Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Cells ; 13(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38727283

ABSTRACT

The unfolded protein response is an intricate system of sensor proteins in the endoplasmic reticulum (ER) that recognizes misfolded proteins and transmits information via transcription factors to either regain proteostasis or, depending on the severity, to induce apoptosis. The main transmembrane sensor is IRE1α, which contains cytoplasmic kinase and RNase domains relevant for its activation and the mRNA splicing of the transcription factor XBP1. Mast cell leukemia (MCL) is a severe form of systemic mastocytosis. The inhibition of IRE1α in the MCL cell line HMC-1.2 has anti-proliferative and pro-apoptotic effects, motivating us to elucidate the IRE1α interactors/regulators in HMC-1.2 cells. Therefore, the TurboID proximity labeling technique combined with MS analysis was applied. Gene Ontology and pathway enrichment analyses revealed that the majority of the enriched proteins are involved in vesicle-mediated transport, protein stabilization, and ubiquitin-dependent ER-associated protein degradation pathways. In particular, the AAA ATPase VCP and the oncoprotein MTDH as IRE1α-interacting proteins caught our interest for further analyses. The pharmacological inhibition of VCP activity resulted in the increased stability of IRE1α and MTDH as well as the activation of IRE1α. The interaction of VCP with both IRE1α and MTDH was dependent on ubiquitination. Moreover, MTDH stability was reduced in IRE1α-knockout cells. Hence, pharmacological manipulation of IRE1α-MTDH-VCP complex(es) might enable the treatment of MCL.


Subject(s)
Endoribonucleases , Leukemia, Mast-Cell , Protein Serine-Threonine Kinases , Humans , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Endoribonucleases/metabolism , Cell Line, Tumor , Leukemia, Mast-Cell/metabolism , Leukemia, Mast-Cell/pathology , Endoplasmic Reticulum-Associated Degradation , Valosin Containing Protein/metabolism , Valosin Containing Protein/genetics , Membrane Proteins/metabolism
2.
Int J Mol Sci ; 24(4)2023 Feb 09.
Article in English | MEDLINE | ID: mdl-36834926

ABSTRACT

Activating mutations in KIT (CD117) have been associated with several diseases, including gastrointestinal stromal tumors and mastocytosis. Rapidly progressing pathologies or drug resistance highlight the need for alternative treatment strategies. Previously, we reported that the adaptor molecule SH3 binding protein 2 (SH3BP2 or 3BP2) regulates KIT expression at the transcriptional level and microphthalmia-associated transcription factor (MITF) expression at the post-transcriptional level in human mast cells and gastrointestinal stromal tumor (GIST) cell lines. Lately, we have found that the SH3BP2 pathway regulates MITF through miR-1246 and miR-5100 in GIST. In this study, miR-1246 and miR-5100 were validated by qPCR in the SH3BP2-silenced human mast cell leukemia cell line (HMC-1). MiRNA overexpression reduces MITF and MITF-dependent target expression in HMC-1. The same pattern was observed after MITF silencing. In addition, MITF inhibitor ML329 treatment reduces MITF expression and affects the viability and cell cycle progression in HMC-1. We also examine whether MITF downregulation affected IgE-dependent mast cell degranulation. MiRNA overexpression, MITF silencing, and ML329 treatment reduced IgE-dependent degranulation in LAD2- and CD34+-derived mast cells. These findings suggest MITF may be a potential therapeutic target for allergic reactions and deregulated KIT mast-cell-mediated disorders.


