Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
Leukemia ; 36(7): 1843-1849, 2022 07.
Article in English | MEDLINE | ID: mdl-35654819

ABSTRACT

Mutations of the JAK2 gene are frequent aberrations in the aging hematopoietic system and in myeloid neoplasms. While JAK-inhibitors efficiently reduce hyperinflammation induced by the constitutively active mutated JAK2 kinase, the malignant clone and abundance of mutated cells remains rather unaffected. Here, we sought to assess for genetic vulnerabilities of JAK2-mutated clones. We identified lysine-specific demethylase KDM4C as a selective genetic dependency that persists upon JAK-inhibitor treatment. Genetic inactivation of KDM4C in human and murine JAK2-mutated cells resulted in loss of cell competition and reduced proliferation. These findings led to reduced disease penetrance and improved survival in xenograft models of human JAK2-mutated cells. KDM4C deleted cells showed alterations in target histone residue methylation and target gene expression, resulting in induction of cellular senescence. In summary, these data establish KDM4C as a specific dependency and therapeutic target in JAK2-mutated cells that is essential for oncogenic signaling and prevents induction of senescence.


Subject(s)
Histone Demethylases , Neoplasms , Animals , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Methylation , Mice , Neoplasms/genetics , Signal Transduction
3.
Nature ; 588(7836): 157-163, 2020 12.
Article in English | MEDLINE | ID: mdl-33239784

ABSTRACT

Janus kinases (JAKs) mediate responses to cytokines, hormones and growth factors in haematopoietic cells1,2. The JAK gene JAK2 is frequently mutated in the ageing haematopoietic system3,4 and in haematopoietic cancers5. JAK2 mutations constitutively activate downstream signalling and are drivers of myeloproliferative neoplasm (MPN). In clinical use, JAK inhibitors have mixed effects on the overall disease burden of JAK2-mutated clones6,7, prompting us to investigate the mechanism underlying disease persistence. Here, by in-depth phosphoproteome profiling, we identify proteins involved in mRNA processing as targets of mutant JAK2. We found that inactivation of YBX1, a post-translationally modified target of JAK2, sensitizes cells that persist despite treatment with JAK inhibitors to apoptosis and results in RNA mis-splicing, enrichment for retained introns and disruption of the transcriptional control of extracellular signal-regulated kinase (ERK) signalling. In combination with pharmacological JAK inhibition, YBX1 inactivation induces apoptosis in JAK2-dependent mouse and primary human cells, causing regression of the malignant clones in vivo, and inducing molecular remission. This identifies and validates a cell-intrinsic mechanism whereby differential protein phosphorylation causes splicing-dependent alterations of JAK2-ERK signalling and the maintenance of JAK2V617F malignant clones. Therapeutic targeting of YBX1-dependent ERK signalling in combination with JAK2 inhibition could thus eradicate cells harbouring mutations in JAK2.


Subject(s)
Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Neoplasms/genetics , Neoplasms/pathology , Y-Box-Binding Protein 1/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cells, Cultured , Clone Cells/metabolism , Clone Cells/pathology , Female , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Introns/genetics , Janus Kinase 2/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Male , Mice , Mutation , Neoplasm Transplantation , Neoplasms/drug therapy , Phosphoproteins/analysis , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteome/analysis , Proteomics , RNA Splicing/genetics , Remission Induction , Y-Box-Binding Protein 1/antagonists & inhibitors , Y-Box-Binding Protein 1/chemistry
4.
Nat Cell Biol ; 22(11): 1382-1395, 2020 11.
Article in English | MEDLINE | ID: mdl-33077911

ABSTRACT

Mutant KRAS modulates the metabolic plasticity of cancer cells to confer a growth advantage during hypoxia, but the molecular underpinnings are largely unknown. Using a lipidomic screen, we found that PLCγ1 is suppressed during hypoxia in KRAS-mutant human lung adenocarcinoma cancer cell lines. Suppression of PLCγ1 in hypoxia promotes a less oxidative cancer cell metabolism state, reduces the formation of mitochondrial reactive oxygen species and switches tumour bioenergetics towards glycolysis by impairing Ca2+ entry into the mitochondria. This event prevents lipid peroxidation, antagonizes apoptosis and increases cancer cell proliferation. Accordingly, loss of function of Plcg1 in a mouse model of KrasG12D-driven lung adenocarcinoma increased the expression of glycolytic genes, boosted tumour growth and reduced survival. In patients with KRAS-mutant lung adenocarcinomas, low PLCγ1 expression correlates with increased expression of hypoxia markers and predicts poor patient survival. Thus, our work reveals a mechanism of cancer cell adaptation to hypoxia with potential therapeutic value.


