fewer proplatelets. Cultured WT and NHD13 MK were also cytospun onto glass slides, labeled with fluorescent-tagged F-actin, α/β-tubulin and myosin IIa, and their cytoskeleton compared. Interestingly WT MK had actin either distributed evenly or predominantly in the periphery of the cytoplasm, NHD13 MK displayed only the former phenotype. Additionally, proplatelets lacked actin cytoplasmic extensions. The results from the present thesis demonstrate actin expression and architecture are impaired in dysplastic MK expressing the NHD13 leukemic fusion gene and leads to macrothromcytopenia. Understanding the molecular mechanisms of abnormal MK differentiation in MDS is important as many MDS patients die of hemorrhagic complications. / Master of Science
2

Mise au point d'un modèle de leucémie chez la Drosophile

Casgrain, Amélie January 2005 (has links)
Mémoire numérisé par la Direction des bibliothèques de l'Université de Montréal.
3

Elucidating the role of altered DNA damage response in Nup98-associated leukaemia

Nilles, Nadine 01 March 2018 (has links)
Acute myeloid leukaemia is a heterogeneous disease characterized by uncontrolled proliferation of neoplastic haematopoietic precursor cells, which leads to the disruption of normal haematopoiesis and bone marrow failure. Impaired haematopoiesis is often associated with balanced chromosomal translocations that involve the nucleoporin Nup98 fused to around 30 different partner genes, such as the homeobox genes HOXA9 and PMX1. Nup98-associated AML is characterized by poor prognosis and poor treatment outcome for the patients. The aim of the study was to elucidate the mechanisms underlying chemotherapy-resistance. Previous experiments showed that the expression of Nup98 fusion proteins leads to changes in nuclear organization. Based on these observations, we hypothesize that the expression of Nup98 fusion proteins affect DNA double-strand break (DSB) repair. Our work shows that the expression of Nup98-HoxA9 and Nup98-HHEX in U2OS cells does not induce any DSBs. Further, we examined the repair phenotype of exogenously induced DSBs. Experiments carried out using etoposide (ETO) or neocarzinostatin (NCS) revealed that Nup98 fusion proteins affect non-homologous end joining (NHEJ). The second major DSB repair pathway, homologous recombination (HR), remains unaffected by Nup98 fusion proteins. The repair phenotype showed that at most timepoints analyzed, cells expressing Nup98 fusion proteins present less DSBs that control cells. We further performed single cell gel electrophoresis assays, also called COMET assay. This assay determines the amount of broken DNA at the single cell level. COMET assays showed that cells expressing Nup98-HoxA9 get equally damaged as control cells. Taken together, these results show that Nup98-HoxA9 induces faster DNA repair by affecting NHEJ. Additional experiments, pointed toward a role of p53 in the effect of Nup98 fusion proteins on DSB repair. Monitoring the repair phenotype in a wild-type and p53 depletion background, revealed that the effect of Nup98-HoxA9 on NHEJ is partially p53 dependent. A further search for the potentially implicated factor in the accelerated NHEJ remained inconclusive so far. In conclusion, Nup98-HoxA9 induces accelerated NHEJ in a partially p53-dependent manner. / Option Biologie moléculaire du Doctorat en Sciences / info:eu-repo/semantics/nonPublished
4

Nucleoporin-Related Leukemia: Nucleoporin rearrangements and their impact on nucleocytoplasmic transport and the proteome

