158 results on '"Böhmer FD"'
Search Results
2. Diabetes mellitus activates signal transduction pathways resulting in vascular endothelial growth factor resistance of human monocytes.
- Author
-
Tchaikovski V, Olieslagers S, Böhmer FD, and Waltenberger J
- Published
- 2009
- Full Text
- View/download PDF
3. Context-specific effects of NOX4 inactivation in acute myeloid leukemia (AML).
- Author
-
Demircan MB, Schnoeder TM, Mgbecheta PC, Schröder K, Böhmer FD, and Heidel FH
- Subjects
- Animals, Humans, Mice, Mice, Knockout, Mutation, Reactive Oxygen Species metabolism, fms-Like Tyrosine Kinase 3 genetics, Leukemia, Myeloid, Acute pathology, Myeloproliferative Disorders genetics, NADPH Oxidase 4 genetics
- Abstract
Purpose: Oxidative stress has been linked to initiation and progression of cancer and recent studies have indicated a potential translational role regarding modulation of ROS in various cancers, including acute myeloid leukemia (AML). Detailed understanding of the complex machinery regulating ROS including its producer elements in cancer is required to define potential translational therapeutic use. Based on previous studies in acute myeloid leukemia (AML) models, we considered NADPH oxidase (NOX) family members, specifically NOX4 as a potential target in AML., Methods: Pharmacologic inhibition and genetic inactivation of NOX4 in murine and human models of AML were used to understand its functional role. For genetic inactivation, CRISPR-Cas9 technology was used in human AML cell lines in vitro and genetically engineered knockout mice for Nox4 were used for deletion of Nox4 in hematopoietic cells via Mx1-Cre recombinase activation., Results: Pharmacologic NOX inhibitors and CRISPR-Cas9-mediated inactivation of NOX4 and p22-phox (an essential NOX component) decreased proliferative capacity and cell competition in FLT3-ITD-positive human AML cells. In contrast, conditional deletion of Nox4 enhanced the myeloproliferative phenotype of an FLT3-ITD induced knock-in mouse model. Finally, Nox4 inactivation in normal hematopoietic stem and progenitor cells (HSPCs) caused a minor reduction in HSC numbers and reconstitution capacity., Conclusion: The role of NOX4 in myeloid malignancies appears highly context-dependent and its inactivation results in either enhancing or inhibitory effects. Therefore, targeting NOX4 in FLT3-ITD positive myeloid malignancies requires additional pre-clinical assessment., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
4. Combined Activity of the Redox-Modulating Compound Setanaxib (GKT137831) with Cytotoxic Agents in the Killing of Acute Myeloid Leukemia Cells.
- Author
-
Demircan MB, Mgbecheta PC, Kresinsky A, Schnoeder TM, Schröder K, Heidel FH, and Böhmer FD
- Abstract
Acute myeloid leukemia (AML) cells harbor elevated levels of reactive oxygen species (ROS), which promote cell proliferation and cause oxidative stress. Therefore, the inhibition of ROS formation or elevation beyond a toxic level have been considered as therapeutic strategies. ROS elevation has recently been linked to enhanced NADPH oxidase 4 (NOX4) activity. Therefore, the compound Setanaxib (GKT137831), a clinically advanced ROS-modulating substance, which has initially been identified as a NOX1/4 inhibitor, was tested for its inhibitory activity on AML cells. Setanaxib showed antiproliferative activity as single compound, and strongly enhanced the cytotoxic action of anthracyclines such as daunorubicin in vitro. Setanaxib attenuated disease in a mouse model of FLT3-ITD driven myeloproliferation in vivo. Setanaxib did not significantly inhibit FLT3-ITD signaling, including FLT3 autophosphorylation, activation of STAT5, AKT, or extracellular signal regulated kinase 1 and 2 (ERK1/2). Surprisingly, the effects of Setanaxib on cell proliferation appeared to be independent of the presence of NOX4 and were not associated with ROS quenching. Instead, Setanaxib caused elevation of ROS levels in the AML cells and importantly, enhanced anthracycline-induced ROS formation, which may contribute to the combined effects. Further assessment of Setanaxib as potential enhancer of cytotoxic AML therapy appears warranted.
- Published
- 2022
- Full Text
- View/download PDF
5. The translation attenuating arginine-rich sequence in the extended signal peptide of the protein-tyrosine phosphatase PTPRJ/DEP1 is conserved in mammals.
- Author
-
Karagyozov L, Grozdanov PN, and Böhmer FD
- Subjects
- Animals, Arginine genetics, Codon, Initiator genetics, Humans, RNA, