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

SHP2/PTPN11 PROTEIN-TYROSINE PHOSPHATASE PROMOTES MAST CELL HOMEOSTASIS AND SYSTEMIC MASTOCYTOSIS

Sharma, NAMIT 25 June 2013 (has links)
KIT receptor (CD117) is a receptor tyrosine kinase crucial for homeostasis of mast cells (MCs) in tissues and recruitment to sites of inflammation and tumors in response to its ligand Stem cell factor (SCF). Gain of function mutations in KIT (e.g. D816V) are frequently observed in systemic mastocytosis and other cancer types. Src Homology 2 domain containing phosphatase-2 (SHP2 or PTPN11) is a protein tyrosine phosphatase that promotes cell proliferation, survival and motility in multiple pathways and cell types. To study SHP2 function in MCs, we generated novel MC-specific Shp2 knock-out (KO) mice (MC-shp2 KO). These mice had reduced numbers of MCs in skin and peritoneum, and defective contact hypersensitivity responses compared to control mice, consistent with SHP2 promoting MC homeostasis. Using an inducible SHP2 KO bone marrow-derived MC (BMMC) culture model, we found that SHP2 KO cells were prone to apoptosis and had no MC repopulating activity in vivo. Mechanistically, SHP2 enhanced ERK activation and downregulation of pro-apoptotic protein Bim. SHP2 KO BMMCs also had defects in chemotaxis towards SCF, due to impaired activation of a Lyn/Vav/Rac pathway in SHP2 KO BMMCs. This correlated with defects in cell spreading, and F-actin polymerization in response to SCF. Treatment of BMMCs with a SHP2 inhibitor (II-B08) also led to reduced chemotaxis, consistent with SHP2 phosphatase activity being required for KIT-induced chemotaxis. Lastly, we tested whether SHP2 regulates oncogenic KIT signaling using a P815 mouse mastocytoma model. Stable silencing of SHP2 in P815 cells led to reduced cell growth and survival in vitro, and less aggressive systemic mastocytosis development in syngeneic mice. Overall, these studies identify SHP2 as a key node in SCF/KIT and oncogenic KIT pathways, and as a potential therapeutic target in several human diseases. / Thesis (Ph.D, Biochemistry) -- Queen's University, 2013-06-25 12:03:57.818
2

Systemic Mastocytosis with associated CMML

Tawadros, Fady, Chakraborty, Kanishka 05 April 2018 (has links)
Systemic mastocytosis refers to a heterogeneous group of clinical disorders characterized by excessive mast cell accumulation in one or multiple organs. Mastocytosis is now considered as a separate disease category in the 2016 WHO classification of myeloid neoplasm and acute leukemia. It is no longer considered as a subgroup of meyloproliferate neoplasms. The clinical presentation of mastocytosis is heterogeneous ranging from skin-limited disease (cutaneous mastocytosis) to a more aggressive form with extra cutaneous presentation (systemic mastocytosis) with or without skin involvement. We are presenting a case of systemic mastocytosis that aroused in a patient who carried diagnosis of CMML for almost 2 years. The worsening B symptoms along with worsening splenomegaly were the driving factor for further investigations including Bone Marrow biopsy which revealed the diagnosis. A 74 year old Caucasian male with past oncology history of Chronic myelomoncytic leukemia diagnosed after persistant monocytosis on complete blood count . Patient presented with gradual onset of low grade fever , weight loss and night sweating , CT abdomen showed hepatosplenomegaly. core biopsy of the liver showed portal and lobular infiltrate consistent with involvement by mastocytes and extra medullary hematopoiesis. The infiltrate was positive for CD117, CD33, CD68, myeloperoxidase and CD163. Patient had bone marrow biopsy which showed increased CD117 positive cells consistent with involvement by systemic mastocytosis. The core biopsy showed multifocal nodules of spindle cells with fibrosis which was morphologically consistent with abnormal mast cells. Immunohistochemistry for CD117 was strongly positive in the spindle cell nodules and scattered polygonal cell nodules. KIT D816V mutation was detected. Patient met criteria for diagnosis of systemic mastocytosis with presence of previous diagnosis of CMML and classified as Systemic mastocytosis with an associated hematologic neoplasm (SM-AHN). Due to patient multiple comorbiditeis , he was not a candidate for Allo HCT. In an attempt to control his disease , patient was started on dose reduced Dacogen, but his functional status continued to delined and eventually dacogen was discontinued and patient was placed on best supportive car Conclusion Systemic mastocytosis is a rare entity with heterogeneous clinical presentation, highly variable disease course and consequently survival rates.Though recent advances in understanding genetic and molecular basis of disease, bone marrow transplantation remains the only treatment with possible curative potential in patients with advanced form of mastocytosis though carrying substantial mortality risk .Further understanding of Kit mutation might be able to offer a highly effective medication with durable response in a fashion similar to the success story of gleevac with CML treatment .
3