Subject(s)
Adenocarcinoma of Lung/enzymology , Lung Neoplasms/enzymology , Mutation , Phospholipase C gamma/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Hypoxia , A549 Cells , Adaptation, Physiological , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Animals , Cell Proliferation , Cell Survival , Energy Metabolism , Female , Humans , Lipid Peroxidation , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice, Inbred NOD , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/pathology , Phospholipase C gamma/genetics , Signal Transduction
5.
Nat Commun ; 10(1): 2031, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31048683

ABSTRACT

Mutations in the nucleophosmin 1 (NPM1) gene are considered founder mutations in the pathogenesis of acute myeloid leukemia (AML). To characterize the genetic composition of NPM1 mutated (NPM1mut) AML, we assess mutation status of five recurrently mutated oncogenes in 129 paired NPM1mut samples obtained at diagnosis and relapse. We find a substantial shift in the genetic pattern from diagnosis to relapse including NPM1mut loss (n = 11). To better understand these NPM1mut loss cases, we perform whole exome sequencing (WES) and RNA-Seq. At the time of relapse, NPM1mut loss patients (pts) feature distinct mutational patterns that share almost no somatic mutation with the corresponding diagnosis sample and impact different signaling pathways. In contrast, profiles of pts with persistent NPM1mut are reflected by a high overlap of mutations between diagnosis and relapse. Our findings confirm that relapse often originates from persistent leukemic clones, though NPM1mut loss cases suggest a second "de novo" or treatment-associated AML (tAML) as alternative cause of relapse.


Subject(s)
Clonal Evolution , Leukemia, Myeloid, Acute/genetics , Neoplasm Recurrence, Local/genetics , Neoplasms, Second Primary/genetics , Nuclear Proteins/genetics , Adult , Aged , DNA Mutational Analysis , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Mutation , Neoplasm Recurrence, Local/pathology , Neoplasms, Second Primary/pathology , Nucleophosmin , Exome Sequencing
6.
Oncogene ; 38(24): 4773-4787, 2019 06.
Article in English | MEDLINE | ID: mdl-30820040

ABSTRACT

The receptor tyrosine kinase FLT3 is expressed in myeloid and lymphoid progenitor cells. Activating mutations in FLT3 occur in 25-30% of acute myeloid leukaemia (AML) patients. Most common are internal tandem duplications of sequence (ITD) leading to constitutive FLT3-ITD kinase activity with an altered signalling quality promoting leukaemic cell transformation. Here, we observed the attenuating role of the receptor-like protein tyrosine phosphatase (RPTP) CD45/Ptprc in FLT3 signalling in vivo. Low level expression of this abundant RPTP correlates with a poor prognosis of FLT3-ITD-positive AML patients. To get a further insight into the regulatory role of Ptprc in FLT3-ITD activity in vivo, Ptprc knock-out mice were bred with FLT3-ITD knock-in mice. Inactivation of the Ptprc gene in FLT3-ITD mice resulted in a drastically shortened life span and development of severe monocytosis, a block in B-cell development and anaemia. The myeloproliferative phenotype was associated with extramedullary haematopoiesis, splenohepatomegaly and severe alterations of organ structures. The phenotypic alterations were associated with increased transforming signalling of FLT3-ITD, including activation of its downstream target STAT5. These data reveal the capacity of Ptprc for the regulation of FLT3-ITD signalling activity in vivo. In addition, histopathology and computed tomography (CT) revealed an unexpected bone phenotype; the FLT3-ITD Ptprc-/- mice, but none of the controls, showed pronounced alterations in bone morphology and, in part, apparent features of osteoporosis. In the spleen, ectopic bone formation was observed. The observed bone phenotypes suggest a previously unappreciated capacity of FLT3-ITD (and presumably FLT3) to regulate bone development/remodelling, which is under negative control of CD45/Ptprc.


