7 research outputs found

    Identification of Bromodomain-Containing Protein 4 (BRD4) as a Novel Marker and Epigenetic Target in Systemic Mastocytosis and Mast Cell Leukemia

    No full text
    Les mastocytes humains (MC) sont des cellules tissulaires d’origine hĂ©matopoĂŻĂ©tique impliquĂ©es dans une sĂ©rie de processus physiologiques et pathologiques. Les recherches sur les MC ont Ă©tĂ© entravĂ©es pendant longtemps en raison de l'accĂšs limitĂ© Ă  des populations pures de ces cellules. Nous avons Ă©tabli une nouvelle lignĂ©e humaine de MC, ROSAKIT WT, dont les propriĂ©tĂ©s sont similaires Ă  celles des MC primaires, constituant un nouvel outil pour la recherche sur les fonctions des MC humains, et permettant le criblage Ă  haut dĂ©bit de thĂ©rapies anti-allergiques. Les MC sont impliquĂ©s dans les mastocytoses, oĂč ils s’accumulent pathologiquement dans divers tissus. Bien que la plupart des cas de mastocytoses systĂ©miques (SM) sont chroniques et indolents, les patients atteints de SM avancĂ©e (SM agressive; ASM, et leucĂ©mie Ă  mastocytes; MCL) ont un mauvais pronostic, car la plupart des thĂ©rapies disponibles ne sont pas curatives. Afin de mieux comprendre la physiopathologie des formes avancĂ©es de SM et pour trouver de nouvelles approches pour le traitement, nous avons profitĂ© de la disponibilitĂ© des cellules ROSAKIT WT pour Ă©tablir un nouveau sous-clone, la lignĂ©e cellulaire ROSAKIT D816V, reprĂ©sentant un Ă©quivalent des cellules nĂ©oplasiques s’accumulant dans les SM. L'utilisation de cette lignĂ©e et de cellules des patients nous a permis d’identifier BRD4 comme une nouvelle cible thĂ©rapeutique dans les ASM et les MCL. Nous avons dĂ©montrĂ© que les MC nĂ©oplasiques de patients avec ASM expriment des quantitĂ©s substantielles de BRD4. Fait intĂ©ressant, nous avons aussi dĂ©montrĂ© que les lignĂ©es cellulaires HMC-1 et ROSAKIT D816V expriment aussi BRD4, et que leur prolifĂ©ration est inhibĂ©e par un shRNA BRD4-spĂ©cifique. En outre, nous avons montrĂ© que le mĂ©dicament JQ1, inhibiteur de BRD4, induit une inhibition de la croissance et une apoptose dose-dĂ©pendante dans les mĂȘmes cellules. De plus, nous avons dĂ©montrĂ© que JQ1 supprime Ă©galement la prolifĂ©ration des MC nĂ©oplasiques primaires de patients atteints d’ASM ou de MCL Ă  de faibles concentrations. Enfin, nous avons observĂ© que la midostaurine (PKC412) et l’acide rĂ©tinoĂŻque tout-trans (ATRA) coopĂšrent avec JQ1 pour induire des effets inhibiteurs synergiques sur l’inhibition de la survie des mĂȘmes cellules. En conclusion, nos rĂ©sultats reprĂ©sentent une avancĂ©e sur ce qui Ă©tait prĂ©cĂ©demment connu sur l’implication de BRD4 dans les mastocytoses et nous ont permis d'identifier cette protĂ©ine comme cible thĂ©rapeutique prometteuse dans le traitement des formes avancĂ©es 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 S

    Identification de BRD4 comme nouvelle cible thérapeutique dans le traitement des mastocytoses systémiques agressives (ASM) et des leucémies à mastocytes (MCL)