Rodrigues Mendes, Maria Adélia 08 July 2020 (has links) (PDF)
Chromosomal rearrangements of the nucleoporin genes NUP214 and NUP98 are recurrent in aggressive cases of acute myeloid and lymphoid leukemias. NUP214 and NUP98 are components of the nuclear pore complex, a giant multiprotein structure that mediates nucleocytoplasmic shuttling. The two nucleoporins are enriched in phenylalanine-glycine (FG) repeats, which form the NPC permeability barrier and are essential for the interaction with nuclear transport receptors. NUP214 and NUP98 exhibit high affinity for the nuclear export receptor chromosomal region maintenance 1 (CRM1), which, alone, mediates the nuclear export of thousands of proteins and ribonucleoproteins. In the first part of this project, we report that the leukemogenic fusion proteins SET-NUP214 and DEK-NUP214 affect nucleocytoplasmic transport by perturbing the localization of essential nuclear transport factors, including endogenous nucleoporins and CRM1 nuclear export complexes. We further demonstrate that the two fusion proteins are sensitive to CRM1 inhibition and that targeted inhibition of nuclear export is sufficient to reduce the cell viability and proliferation of patient-derived cell lines with SET-NUP214 and DEK-NUP214 rearrangements. In the second part of the project, we used proximity-dependent biotin identification (BioID) to study the landscape of the NUP98-HOXA9 and SET-NUP214 environments. Though distinct endogenous binding partners have been documented for NUP214 and NUP98 chimeras, their total interactome has not been fully disclosed. Our results suggest that both fusion proteins interact with major regulators of RNA processing, with translation-associated proteins, and that both chimeras perturb the transcriptional program of the tumor suppressor p53. We further purpose that the two fusion proteins affect distinct cellular processes. According to our results, NUP98-HOXA9 likely perturbs Wnt, MAPK and estrogen receptor signaling pathways, as well as the cytoskeleton, the latter likely due to its interaction with the nuclear export receptor CRM1. Conversely, SET-NUP214 appears to affect cellular metabolism, likely due to the interaction with mitochondrial proteins and metabolic regulators. Overall, this research project provided new data supporting that CRM1 might be a possible therapeutic target in NUP214-related leukemia and revealed new clues on the mechanistic actions of nucleoporin fusion proteins. Hence, our findings might be of particular relevance in the search of new druggable targets for the treatment of nucleoporin-related leukemia. / Doctorat en Sciences / info:eu-repo/semantics/nonPublished
5

Études des leucémies de l’enfant induites par les oncogènes de fusion NUP98::KDM5A et CBFA2T3::GLIS2