Subject(s)
Bone and Bones , Leukocyte Common Antigens/genetics , Myeloproliferative Disorders/genetics , Osteoporosis/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Bone Development/genetics , Bone Remodeling/genetics , Cell Transformation, Neoplastic , Cells, Cultured , Choristoma/genetics , Choristoma/metabolism , Embryo, Mammalian , Female , Humans , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukocyte Common Antigens/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/pathology , Osteogenesis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Phenotype , Porosity , Tandem Repeat Sequences/genetics
7.
Cell Commun Signal ; 17(1): 5, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30651113

ABSTRACT

BACKGROUND: Treatment of acute leukemia is challenging and long-lasting remissions are difficult to induce. Innovative therapy approaches aim to complement standard chemotherapy to improve drug efficacy and decrease toxicity. Promising new therapeutic targets in cancer therapy include voltage-gated Kv1.3 potassium channels, but their role in acute leukemia is unclear. We reported that Kv1.3 channels of lymphocytes are blocked by memantine, which is known as an antagonist of neuronal N-methyl-D-aspartate type glutamate receptors and clinically applied in therapy of advanced Alzheimer disease. Here we evaluated whether pharmacological targeting of Kv1.3 channels by memantine promotes cell death of acute leukemia cells induced by chemotherapeutic cytarabine. METHODS: We analyzed acute lymphoid (Jurkat, CEM) and myeloid (HL-60, Molm-13, OCI-AML-3) leukemia cell lines and patients' acute leukemic blasts after treatment with either drug alone or the combination of cytarabine and memantine. Patch-clamp analysis was performed to evaluate inhibition of Kv1.3 channels and membrane depolarization by memantine. Cell death was determined with propidium iodide, Annexin V and SYTOX staining and cytochrome C release assay. Molecular effects of memantine co-treatment on activation of Caspases, AKT, ERK1/2, and JNK signaling were analysed by Western blot. Kv1.3 channel expression in Jurkat cells was downregulated by shRNA. RESULTS: Our study demonstrates that memantine inhibits Kv1.3 channels of acute leukemia cells and in combination with cytarabine potentiates cell death of acute lymphoid and myeloid leukemia cell lines as well as primary leukemic blasts from acute leukemia patients. At molecular level, memantine co-application fosters concurrent inhibition of AKT, S6 and ERK1/2 and reinforces nuclear down-regulation of MYC, a common target of AKT and ERK1/2 signaling. In addition, it augments mitochondrial dysfunction resulting in enhanced cytochrome C release and activation of Caspase-9 and Caspase-3 leading to amplified apoptosis. CONCLUSIONS: Our study underlines inhibition of Kv1.3 channels as a therapeutic strategy in acute leukemia and proposes co-treatment with memantine, a licensed and safe drug, as a potential approach to promote cytarabine-based cell death of various subtypes of acute leukemia.


Subject(s)
Apoptosis/drug effects , Cytarabine/pharmacology , Kv1.3 Potassium Channel/antagonists & inhibitors , Leukemia, Myeloid, Acute/pathology , MAP Kinase Signaling System/drug effects , Memantine/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Adenosine Triphosphate/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochromes c/metabolism , Humans , Kv1.3 Potassium Channel/metabolism , Proto-Oncogene Proteins c-myc/metabolism
8.
J Cancer Res Clin Oncol ; 144(10): 1933-1944, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30083817

ABSTRACT

PURPOSE: Cell fate determinants Scrib and Llgl1 influence self-renewal capacity of hematopoietic stem cells (HSCs). Scrib-deficient HSCs are functionally impaired and lack sufficient repopulation capacity during serial transplantation and stress. In contrast, loss of Llgl1 leads to increased HSC fitness, gain of self-renewal capacity and expansion of the stem cell pool. Here, we sought to assess for shared and unique molecular functions of Llgl1 and Scrib by analyzing their interactome in hematopoietic cells. METHODS: Interactome analysis was performed by affinity purification followed by mass spectrometry. Motility, migration and adhesion were assessed on primary murine HSCs, which were isolated by FACS sorting following conditional deletion of Scrib or Llgl1, respectively. Imaging of Scrib-deficient HSCs was performed by intravital 2-photon microscopy. RESULTS: Comparison of Scrib and Llgl1 interactome analyses revealed involvement in common and unique cellular functions. Migration and adhesion were among the cellular functions connected to Scrib but not to Llgl1. Functional validation of these findings confirmed alterations in cell adhesion and migration of Scrib-deficient HSCs in vitro and in vivo. In contrast, genetic inactivation of Llgl1 did not affect adhesion or migratory capacity of hematopoietic stem cells. CONCLUSION: Our data provide first evidence for an evolutionarily conserved role of the cell fate determinant Scrib in HSC adhesion and migration in vitro and in vivo, a unique function that is not shared with its putative complex partner Llgl1.


Subject(s)
Cell Adhesion , Cell Lineage , Cell Movement , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Proteome/analysis , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytoskeletal Proteins , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Protein Interaction Domains and Motifs
10.
Nat Commun ; 8(1): 311, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28827518

ABSTRACT

Graft-vs.-host disease (GvHD) is a major complication of allogenic hematopoietic stem-cell(HSC) transplantation. GvHD is associated with loss of endothelial thrombomodulin, but the relevance of this for the adaptive immune response to transplanted HSCs remains unknown. Here we show that the protease-activated protein C (aPC), which is generated by thrombomodulin, ameliorates GvHD aPC restricts allogenic T-cell activation via the protease activated receptor (PAR)2/PAR3 heterodimer on regulatory T-cells (Tregs, CD4+FOXP3+). Preincubation of pan T-cells with aPC prior to transplantation increases the frequency of Tregs and protects from GvHD. Preincubation of human T-cells (HLA-DR4-CD4+) with aPC prior to transplantation into humanized (NSG-AB°DR4) mice ameliorates graft-vs.-host disease. The protective effect of aPC on GvHD does not compromise the graft vs. leukaemia effect in two independent tumor cell models. Ex vivo preincubation of T-cells with aPC, aPC-based therapies, or targeting PAR2/PAR3 on T-cells may provide a safe and effective approach to mitigate GvHD.Graft-vs.-host disease is a complication of allogenic hematopoietic stem cell transplantation, and is associated with endothelial dysfunction. Here the authors show that activated protein C signals via PAR2/PAR3 to expand Treg cells, mitigating the disease in mice.


Subject(s)
Graft vs Host Disease/immunology , Protein C/immunology , Receptor, PAR-2/immunology , Receptors, Proteinase-Activated/immunology , Receptors, Thrombin/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Kaplan-Meier Estimate , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Protein C/metabolism , Protein Multimerization , Receptor, PAR-2/chemistry , Receptor, PAR-2/metabolism , Receptors, Proteinase-Activated/chemistry , Receptors, Proteinase-Activated/metabolism , Receptors, Thrombin/chemistry , Receptors, Thrombin/metabolism , Signal Transduction/immunology , T-Lymphocytes, Regulatory/metabolism , Transplantation, Homologous
11.
Oncotarget ; 8(16): 26613-26624, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28460451

ABSTRACT

Fms-like tyrosine kinase 3 (FLT3) with internal tandem duplications (ITD) is a major oncoprotein in acute myeloid leukemia (AML), and confers an unfavorable prognosis. Interference with FLT3ITD signaling is therefore pursued as a promising therapeutic strategy. In this study we show that abrogation of FLT3ITD glycoprotein maturation using low doses of the N-glycosylation inhibitor tunicamycin has anti-proliferative and pro-apoptotic effects on FLT3ITD-expressing human and murine cell lines. This effect is mediated in part by arresting FLT3ITD in an underglycosylated state and thereby attenuating FLT3ITD-driven AKT and ERK signaling. In addition, tunicamycin caused pronounced endoplasmatic reticulum stress and apoptosis through activation of protein kinase RNA-like endoplasmic reticulum kinase (PERK) and activation of the gene encoding CCAAT-enhancer-binding protein homologous protein (CHOP). PERK inhibition with a small molecule attenuated CHOP induction and partially rescued cells from apoptosis. Combination of tunicamycin with potent FLT3ITD kinase inhibitors caused synergistic cell killing, which was highly selective for cell lines and primary AML cells expressing FLT3ITD. Although tunicamycin is currently not a clinically applicable drug, we propose that mild inhibition of N-glycosylation may have therapeutic potential in combination with FLT3 kinase inhibitors for FLT3ITD-positive AML.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Duplication , Leukemia, Myeloid, Acute/genetics , Protein Kinase Inhibitors/pharmacology , Tandem Repeat Sequences , fms-Like Tyrosine Kinase 3/genetics , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Drug Synergism , Endoplasmic Reticulum Stress , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Glycosylation/drug effects , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins c-akt/metabolism , Tumor Cells, Cultured , Tunicamycin/pharmacology , fms-Like Tyrosine Kinase 3/metabolism
12.
J Cancer Res Clin Oncol ; 143(5): 807-820, 2017 May.
Article in English | MEDLINE | ID: mdl-28233092

ABSTRACT

PURPOSE: Myeloproliferative neoplasms (MPN) are clonal disorders of hematopoietic stem- and progenitor cells. Mutation of Janus-Kinase 2 (JAK2) is the most frequent genetic event detected in Philadelphia-negative MPN. In advanced phases, the clinical hallmark of the disease is a striking inflammatory syndrome. So far, the cellular and molecular basis of inflammation is not fully understood. We, therefore, sought to investigate the relationship of activating JAK2 mutation and aberrant cytokine expression in MPN. METHODS: Cytokine array was performed to identify Jak2V617F-related cytokine expression and secretion. CXCL10 mRNA expression was analyzed by qPCR in peripheral blood cells. To exclude paracrine/autocrine stimulation as a potential mechanism, we generated Ba/F3-EpoR-JAK2WT or EpoR-JAK2V617F cells lacking CXCL10 receptor. Pharmacologic inhibition of JAK2 kinase was achieved by JAK-Inhibitor treatment. Signaling pathways and downstream effectors were characterized by Western blotting, immunofluorescence microscopy, luciferase reporter assays, qPCR, and chromatin-immunoprecipitation studies. RESULTS: We identified CXCL10 as the most highly induced cytokine in JAK2-mutated cell lines. In MPN patients, CXCL10 is highly expressed in JAK2V617F but not JAK2WT MPN or healthy donor controls. Moreover, CXCL10 expression correlates with JAK2V617F allelic burden. High CXCL10 correlates with the presence of clinical risk factors but not with clinical symptoms and quality of life. Pharmacologic inhibition of mutated JAK2 kinase inhibits CXCL10 expression. NFκB signaling is activated downstream of JAK2V617F receptor and directly induces CXCL10 expression. CONCLUSIONS: Our data provide first evidence for a link between oncogenic JAK2V617F signaling and cell intrinsic induction of CXCL10 induced by activated NFkB signaling.


Subject(s)
Chemokine CXCL10/biosynthesis , Janus Kinase 2/genetics , Myeloproliferative Disorders/metabolism , Animals , Cell Line, Tumor , Chemokine CXCL10/genetics , Humans , Janus Kinase 2/metabolism , Mice , Mutation , Myeloproliferative Disorders/blood , Myeloproliferative Disorders/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
13.
Toxins (Basel) ; 8(12)2016 12 06.
Article in English | MEDLINE | ID: mdl-27929418

ABSTRACT

RTA, the catalytic A-subunit of the ribosome inactivating A/B toxin ricin, inhibits eukaryotic protein biosynthesis by depurination of 28S rRNA. Although cell surface binding of ricin holotoxin is mainly mediated through its B-subunit (RTB), sole application of RTA is also toxic, albeit to a significantly lower extent, suggesting alternative pathways for toxin uptake and transport. Since ricin toxin trafficking in mammalian cells is still not fully understood, we developed a GFP-based reporter assay in yeast that allows rapid identification of cellular components required for RTA uptake and subsequent transport through a target cell. We hereby show that Ypt6p, Sft2p and GARP-complex components play an important role in RTA transport, while neither the retromer complex nor COPIB vesicles are part of the transport machinery. Analyses of yeast knock-out mutants with chromosomal deletion in genes whose products regulate ADP-ribosylation factor GTPases (Arf-GTPases) and/or retrograde Golgi-to-ER (endoplasmic reticulum) transport identified Sso1p, Snc1p, Rer1p, Sec22p, Erv46p, Gea1p and Glo3p as novel components in RTA transport, suggesting the developed reporter assay as a powerful tool to dissect the multistep processes of host cell intoxication in yeast.


Subject(s)
Ricin/metabolism , Saccharomyces cerevisiae/metabolism , Biological Assay , Green Fluorescent Proteins/genetics , Protein Transport , Saccharomyces cerevisiae/genetics
14.
Anticancer Res ; 36(12): 6249-6258, 2016 12.
Article in English | MEDLINE | ID: mdl-27919943

ABSTRACT

BACKGROUND/AIM: The presence of FLT3-Internal tandem duplications (ITDs) in human acute myeloid leukemia (AML) is associated with a dismal prognosis. Altered cell-cycle activity has been reported in FLT3-ITD-positive AML; however, the mechanisms by which this oncogene influences cell-cycle activity remained so far elusive. MATERIALS AND METHODS: A phospho-kinomic screen was used to identify downstream effectors of FLT3-ITD. Validation and functional characterization was performed by western blotting, cell-cycle analysis and apoptosis assays. RESULTS: We identified aberrant phosphorylation of CDC25C-T48 in FLT3-ITD mutated cells. Forced expression of CDC25C affected cell-cycle progression but did not affect sensitivity to cellular stress. CONCLUSION: Depending on the oncogenic background, CDC25C may reveal protective or oncogenic functions. Our results identify CDC25C as a downstream target of the mutated tyrosine kinase FLT3-ITD affecting cell-cycle regulation in a model of AML.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , cdc25 Phosphatases/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Humans , Phosphorylation
15.
Haematologica ; 101(3): e81-5, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26589910

Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Gene Expression Regulation, Neoplastic , Multidrug Resistance-Associated Proteins/genetics , Neoplasm Proteins/genetics , Polycythemia Vera/genetics , Primary Myelofibrosis/genetics , Pyrazoles/pharmacology , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/immunology , ATP Binding Cassette Transporter, Subfamily G, Member 2/immunology , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymphocyte Activation/drug effects , Multidrug Resistance-Associated Proteins/immunology , Mutation , Neoplasm Proteins/immunology , Nitriles , Phytohemagglutinins/pharmacology , Polycythemia Vera/drug therapy , Polycythemia Vera/immunology , Polycythemia Vera/pathology , Primary Cell Culture , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/immunology , Primary Myelofibrosis/pathology , Protein Kinase Inhibitors/pharmacology , Pyrimidines , Signal Transduction , Treatment Outcome
17.
PLoS One ; 7(7): e40853, 2012.
Article in English | MEDLINE | ID: mdl-22815843

ABSTRACT

Clinical development of imatinib in CML established continuous target inhibition as a paradigm for successful tyrosine kinase inhibitor (TKI) therapy. However, recent reports suggested that transient potent target inhibition of BCR-ABL by high-dose TKI (HD-TKI) pulse-exposure is sufficient to irreversibly commit cells to apoptosis. Here, we report a novel mechanism of prolonged intracellular TKI activity upon HD-TKI pulse-exposure (imatinib, dasatinib) in BCR-ABL-positive cells. Comprehensive mechanistic exploration revealed dramatic intracellular accumulation of TKIs which closely correlated with induction of apoptosis. Cells were rescued from apoptosis upon HD-TKI pulse either by repetitive drug wash-out or by overexpression of ABC-family drug transporters. Inhibition of ABCB1 restored sensitivity to HD-TKI pulse-exposure. Thus, our data provide evidence that intracellular drug retention crucially determines biological activity of imatinib and dasatinib. These studies may refine our current thinking on critical requirements of TKI dose and duration of target inhibition for biological activity of TKIs.


Subject(s)
Apoptosis/drug effects , Intracellular Space/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Annexin A5/metabolism , Benzamides , Caspase 3/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Imatinib Mesylate , Intracellular Space/drug effects , Kinetics , Piperazines/pharmacology , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Time Factors
18.
Mol Biol Cell ; 21(15): 2543-54, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20519439

ABSTRACT

We report that a toxic polypeptide retaining the potential to refold upon dislocation from the endoplasmic reticulum (ER) to the cytosol (ricin A chain; RTA) and a misfolded version that cannot (termed RTA(Delta)), follow ER-associated degradation (ERAD) pathways in Saccharomyces cerevisiae that substantially diverge in the cytosol. Both polypeptides are dislocated in a step mediated by the transmembrane Hrd1p ubiquitin ligase complex and subsequently degraded. Canonical polyubiquitylation is not a prerequisite for this interaction because a catalytically inactive Hrd1p E3 ubiquitin ligase retains the ability to retrotranslocate RTA, and variants lacking one or both endogenous lysyl residues also require the Hrd1p complex. In the case of native RTA, we established that dislocation also depends on other components of the classical ERAD-L pathway as well as an ongoing ER-Golgi transport. However, the dislocation pathways deviate strikingly upon entry into the cytosol. Here, the CDC48 complex is required only for RTA(Delta), although the involvement of individual ATPases (Rpt proteins) in the 19S regulatory particle (RP) of the proteasome, and the 20S catalytic chamber itself, is very different for the two RTA variants. We conclude that cytosolic ERAD components, particularly the proteasome RP, can discriminate between structural features of the same substrate.


Subject(s)
Endoplasmic Reticulum/metabolism , Protein Folding , Ricin/chemistry , Ricin/metabolism , Cytosol/metabolism , Gene Deletion , Gene Library , Golgi Apparatus/metabolism , Lysine/metabolism , Models, Biological , Molecular Chaperones/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Protein Transport , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...