Nouvelles approches thérapeutiques au cours des mastocytoses systémiques avancées KIT D816V+ résistantes aux inhibiteurs de tyrosine kinases / New therapeutic approaches for KIT D816V+ advanced systemic mastocytosis resistant to tyrosine kinase inhibitors

Bibi, Siham 13 December 2016 (has links)
Les mastocytoses systémiques (SM) constituent un groupe hétérogène de maladies rares, caractérisées par l’accumulation anormale de mastocytes malins dans la moelle osseuse et dans d’autres organes extra-cutanés. La majorité des patients avec SM ont une mutation activatrice du gène KIT, le plus souvent la mutation KIT D816V, retrouvée chez plus de 90% de tous les patients. Cette mutation induit l’activation constitutive du récepteur KIT en déclenchant de façon aberrante une cascade de voies de signalisation, dont la voie PI3K/AKT et JAK/STAT5, aboutissant à l’inhibition de l’apoptose et à l’augmentation de la prolifération et de la survie des mastocytes malins. Cependant, l’efficacité des inhibiteurs de tyrosines kinases (ITKs) sur cette mutation est limitée à cause de la résistance et/ou de toxicité liée à un manque de spécificité. Il est donc nécessaire de trouver de nouvelles approches thérapeutiques afin de contourner cette résistance au cours des SM KIT D816V+ avancées. Nous avons utilisé une approche consistant à cibler de façon combinée des molécules activées en aval de KIT D816V, comme AKT et STAT5, par des inhibiteurs pharmacologiques. Ceci nous a permis d’identifier une combinaison synergique entre un inhibiteur d’AKT (GSK690693) et un inhibiteur de STAT5 (BP-1-102). Ces composés sont capables d’inhiber la prolifération des cellules KIT D816V+ seuls ou en combinaison, mais à de très fortes concentrations, malheureusement non utilisables en thérapeutique. Néanmoins, ces premiers résultats ont permis de valider AKT et STAT5 comme cibles potentielles dans le traitement des SM avancées. La seconde approche employée a été de cibler directement le récepteur KIT D816V par des inhibiteurs pharmacologiques. A l’issu d’un criblage, nous avons identifié trois composés - BLU2317, BLU2718 et DCC-2618 - capables d’inhiber sélectivement la phosphorylation de KIT D816V. Ces composés inhibent la prolifération des cellules ROSAKIT D816V et HMC-1.2, et induisent l’apoptose des cellules de façon dose-dépendante. Bien que les effets de ces trois composés soient similaires, DCC-2618 agit à des concentrations plus faibles par rapport aux composés BLU2317 et BLU2718. Afin d’apprécier l’efficacité in vivo de DCC-2618, nous avons d’abord établi un nouveau modèle de SM basé sur l’injection intraveineuse des cellules ROSAKIT D816V-Gluc exprimant la Gaussia luciferase (Gluc) dans des souris NSG. La présence de la Gluc sécrétée par les cellules ROSAKIT D816V-Gluc facilite la mise en évidence de prise de greffe et permet un contrôle précis de la progression de la maladie. Ce modèle reproduit, au bout de 4 semaines, chez toutes les souris greffées, une SM avancée similaire à celle retrouvée chez l’homme, avec atteinte de la moelle osseuse, du sang, de la rate et du foie, tandis que la dégradation de l’état général des souris n’est observée qu’à partir de 12 semaines. Ce nouveau modèle offre suffisamment de temps pour explorer la cinétique de la progression de la maladie et surtout pour effectuer des études pharmacologiques précliniques. L’évaluation de l’effet de DCC-2618 in vivo a été réalisée sur ce modèle. Etonnamment, DCC-2618 n’a pas été capable d’inhiber la progression de la maladie chez les souris traitées, bien qu’atteignant des concentrations élevées dans la moelle osseuse et le plasma des souris traitées. Néanmoins, DCC-2618 s’est montré capable d’inhiber la phosphorylation de KIT dans les cellules issues de la moelle osseuse des souris traitées. En revanche, contrairement aux effets observés in vitro, DCC-2618 a induit une surexpression de phospho-ERK1/2 dans les cellules malignes des souris greffées. Ceci suggère qu’ERK1/2 joue un rôle important dans la résistance au composé DCC-2618 et éventuellement à d’autres ITKs, indépendamment du récepteur KIT. ERK1/2 pourrait donc être une nouvelle cible