    No full text
    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 SMLes mastocytes humains (MC) sont des cellules tissulaires d’origine hĂ©matopoĂŻĂ©tique impliquĂ©es dans une sĂ©rie de processus physiologiques et pathologiques. Les recherches sur les MC ont Ă©tĂ© entravĂ©es pendant longtemps en raison de l'accĂšs limitĂ© Ă  des populations pures de ces cellules. Nous avons Ă©tabli une nouvelle lignĂ©e humaine de MC, ROSAKIT WT, dont les propriĂ©tĂ©s sont similaires Ă  celles des MC primaires, constituant un nouvel outil pour la recherche sur les fonctions des MC humains, et permettant le criblage Ă  haut dĂ©bit de thĂ©rapies anti-allergiques. Les MC sont impliquĂ©s dans les mastocytoses, oĂč ils s’accumulent pathologiquement dans divers tissus. Bien que la plupart des cas de mastocytoses systĂ©miques (SM) sont chroniques et indolents, les patients atteints de SM avancĂ©e (SM agressive; ASM, et leucĂ©mie Ă  mastocytes; MCL) ont un mauvais pronostic, car la plupart des thĂ©rapies disponibles ne sont pas curatives. Afin de mieux comprendre la physiopathologie des formes avancĂ©es de SM et pour trouver de nouvelles approches pour le traitement, nous avons profitĂ© de la disponibilitĂ© des cellules ROSAKIT WT pour Ă©tablir un nouveau sous-clone, la lignĂ©e cellulaire ROSAKIT D816V, reprĂ©sentant un Ă©quivalent des cellules nĂ©oplasiques s’accumulant dans les SM. L'utilisation de cette lignĂ©e et de cellules des patients nous a permis d’identifier BRD4 comme une nouvelle cible thĂ©rapeutique dans les ASM et les MCL. Nous avons dĂ©montrĂ© que les MC nĂ©oplasiques de patients avec ASM expriment des quantitĂ©s substantielles de BRD4. Fait intĂ©ressant, nous avons aussi dĂ©montrĂ© que les lignĂ©es cellulaires HMC-1 et ROSAKIT D816V expriment aussi BRD4, et que leur prolifĂ©ration est inhibĂ©e par un shRNA BRD4-spĂ©cifique. En outre, nous avons montrĂ© que le mĂ©dicament JQ1, inhibiteur de BRD4, induit une inhibition de la croissance et une apoptose dose-dĂ©pendante dans les mĂȘmes cellules. De plus, nous avons dĂ©montrĂ© que JQ1 supprime Ă©galement la prolifĂ©ration des MC nĂ©oplasiques primaires de patients atteints d’ASM ou de MCL Ă  de faibles concentrations. Enfin, nous avons observĂ© que la midostaurine (PKC412) et l’acide rĂ©tinoĂŻque tout-trans (ATRA) coopĂšrent avec JQ1 pour induire des effets inhibiteurs synergiques sur l’inhibition de la survie des mĂȘmes cellules. En conclusion, nos rĂ©sultats reprĂ©sentent une avancĂ©e sur ce qui Ă©tait prĂ©cĂ©demment connu sur l’implication de BRD4 dans les mastocytoses et nous ont permis d'identifier cette protĂ©ine comme cible thĂ©rapeutique prometteuse dans le traitement des formes avancĂ©es de SM

    Preclinical human models and emerging therapeutics for advanced systemic mastocytosis

    No full text
    Mastocytosis is a term used to denote a group of rare diseases characterized by an abnormal accumulation of neoplastic mast cells in various tissues and organs. In most patients with systemic mastocytosis, the neoplastic cells carry activating mutations in; KIT; Progress in mastocytosis research has long been hindered by the lack of suitable; in vitro; models, such as permanent human mast cell lines. In fact, only a few human mast cell lines are available to date: HMC-1, LAD1/2, LUVA, ROSA and MCPV-1. The HMC-1 and LAD1/2 cell lines were derived from patients with mast cell leukemia. By contrast, the more recently established LUVA, ROSA and MCPV-1 cell lines were derived from CD34; +; cells of non-mastocytosis donors. While some of these cell lines (LAD1/2, LUVA, ROSA; KIT WT; and MCPV-1) do not harbor; KIT; mutations, HMC-1 and ROSA; KIT D816V; cells exhibit activating; KIT; mutations found in mastocytosis and have thus been used to study disease pathogenesis. In addition, these cell lines are increasingly employed to validate new therapeutic targets and to screen for effects of new targeted drugs. Recently, the ROSA; KIT D816V; subclone has been successfully used to generate a unique; in vivo; model of advanced mastocytosis by injection into immunocompromised mice. Such a model may allow; in vivo; validation of data obtained; in vitro; with targeted drugs directed against mastocytosis. In this review, we discuss the major characteristics of all available human mast cell lines, with particular emphasis on the use of HMC-1 and ROSA; KIT D816V; cells in preclinical therapeutic research in mastocytosis

    A new human mast cell line expressing a functional IgE receptor converts to tumorigenic growth by KIT D816V transfection

    No full text
    International audienceIn systemic mastocytosis (SM), clinical problems arise from factor-independent proliferation of mast cells (MCs) and the increased release of mediators by MCs, but no human cell line model for studying MC activation in the context of SM is available. We have created a stable stem cell factor (SCF)-dependent human MC line, ROSA(KIT WT), expressing a fully functional immunoglobulin E (IgE) receptor. Transfection with KIT D816V converted ROSA(KIT WT) cells into an SCF-independent clone, ROSA(KIT D816V), which produced a mastocytosis-like disease in NSG mice. Although several signaling pathways were activated, ROSA(KIT D816V) did not exhibit an increased, but did exhibit a decreased responsiveness to IgE-dependent stimuli. Moreover, NSG mice bearing ROSA(KIT D816V)-derived tumors did not show mediator-related symptoms, and KIT D816V-positive MCs obtained from patients with SM did not show increased IgE-dependent histamine release or CD63 upregulation. Our data show that KIT D816V is a disease-propagating oncoprotein, but it does not activate MCs to release proinflammatory mediators, which may explain why mediator-related symptoms in SM occur preferentially in the context of a coexisting allergy. ROSA(KIT D816V) may provide a valuable tool for studying the pathogenesis of mastocytosis and should facilitate the development of novel drugs for treating SM patients
    corecore