Roussy, Mathieu 12 1900 (has links)
Acute myeloid leukemia (AML) is a genetically heterogeneous disease and represents about 20% of pediatric leukemias. Survival rates vary depending on subtypes but are particularly unfavorable for acute megakaryoblastic leukemia (AMKL), a rare subtype of AML that usually affects children under 3 years old (≤ 30% survival for certain subtypes of AMKL). In pediatrics, genetic rearrangement leading to the expression of a chimeric fusion gene are present in many cases and are considered initiator events in the development of leukemia. In AMKL cases, more than 70% of them exhibit such rearrangement. Several of these chimeric transcripts, such as NUP98::KDM5A and CBFA2T3::GLIS2, occur in a higher proportion of cases. The analysis of the transcriptome from pediatric leukemic cases allowed us to identify new chimeric fusion transcripts in pediatric leukemias. Specifically, we discovered BPTF as a new fusion partner of NUP98 in the case of acute megakaryoblastic leukemia (AMKL), and the ACIN1::NUTM1 fusion in B-cell lymphoid leukemias. These studies have refined the molecular classification of these leukemias and provided tools for diagnosis and disease monitoring. The hypothesis of my thesis is that the NUP98::KDM5A and CBFA2T3::GLIS2 fusions are oncogenic and their expression in normal human hematopoietic and progenitor cells leads to transformation into acute megakaryoblastic leukemia in immunodeficient recipient mice, allowing for the generation of renewable xenograft models. My work has contributed to the generation of AMKL models with NUP98::KDM5A (N5A) and CBFA2T3::GLIS2 (CG2) fusions. To do this, we optimized a pipeline for transducing these chimeric genes in CD34+ cells isolated from cord blood, followed by transplantation into immunodeficient mice. These xenograft models phenocopy the leukemia of patients from a morphological, immunophenotypic, and transcriptomic standpoint. These synthetic AMKL models can be serially transplanted into mice and have a high frequency of leukemic stem cells. I also contributed to the development of a unique patient-derived xenograft (PDX) model derived from primary cells of a patient with an NUP98::BPTF genotype AMKL leukemia. These synthetic and PDX models then served as substrates for my experiments and those of several members of our laboratory. My research has allowed us to identify and characterize new biomarkers specific to NUP98- rearranged and CBFA2T3::GLIS2 positive AMKL. Taking advantage of the biomass generated by these AMKL leukemia models, we conducted transcriptomic and proteomic studies of the membrane surface. These results were compared to normal cells isolated from cord blood to identify several surface proteins specific to each leukemia genotype and shed light on new potential biomarkers. Furthermore, we confirmed the sensitivity of our AMKL models to JAK-STAT pathway inhibitors and performed synergy assays between JAK-STAT and the PI3K-AKT-mTOR pathway inhibitors. These experiments demonstrated the synergistic induction of apoptosis in our models upon the combine exposure to JAK-STAT and PI3K-AKT-mTOR pathway inhibitors. These works allowed us to identify potential therapeutic vulnerabilities of AMKL. Finally, since research on AMKL is affected by the limited number of patient samples, the human models and molecular data presented in this thesis constitute an invaluable resource to accelerate translational research for these high-risk leukemias. / La leucémie myéloïde aiguë (LMA) est une maladie hétérogène sur le plan génétique et représente environ 20% des leucémies pédiatriques. Les taux de survie varient selon les sous- types mais sont particulièrement défavorables pour les leucémies aiguës mégacaryoblastiques (AMKL), un sous-type rare de LMA touchant généralement les enfants de moins de 3 ans (≤ 30% de survie pour certains sous-types d’AMKL). En pédiatrie, les réarrangements génétiques entraînant l’expression d’un gène de fusion chimérique sont présentes dans un grand nombre de cas et sont considérées comme des événements initiateurs à l’origine de la leucémie. Chez les leucémies de type AMKL, c’est plus de 70% des cas qui présentent un tel réarrangement. Quelques-uns de ces transcrits chimériques, tels que NUP98::KDM5A et CBFA2T3::GLIS2, surviennent dans une plus grande proportion des cas. Dans le cadre de mes recherches, l’analyse du transcriptome de leucémies pédiatriques nous ont permis de mettre en évidence de nouveaux transcrits chimériques. Notamment, nous avons découvert BPTF comme étant un nouveau partenaire de fusion de NUP98 dans le cas d’une AMKL, ainsi que la fusion ACIN1::NUTM1 chez des leucémies lymphoïdes à cellules B. Ces travaux ont permis de raffiner la classification moléculaire de ces leucémies et propose de nouvelles approches pour le diagnostic et le suivi de la maladie. L’hypothèse de ma thèse est que les fusions NUP98::KDM5A et CBFA2T3::GLIS2 sont oncogéniques et leur expression chez des cellules souches hématopoïétiques et progénitrices humaines normales entraîne une transformation en leucémie aiguë mégacaryoblastique dans les souris receveuses immunodéficientes, permettant de générer des modèles de xénogreffe. Mes travaux ont contribué à la génération de modèles d’AMKL arborant les fusions NUP98::KDM5A ainsi que CBFA2T3::GLIS2. Pour ce faire, nous avons optimisé un processus de transduction de ces gènes chimériques chez des cellules CD34+ isolées de sang de cordon, suivi de transplantation chez la souris immunodéficiente. Ces modèles de xénogreffe récapitulent la leucémie des patients aux points de vue morphologique, immunophenotypique et transcriptomique. Ces modèles synthétiques d’AMKL peuvent être transplantés de manière sériée en souris et présentent une fréquence élevée de cellules souches leucémiques. De plus, nous avons aussi développé un modèle pdx unique (patient derived xenograft) dérivé des cellules primaires d’un patient atteint d’une leucémie AMKL présentant la fusions NUP98::BPTF. Ces modèles synthétiques et pdx ont ensuite servi de substrats à mes expériences ainsi que celles de plusieurs membres du laboratoire. Mes recherches ont permis d’identifier et de caractériser de nouveaux biomarqueurs spécifiques aux AMKL présentant un transcrit de NUP98 réarrangé et CBFA2T3::GLIS2. Tirant avantage de la biomasse générée par ces modèles de leucémie AMKL, nous avons fait des études transcriptomiques et protéomiques de la surface membranaire de nos modèles. Ces résultats furent comparés aux cellules normales isolées de sang de cordon afin d’identifier plusieurs protéines de surface spécifiques aux leucémies initiées par NUP98 réarrangé et CBFA2T3::GLIS2 afin de mettre en lumière de nouveaux biomarqueurs potentiels. De plus, nous avons aussi confirmé la sensibilité de nos modèles AMKL aux inhibiteurs de la voie JAK-STAT ainsi que démontré l’induction synergique de l’apoptose de nos modèles en présence des inhbitieurs combinés des voies JAK-STAT et PI3K-AKT-mTOR. Finalement, puisque la recherche sur les AMKL est ralentie par la quantité limitante d’échantillons de patient, les modèles humains et les données moléculaires présentés dans cette thèse constituent une ressource inestimable afin d’accélérer la recherche translationnelle pour ces leucémies à haut risque.
6