Subject(s)
Leukemia, Mast-Cell , MicroRNAs , Microphthalmia-Associated Transcription Factor , Humans , Cell Death/genetics , Down-Regulation , Gastrointestinal Stromal Tumors/metabolism , Gastrointestinal Stromal Tumors/pathology , Immunoglobulin E/metabolism , Leukemia, Mast-Cell/metabolism , Mast Cells/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , MicroRNAs/genetics
3.
Hematol Oncol ; 41(3): 520-534, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36383121

ABSTRACT

Prevention of fatal side effects during cancer therapy of cancer patients with high-dosed pharmacological inhibitors is to date a major challenge. Moreover, the development of drug resistance poses severe problems for the treatment of patients with leukemia or solid tumors. Particularly drug-mediated dimerization of RAF kinases can be the cause of acquired resistance, also called "paradoxical activation." In the present work we re-analyzed the effects of different tyrosine kinase inhibitors (TKIs) on the proliferation, metabolic activity, and survival of the Imatinib-resistant, KIT V560G, D816V-expressing human mast cell (MC) leukemia (MCL) cell line HMC-1.2. We observed that low concentrations of the TKIs Nilotinib and Ponatinib resulted in enhanced proliferation, suggesting paradoxical activation of the MAPK pathway. Indeed, these TKIs caused BRAF-CRAF dimerization, resulting in ERK1/2 activation. The combination of Ponatinib with the MEK inhibitor Trametinib, at nanomolar concentrations, effectively suppressed HMC-1.2 proliferation, metabolic activity, and induced apoptotic cell death. Effectiveness of this drug combination was recapitulated in the human KIT D816V MC line ROSAKIT D816V and in KIT D816V hematopoietic progenitors obtained from patient-derived induced pluripotent stem cells (iPS cells) and systemic mastocytosis patient samples. In conclusion, mutated KIT-driven Imatinib resistance and possible TKI-induced paradoxical activation can be efficiently overcome by a low concentration Ponatinib and Trametinib co-treatment, potentially reducing the negative side effects associated with MCL therapy.


Subject(s)
Leukemia, Mast-Cell , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Leukemia, Mast-Cell/metabolism , Leukemia, Mast-Cell/pathology , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/pharmacology , Mast Cells/metabolism , Mast Cells/pathology , Proto-Oncogene Proteins c-kit/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Mutation
4.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 44(6): 1117-1122, 2022 Dec.
Article in Chinese | MEDLINE | ID: mdl-36621796

ABSTRACT

Mast cell leukemia(MCL)is an extremely rare type of leukemia with high heterogeneity in clinical practice.MCL needs to be diagnosed by means of bone marrow routine and pathology,flow immunophenotyping,and cytogenetics and molecular biological testing.This article retrospectively studied the clinical data including the clinical features,diagnosis,treatment,and prognosis of two patients with MCL,aiming to improve the understanding of MCL and provide a new reference for the clinical diagnosis,treatment,and basic medical research of this disease.


Subject(s)
Leukemia, Mast-Cell , Humans , Leukemia, Mast-Cell/diagnosis , Leukemia, Mast-Cell/metabolism , Leukemia, Mast-Cell/pathology , Retrospective Studies , Bone Marrow/pathology
5.
Clin Cancer Res ; 24(24): 6396-6407, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30171047

ABSTRACT

PURPOSE: STA-1474, prodrug of the heat shock protein 90 inhibitor (HSP90i) ganetespib, previously demonstrated activity in canine preclinical models of cancer; interestingly, prolonged infusions were associated with improved biologic activity. The purpose of this study was to identify the ideal treatment schedule for HSP90i in preclinical models of KIT-driven malignancies and in dogs with spontaneous mast cell tumors (MCT), where KIT is a known driver. EXPERIMENTAL DESIGN: In vitro and murine xenograft experiments and clinical studies in dogs with MCTs were used to define the effects of HSP90i-dosing regimen on client protein downregulation and antitumor activity. RESULTS: Continuous HSP90 inhibition led to durable destabilization of client proteins in vitro; however, transient exposure required >10× drug for comparable effects. In vivo, KIT was rapidly degraded following a single dose of HSP90i but returned to baseline levels within a day. HSP90 levels increased and stabilized 16 hours after HSP90i and were not elevated following a subsequent near-term exposure, providing a functional pool of chaperone to stabilize proteins and a means for greater therapeutic activity upon HSP90i reexposure. HSP90i administered on days 1 and 2 (D1/D2) demonstrated increased biologic activity compared with D1 treatment in KIT or EGFR-driven murine tumor models. In a trial of dogs with MCT, D1/D2 dosing of HSP90i was associated with sustained KIT downregulation, 50% objective response rate and 100% clinical benefit rate compared with D1 and D1/D4 schedules. CONCLUSIONS: These data provide further evidence that prolonged HSP90i exposure improves biologic activity through sustained downregulation of client proteins.