thérapeutique d’intérêt dans le traitement des SM résistantes aux ITKs / Systemic mastocytosis (SM) is a heterogeneous group of rare diseases characterized by abnormal accumulation of malignant mast cells (MCs) in the bone marrow (BM) and other extra-cutaneous organs. The majority of SM patients have an activating mutation in the KIT gene, usually the D816V point mutation, which is found in more than 90% of all patients. This mutation induces constitutive activation of the KIT receptor by triggering a cascade of signaling pathways, including the PI3K/AKT and the JAK/STAT5 pathways, resulting in the inhibition of apoptosis and increased survival and proliferation of malignant mast cells. However, the efficacy of the tyrosine kinase inhibitors (TKIs) on this mutation is limited due to resistance and/or toxicity associated with a lack of specificity. It is therefore critical to find new therapeutic approaches to overcome this resistance to TKIs, particularly for advanced KIT D816V+ SM. In the present thesis, we have used an approach consisting in targeting molecules activated downstream of KIT D816V, such as AKT and STAT5, using pharmacological inhibitors in combination. This allowed us to identify a synergistic combination of an AKT inhibitor (GSK690693) and an inhibitor of STAT5 (BP-1-102). These compounds are able to inhibit proliferation of KIT D816V+ cells, alone or in combination, but at very high concentrations, unfortunately not useful therapeutically. Nevertheless, these initial results have validated STAT5 and AKT as potential targets for the treatment of advanced SM. The second approach used was to target directly the KIT D816V receptor by pharmacological inhibitors. After a large screening, we identified three compounds - BLU2317, BLU2718 and DCC-2618 - which selectively inhibit the phosphorylation of KIT D816V. These compounds inhibit the proliferation of ROSAKIT D816V and HMC-1.2 cells, and induce apoptosis of these cells in a dose-dependent manner. Although the effects of these three compounds are similar, the DCC-2618 compound acts at lower concentrations relative to BLU2317 and BLU2718 compounds. In order to assess the in vivo efficacy of DCC-2618, we first established a new model of SM based on intravenous injection of cells expressing Gaussia luciferase (Gluc), ROSAKIT D816V-Gluc cells, in NSG mice. The presence of the secreted Gluc in ROSAKIT D816V-Gluc cells facilitates the detection of engraftment and allows precise monitoring of disease progression. This model reproduced within four weeks, in all grafted mice, an advanced SM similar to the one found in humans, with neoplastic MCs infiltration in BM, blood, spleen and liver, while the terminal deterioration of the clinical condition of the mice was observed after 12 weeks. Thus, this new in vivo model allows modulating the aggressiveness of the disease by varying the number of injected cells. It provides sufficient time to explore the kinetics of disease progression and especially to conduct preclinical pharmacological studies. We then evaluated the effect of DCC-2618 compound in vivo on this model. Surprisingly, DCC-2618 was not able to inhibit disease progression in treated mice, although it reached high concentrations in the BM and the plasma of treated mice. Nevertheless, we showed that the compound was able to inhibit the phosphorylation of the KIT receptor in cells derived from the BM of treated mice. In addition, contrasting to the effects observed in vitro, DCC-2618 induced an over-expression of phospho-ERK1/2 in the malignant cells of transplanted mice. This suggests that ERK1/2 may play a critical role in the resistance to DCC-2618, and possibly to other TKIs, independently of the KIT receptor. ERK1/2 could thus be a new interesting therapeutic target in the treatment of advanced SM resistant to TKIs
4

Identification de BRD4 comme nouvelle cible thérapeutique dans le traitement des mastocytoses systémiques agressives (ASM) et des leucémies à mastocytes (MCL) / Identification of Bromodomain-Containing Protein 4 (BRD4) as a Novel Marker and Epigenetic Target in Systemic Mastocytosis and Mast Cell Leukemia