Étude de la fusion humaine NUP98-HOXA9 chez la drosophile

Gavory, Gwenaëlle 12 1900 (has links)
No description available.
7

Les oncogènes NUP98-PHF23 et NUP98-HOXD13 confèrent un potentiel aberrant d’auto-renouvellement aux progéniteurs thymiques

Tardif, Magalie 09 1900 (has links)
No description available.
8

REMANIEMENTS DU GENE NUP98 DANS LES HEMOPATHIES MALIGNES HUMAINES

Petit, Arnaud 09 September 2010 (has links) (PDF)
Les hémopathies malignes humaines sont associées à des remaniements chromosomiques récurrents. NUP98 est un gène à multipartenaires, remanié de manière récurrente dans les hémopathies malignes. Il code pour une nucléoporine qui participe au complexe de pore nucléaire. Deux nouveaux partenaires CCDC28A en 6q24 et HMGB3 en Xq28 ont été caractérisés chez deux patients présentant une hémopathie maligne avec remaniement de NUP98. Une fusion NUP98-NDS1 est décrite dans une translocation complexe. Une revue rétrospective des cas publiés indiquent que les remaniements du gène NUP98 sont rares. Ils surviennent essentiellement dans des hémopathies myéloïdes de novo, parfois dans les leucémies lymphoïdes T, jamais dans les leucémies lymphoïdes B. Les gènes partenaires comprennent des gènes à homéboites, des gènes remaniant la chromatine et des gènes codant pour des protéines cytoplasmiques. En proportion, les partenaires les plus fréquemment remaniés sont les gènes HOX. Les hémopathies sont toujours à caryotypes simples. Il existe une corrélation génotype/phénotype. La protéine CCDC28A, de fonction inconnue, est de localisation cytoplasmique. Une protéine analogue, CCDC28B, est impliquée en génétique humaine dans le syndrome de Bardet-Biedl. Le transcrit CCDC28A est exprimé précocement dans l'embryogénèse et dans tous les tissus humains. HMGB3 est un gène de l'hématopoïèse murine et des cellules souches embryonnaires. L'étude fonctionnelle des fusions NUP98-CCDC28A et NUP98-HMGB3, dans des progéniteurs hématopoïétiques murins, greffés chez la souris irradiée, a montré leurs forts pouvoirs transformants. Ces fusions induisent des leucémies aigues rapidement létales. De manière originale, ces fusions dérégulent peu les HOX, et ne dérégulent pas MEIS1, contrairement à plusieurs fusions NUP98, notamment NUP98-HOXA9, suggérant un mécanisme de transformation différent, situé en aval de la voie canonique Hoxa9-Meis1.

Page generated in 0.0213 seconds