Subject(s)
Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Leukemia, Mast-Cell/etiology , Leukemia, Mast-Cell/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Dogs , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Leukemia, Mast-Cell/drug therapy , Leukemia, Mast-Cell/pathology , Mice , Oncogenes , Proteolysis , Proto-Oncogene Proteins c-kit/genetics , Treatment Outcome , Triazoles/pharmacology , Xenograft Model Antitumor Assays
7.
Leukemia ; 32(4): 1016-1022, 2018 04.
Article in English | MEDLINE | ID: mdl-29249817

ABSTRACT

Systemic mastocytosis (SM) is a mast cell (MC) neoplasm with complex pathology and a variable clinical course. In aggressive SM (ASM) and MC leukemia (MCL), responses to conventional drugs are poor and the prognosis is dismal. R763 is a multi-kinase inhibitor that blocks the activity of Aurora-kinase-A/B, ABL1, AKT and FLT3. We examined the effects of R763 on proliferation and survival of neoplastic MC. R763 produced dose-dependent inhibition of proliferation in the human MC lines HMC-1.1 (IC50 5-50 nM), HMC-1.2 (IC50 1-10 nM), ROSAKIT WT (IC50 1-10 nM), ROSAKIT D816V (IC50 50-500 nM) and MCPV-1.1 (IC50 100-1000 nM). Moreover, R763 induced growth inhibition in primary neoplastic MC in patients with ASM and MCL. Growth-inhibitory effects of R763 were accompanied by signs of apoptosis and a G2/M cell cycle arrest. R763 also inhibited phosphorylation of KIT, BTK, AKT and STAT5 in neoplastic MC. The most sensitive target appeared to be STAT5. In fact, tyrosine phosphorylation of STAT5 was inhibited by R763 at 10 nM. At this low concentration, R763 produced synergistic growth-inhibitory effects on neoplastic MC when combined with midostaurin or dasatinib. Together, R763 is a novel promising multi-kinase inhibitor that blocks STAT5 activation and thereby overrides drug-resistance in neoplastic MC.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Mast Cells/drug effects , Phosphorylation/drug effects , STAT5 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Dasatinib/pharmacology , Dogs , Drug Synergism , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Leukemia, Mast-Cell/drug therapy , Leukemia, Mast-Cell/metabolism , Male , Mast Cells/metabolism , Mastocytosis, Systemic/drug therapy , Mastocytosis, Systemic/metabolism , Middle Aged , Norbornanes/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/pharmacology , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Young Adult
8.
Naunyn Schmiedebergs Arch Pharmacol ; 389(7): 671-94, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27132234

ABSTRACT

Mast cell activation disease (MCAD) is a term referring to a heterogeneous group of disorders characterized by aberrant release of variable subsets of mast cell (MC) mediators together with accumulation of either morphologically altered and immunohistochemically identifiable mutated MCs due to MC proliferation (systemic mastocytosis [SM] and MC leukemia [MCL]) or morphologically ordinary MCs due to decreased apoptosis (MC activation syndrome [MCAS] and well-differentiated SM). Clinical signs and symptoms in MCAD vary depending on disease subtype and result from excessive mediator release by MCs and, in aggressive forms, from organ failure related to MC infiltration. In most cases, treatment of MCAD is directed primarily at controlling the symptoms associated with MC mediator release. In advanced forms, such as aggressive SM and MCL, agents targeting MC proliferation such as kinase inhibitors may be provided. Targeted therapies aimed at blocking mutant protein variants and/or downstream signaling pathways are currently being developed. Other targets, such as specific surface antigens expressed on neoplastic MCs, might be considered for the development of future therapies. Since clinicians are often underprepared to evaluate, diagnose, and effectively treat this clinically heterogeneous disease, we seek to familiarize clinicians with MCAD and review current and future treatment approaches.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Histamine Antagonists/therapeutic use , Immunosuppressive Agents/therapeutic use , Leukemia, Mast-Cell/drug therapy , Mast Cells/drug effects , Mastocytosis, Systemic/drug therapy , Animals , Antineoplastic Agents/adverse effects , Apoptosis , Cell Degranulation/drug effects , Histamine Antagonists/adverse effects , Humans , Immunosuppressive Agents/adverse effects , Leukemia, Mast-Cell/immunology , Leukemia, Mast-Cell/metabolism , Leukemia, Mast-Cell/pathology , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/pathology , Mastocytosis, Systemic/immunology , Mastocytosis, Systemic/metabolism , Mastocytosis, Systemic/pathology , Molecular Targeted Therapy , Treatment Outcome
9.
Leukemia ; 30(4): 914-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26678095