Wedeh, Ghaith 29 January 2016 (has links)
Les mastocytes humains (MC) sont des cellules tissulaires d’origine hématopoïétique impliquées dans une série de processus physiologiques et pathologiques. Les recherches sur les MC ont été entravées pendant longtemps en raison de l'accès limité à des populations pures de ces cellules. Nous avons établi une nouvelle lignée humaine de MC, ROSAKIT WT, dont les propriétés sont similaires à celles des MC primaires, constituant un nouvel outil pour la recherche sur les fonctions des MC humains, et permettant le criblage à haut débit de thérapies anti-allergiques. Les MC sont impliqués dans les mastocytoses, où ils s’accumulent pathologiquement dans divers tissus. Bien que la plupart des cas de mastocytoses systémiques (SM) sont chroniques et indolents, les patients atteints de SM avancée (SM agressive; ASM, et leucémie à mastocytes; MCL) ont un mauvais pronostic, car la plupart des thérapies disponibles ne sont pas curatives. Afin de mieux comprendre la physiopathologie des formes avancées de SM et pour trouver de nouvelles approches pour le traitement, nous avons profité de la disponibilité des cellules ROSAKIT WT pour établir un nouveau sous-clone, la lignée cellulaire ROSAKIT D816V, représentant un équivalent des cellules néoplasiques s’accumulant dans les SM. L'utilisation de cette lignée et de cellules des patients nous a permis d’identifier BRD4 comme une nouvelle cible thérapeutique dans les ASM et les MCL. Nous avons démontré que les MC néoplasiques de patients avec ASM expriment des quantités substantielles de BRD4. Fait intéressant, nous avons aussi démontré que les lignées cellulaires HMC-1 et ROSAKIT D816V expriment aussi BRD4, et que leur prolifération est inhibée par un shRNA BRD4-spécifique. En outre, nous avons montré que le médicament JQ1, inhibiteur de BRD4, induit une inhibition de la croissance et une apoptose dose-dépendante dans les mêmes cellules. De plus, nous avons démontré que JQ1 supprime également la prolifération des MC néoplasiques primaires de patients atteints d’ASM ou de MCL à de faibles concentrations. Enfin, nous avons observé que la midostaurine (PKC412) et l’acide rétinoïque tout-trans (ATRA) coopèrent avec JQ1 pour induire des effets inhibiteurs synergiques sur l’inhibition de la survie des mêmes cellules. En conclusion, nos résultats représentent une avancée sur ce qui était précédemment connu sur l’implication de BRD4 dans les mastocytoses et nous ont permis d'identifier cette protéine comme cible thérapeutique prometteuse dans le traitement des formes avancées de SM. / Human mast cells (MCs) are hematopoietic stem cell (HSC)-derived, tissue-resident, multifaceted cells involved in a myriad of physiological and pathological processes. Researches on MCs have been hampered for a long time, due to limited access to pure populations of these cells. We have established a new human MC line, ROSAKIT WT, whose properties are similar to those of primary HSC-derived MCs, providing a novel tool for research on human MC functions, and enabling the high-throughput screening of anti-allergic therapies. Among others, MCs are involved in a group of diseases termed mastocytosis, where they accumulate pathologically in various tissues. Although most cases of systemic mastocytosis (SM) are chronic with an indolent course, patients with advanced SM (aggressive SM; ASM, and mast cell leukemia; MCL) have a reduced life expectancy and a poor prognosis, since most of the therapies already available are not curative. In order to better understand the pathophysiology of advanced SM and to. find new approaches for treatment, we took advantage of the availability of the ROSAKIT WT cells to establish a new subclone, the ROSAKIT D816V cell line, representing a paradigm of the neoplastic cells accumulating in SMUsing these malignant cell line and patients’ cells, we identified the epigenetic reader bromodomain-containing protein-4 (BRD4) as a novel drug target in ASM and MCL. Indeed, we demonstrated that neoplastic MCs from ASM patients expressed substantial amounts of BRD4. Interestingly, we then demonstrated that HMC-1 and ROSAKIT D816V cell lines express BRD4, and that their proliferation is inhibited by a BRD4-specific shRNA. Moreover, we showed that the BRD4-targeting drug JQ1 induced a dose-dependent growth inhibition and apoptosis in the same cells. In addition, we demonstrated that JQ1 suppressed also the proliferation of primary neoplastic MCs of patients with ASM or MCL at low concentrations. Finally, we reported that midostaurin (PKC412) and all-trans retinoic acid (ATRA) cooperated with JQ1 in producing synergistic inhibitory effects on the survival of HMC-1 and ROSA cells. Together, our data represent a significant advance over what was previously known on the involvement of BRD4 in mastocytosis and identify this epigenetic reader bromodomain-containing protein as a promising drug target in advanced SM

Page generated in 0.0798 seconds