ABSTRACT

CD123 is the α-subunit of the interleukin-3 receptor; it represents a potential therapeutic target in systemic mastocytosis (SM) given its absent expression on normal/reactive mast cells (MCs) and aberrant expression on neoplastic MCs. We studied 58 SM patients to define CD123 expression patterns by immunohistochemistry and its clinical significance. Two hematopathologists independently scored bone marrow slides using predefined histologic parameters. In all, 23 patients had indolent SM (ISM), 10 aggressive SM (ASM), 23 SM with associated hematological neoplasm (SM-AHN) and 2 had mast cell leukemia (MCL). MC_CD123 expression was demonstrable in 37 (64%) cases; expression rates were 100%, 61%, 57% and 0% in ASM, ISM, SM-AHN and MCL, respectively (P=0.02). Focal proliferation of plasmacytoid dendritic cells (PDCs) around MC aggregates, suggesting a tumor-promoting role for PDCs, was noted in 44 (76%) cases, and was significantly higher in CD123-positive versus -negative cases (87% versus 50%, P=0.005). CD123 expression and its staining intensity had prognostic value in SM-chronic myelomonocytic leukemia and nonindolent SM patients, respectively. These observations suggest that targeting CD123 in SM may have direct (via MCs) and indirect (via PDCs) antitumor effects and clinical trials to that effect require laboratory correlative studies to address the observed target expression heterogeneity.


Subject(s)
Biomarkers, Tumor/metabolism , Hematologic Neoplasms/metabolism , Interleukin-3 Receptor alpha Subunit/metabolism , Leukemia, Mast-Cell/metabolism , Mastocytosis, Systemic/metabolism , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Hematologic Neoplasms/pathology , Humans , Immunoenzyme Techniques , Leukemia, Mast-Cell/pathology , Male , Mastocytosis, Systemic/pathology , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies
10.
Leukemia ; 29(6): 1320-30, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25655194

ABSTRACT

Mast cells (MCs) are critical components of the innate immune system and important for host defense, allergy, autoimmunity, tissue regeneration and tumor progression. Dysregulated MC development leads to systemic mastocytosis (SM), a clinically variable but often devastating family of hematologic disorders. Here we report that induced expression of Lin28, a heterochronic gene and pluripotency factor implicated in driving a fetal hematopoietic program, caused MC accumulation in adult mice in target organs such as the skin and peritoneal cavity. In vitro assays revealed a skewing of myeloid commitment in LIN28B-expressing hematopoietic progenitors, with increased levels of LIN28B in common myeloid and basophil-MC progenitors altering gene expression patterns to favor cell fate choices that enhanced MC specification. In addition, LIN28B-induced MCs appeared phenotypically and functionally immature, and in vitro assays suggested a slowing of MC terminal differentiation in the context of LIN28B upregulation. Finally, interrogation of human MC leukemia samples revealed upregulation of LIN28B in abnormal MCs from patients with SM. This work identifies Lin28 as a novel regulator of innate immune function and a new protein of interest in MC disease.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/physiology , Leukemia, Mast-Cell/pathology , Mast Cells/cytology , Mastocytosis, Systemic/pathology , Myeloid Cells/cytology , RNA-Binding Proteins/metabolism , Aged , Aged, 80 and over , Animals , Blotting, Western , Bone Marrow Transplantation , Cells, Cultured , Female , Flow Cytometry , Hematopoiesis/physiology , Humans , Leukemia, Mast-Cell/metabolism , Leukemia, Mast-Cell/therapy , Male , Mast Cells/metabolism , Mastocytosis, Systemic/metabolism , Mastocytosis, Systemic/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myeloid Cells/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
11.
Leuk Res ; 39(1): 1-5, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25443885

ABSTRACT

Mast cell leukemia (MCL) is a rare form of systemic mastocytosis characterized by leukemic expansion of mostly immature mast cells, organ damage, drug-resistance, and a poor prognosis. Even when treated with chemotherapy, most patients have a life-expectancy of less than one year. However, there are rare patients with MCL in whom the condition is less aggressive and does not cause organ damage within a short time. In these patients, mast cells exhibit a more mature morphology when compared to acute MCL. A recently proposed classification suggests that these cases are referred to as chronic MCL. In the present article, we discuss clinical, histopathological and morphological aspects of acute and chronic MCL.


Subject(s)
Drug Resistance, Neoplasm , Leukemia, Mast-Cell , Acute Disease , Chronic Disease , Humans , Leukemia, Mast-Cell/classification , Leukemia, Mast-Cell/drug therapy , Leukemia, Mast-Cell/metabolism , Leukemia, Mast-Cell/pathology
12.
Cell Mol Life Sci ; 69(16): 2751-64, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22438059

ABSTRACT

Cell adhesion molecule 1 (CADM1), expressed by human lung mast cells (HLMCs), mediates their adhesion to airway smooth muscle (ASM), and contributes to ASM-dependent HLMC proliferation and survival. CADM1 is expressed in alternatively spliced isoforms, but those present in HLMCs and their function are not known. We cloned three functional and one cryptic non-functional isoform with alternative splicing between exons 7/11 and 1/2, respectively, from HLMCs and human MC lines (HMC-1 and LAD2). Differentiated HLMCs and LAD2 cells expressed the functional isoform SP4 containing exons 7/8/11 (~80% of clones), as well as SP1 (exons 7/8/9/11) and a novel SP6 (exons 7/8/9/10/11). In contrast, immature HMC-1 cells expressed only functional SP4. SP4 overexpression in HMC-1 cells and HLMCs augmented homotypic adhesion to a greater extent than SP1 in various conditions. In contrast, CADM1 downregulation abolished homotypic adhesion, indicating that CADM1 is the sole receptor mediating mast cell aggregation. CADM1-mediated adhesion was enhanced by the presence of cell survival factors. SP1 overexpression in HMC-1 cells compromised survival compared to SP4 overexpression or control. CADM1 downregulation resulted in reduced viability and decreased expression of the pro-survival protein Mcl-1(L), but not Blc-2 or Bcl-X(L), and increased caspase-3/7 activity in both HMC-1 cells and HLMCs. This coincided with decreased basal Kit levels in HLMCs. In summary, human MCs express multiple CADM1 isoforms which exhibit differential regulation of survival and homotypic adhesion. The most highly expressed SP4 isoform is likely to contribute to MC aggregation and longevity in mastocytosis, and augment the pathophysiology of allergic diseases.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Adhesion , Immunoglobulins/metabolism , Leukemia, Mast-Cell/pathology , Lung/cytology , Mast Cells/cytology , Mast-Cell Sarcoma/pathology , Blotting, Western , Cell Adhesion Molecule-1 , Cell Aggregation , Cell Survival , Cells, Cultured , Humans , Leukemia, Mast-Cell/metabolism , Lung/metabolism , Mast Cells/metabolism , Mast-Cell Sarcoma/metabolism , Protein Isoforms , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
13.
Leuk Lymphoma ; 53(9): 1795-803, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22300345

ABSTRACT

The aim of the present study was to determine whether dehydroleucodine, xanthatin and 3-benzyloxymethyl-5H-furan-2-one inhibit the activation of human leukemic LAD2 mast cells induced by compound 48/80 or the calcium ionophore A23187. LAD2 cells were preincubated in the presence of test drugs and then challenged with the secretagogues. This study provides the first evidence in favor of the view that dehydroleucodine and xanthatin inhibit the degranulation of LAD2 cells, thus acting as human mast cell stabilizers. These molecules could be effective in the treatment of human diseases associated with inappropriate mast cell activation.


Subject(s)
Furans/pharmacology , Lactones/pharmacology , Mast Cells/metabolism , Sesquiterpenes/pharmacology , Calcimycin/pharmacology , Calcium Ionophores/pharmacology , Cell Degranulation/drug effects , Cell Line, Tumor , Humans , Kinetics , Leukemia, Mast-Cell/metabolism , Leukemia, Mast-Cell/pathology , Mast Cells/physiology , Mast Cells/ultrastructure , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , beta-N-Acetylhexosaminidases/metabolism , p-Methoxy-N-methylphenethylamine/pharmacology
14.
Br J Haematol ; 156(5): 601-11, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22150124

ABSTRACT

Mantle cell lymphoma (MCL) is an aggressive neoplasm with a short survival. Cases with leukaemic MCL and splenomegaly without adenopathies (non-nodal MCL) may have a more indolent course. To gain insights into the biological features underlying this presentation, we investigated the gene expression profile (GEP) and the IGHV mutational status in a cohort of leukaemic MCL cases. Comparison of MCL with other lymphoproliferative disorders (i.e. splenic marginal zone lymphoma, follicular lymphoma, chronic lymphocytic leukaemia) revealed a MCL signature enriched for the following gene categories: mitochondrion, oxidoreductase activity, response to stress, to DNA damage and TP53-pathway. Furthermore, GEP analysis revealed that non-nodal MCL cases were characterized by the down-modulation of the following gene categories: cell projection, actin cytoskeleton organization, cell adhesion (ITGAE, CELSR1, PCDH9) and tumour invasion/progression (PGF, ST14, ETS1, OCIAD1, EZR). Many down-modulated genes were related to the TP53-pathway and to DNA damage response. IGHV status proved unmutated in all nodal and mutated in all non-nodal MCL. Non-nodal leukaemic MCLs display a peculiar clinical presentation, with distinctive biological features, such as mutated IGHV and a transcriptional profile lacking tumour invasion properties, that might contribute to the absence of nodal involvement and to the less aggressive clinical course.


Subject(s)
Actin Cytoskeleton/genetics , Genes, Immunoglobulin Heavy Chain/genetics , Genes, p53/genetics , Leukemia, Mast-Cell/genetics , Lymph Nodes/pathology , ADP-ribosyl Cyclase 1/metabolism , Aged , Aged, 80 and over , Cell Adhesion/genetics , DNA, Neoplasm/genetics , Down-Regulation/genetics , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Humans , Leukemia, Mast-Cell/metabolism , Leukemia, Mast-Cell/pathology , Male , Membrane Glycoproteins/metabolism , Middle Aged , Mutation , Neoplasm Invasiveness
17.
J Immunol ; 181(7): 5024-34, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18802106

ABSTRACT

Mast cells are granular immunocytes that reside in the body's barrier tissues. These cells orchestrate inflammatory responses. Proinflammatory mediators are stored in granular structures within the mast cell cytosol. Control of mast cell granule exocytosis is a major therapeutic goal for allergic and inflammatory diseases. However, the proteins that control granule biogenesis and abundance in mast cells have not been elucidated. In neuroendocrine cells, whose dense core granules are strikingly similar to mast cell granules, granin proteins regulate granulogenesis. Our studies suggest that the Secretogranin III (SgIII) protein is involved in secretory granule biogenesis in mast cells. SgIII is abundant in mast cells, and is organized into vesicular structures. Our results show that over-expression of SgIII in mast cells is sufficient to cause an expansion of a granular compartment in these cells. These novel granules store inflammatory mediators that are released in response to physiological stimuli, indicating that they function as bona fide secretory vesicles. In mast cells, as in neuroendocrine cells, we show that SgIII is complexed with Chromogranin A (CgA). CgA is granulogenic when complexed with SgIII. Our data show that a novel non-granulogenic truncation mutant of SgIII (1-210) lacks the ability to interact with CgA. Thus, in mast cells, a CgA-SgIII complex may play a key role in secretory granule biogenesis. SgIII function in mast cells is unlikely to be limited to its partnership with CgA, as our interaction trap analysis suggests that SgIII has multiple binding partners, including the mast cell ion channel TRPA1.


Subject(s)
Cell Communication/immunology , Chromogranin A/metabolism , Chromogranins/metabolism , Mast Cells/metabolism , Secretory Vesicles/metabolism , Animals , Cell Communication/genetics , Cell Line , Cell Line, Tumor , Chromogranin A/biosynthesis , Chromogranin A/genetics , Chromogranin A/physiology , Chromogranins/biosynthesis , Chromogranins/genetics , Chromogranins/physiology , Humans , Leukemia, Mast-Cell/genetics , Leukemia, Mast-Cell/immunology , Leukemia, Mast-Cell/metabolism , Mast Cells/immunology , Mast Cells/pathology , Mastocytoma/genetics , Mastocytoma/immunology , Mastocytoma/metabolism , Mice , PC12 Cells , Rats , Secretory Vesicles/genetics , Secretory Vesicles/immunology , Secretory Vesicles/pathology , Sequence Deletion
18.
Leuk Res ; 32(5): 761-70, 2008 May.
Article in English | MEDLINE | ID: mdl-17949810

ABSTRACT

Gain-of-function mutations in the proto-oncogene c-kit that induce constitutive kinase activity of its product, KIT protein, are characteristic of human mast cell disease and are believed to play a central role in mast cell leukemia oncogenesis, proliferation and survival. Nuclear overexpression of the Wnt effector beta-catenin and deregulated beta-catenin nuclear signaling can promote malignant transformation in solid tumors and hematologic malignancies. However, a role for beta-catenin in mast cell leukemia has not been described. Nuclear accumulation of beta-catenin is upregulated by its tyrosine phosphorylation, a process that can be exacerbated by deregulated expression of oncogenic tyrosine kinases. Here, we investigated the relationship between activated KIT and beta-catenin signaling in mast cell leukemia. Beta-catenin was tyrosine-phosphorylated in cells with KIT activated by either gain-of-function mutation or incubation with the KIT ligand stem cell factor. Beta-catenin tyrosine phosphorylation depended on KIT activity but not on PI3K-AKT activation. Tyrosine phosphorylation of beta-catenin was associated with its nuclear localization and enhanced transcription of target genes c-myc and cyclin D1. Endogenous KIT and beta-catenin were found to associate in mast cell leukemia cells, and in vitro kinase assay demonstrated that active KIT phosphorylates tyrosine residues of beta-catenin directly. Aberrant beta-catenin-driven transcription caused by deregulated KIT may represent a significant new target for treatment of mast cell leukemia.


Subject(s)
Leukemia, Mast-Cell/metabolism , Proto-Oncogene Proteins c-kit/physiology , Tyrosine/metabolism , beta Catenin/metabolism , Active Transport, Cell Nucleus , Benzamides , Humans , Imatinib Mesylate , Leukemia, Mast-Cell/drug therapy , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Piperazines/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/physiology , Pyrimidines/pharmacology , Signal Transduction
19.
Blood ; 107(2): 752-9, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16189265

ABSTRACT

In most patients with systemic mastocytosis (SM), including aggressive SM and mast cell leukemia (MCL), neoplastic cells express the oncogenic KIT mutation D816V. KIT D816V is associated with constitutive tyrosine kinase (TK) activity and thus represents an attractive drug target. However, imatinib and most other TK inhibitors fail to block the TK activity of KIT D816V. We show that the novel TK-targeting drugs PKC412 and AMN107 counteract TK activity of D816V KIT and inhibit the growth of Ba/F3 cells with doxycycline-inducible expression of KIT D816V as well as the growth of primary neoplastic mast cells and HMC-1 cells harboring this KIT mutation. PKC412 was a superior agent with median inhibitory concentration (IC(50)) values of 50 to 250 nM without differences seen between HMC-1 cells exhibiting or lacking KIT D816V. By contrast, AMN107 exhibited more potent effects in KIT D816V(-) HMC-1 cells. Corresponding results were obtained with Ba/F3 cells exhibiting wild-type or D816V-mutated KIT. The growth-inhibitory effects of PKC412 and AMN107 on HMC-1 cells were associated with induction of apoptosis and down-regulation of CD2 and CD63. PKC412 was found to cooperate with AMN107, imatinib, and cladribine (2CdA) in producing growth inhibition in HMC-1, but synergistic drug interactions were observed only in cells lacking KIT D816V. Together, PKC412 and AMN107 represent promising novel agents for targeted therapy of SM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Synergism , Leukemia, Mast-Cell/drug therapy , Leukemia, Mast-Cell/genetics , Mutation/genetics , Proto-Oncogene Proteins c-kit/genetics , Staurosporine/analogs & derivatives , Anti-Bacterial Agents/pharmacology , Antigens, CD/metabolism , Apoptosis/drug effects , Benzamides , CD2 Antigens/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cladribine/administration & dosage , Down-Regulation , Doxycycline/pharmacology , Drug Evaluation, Preclinical , Drug Interactions , Female , Humans , Imatinib Mesylate , Immunophenotyping , In Vitro Techniques , Leukemia, Mast-Cell/metabolism , Middle Aged , Phosphorylation/drug effects , Piperazines/administration & dosage , Platelet Membrane Glycoproteins/metabolism , Protein Kinase C/antagonists & inhibitors , Pyrimidines/administration & dosage , Staurosporine/pharmacology , Tetraspanin 30
20.
Blood ; 106(8): 2865-70, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-15972446

ABSTRACT

The majority of patients with systemic mast cell disease express the imatinib-resistant Asp816Val (D816V) mutation in the KIT receptor tyrosine kinase. Limited treatment options exist for aggressive systemic mastocytosis (ASM) and mast cell leukemia (MCL). We evaluated whether PKC412, a small-molecule inhibitor of KIT with a different chemical structure from imatinib, may have therapeutic use in advanced SM with the D816V KIT mutation. We treated a patient with MCL (with an associated myelodysplastic syndrome (MDS)/myeloproliferative disorder [MPD]) based on in vitro studies demonstrating that PKC412 could inhibit D816V KIT-transformed Ba/F3 cell growth with a 50% inhibitory concentration (IC50) of 30 nM to 40 nM. The patient exhibited a partial response with significant resolution of liver function abnormalities. In addition, PKC412 treatment resulted in a significant decline in the percentage of peripheral blood mast cells and serum histamine level and was associated with a decrease in KIT phosphorylation and D816V KIT mutation frequency. The patient died after 3 months of therapy due to progression of her MDS/MPD to acute myeloid leukemia (AML). This case indicates that KIT tyrosine kinase inhibition is a feasible approach in SM, but single-agent clinical efficacy may be limited by clonal evolution in the advanced leukemic phase of this disease.


Subject(s)
Aspartic Acid/genetics , Leukemia, Mast-Cell/drug therapy , Leukemia, Mast-Cell/genetics , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/genetics , Staurosporine/analogs & derivatives , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Immunophenotyping , Leukemia, Mast-Cell/metabolism , Leukemia, Mast-Cell/pathology , Middle Aged , Mutation/genetics , Phosphorylation/drug effects , Protein-Tyrosine Kinases/metabolism , Staurosporine/pharmacokinetics , Staurosporine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...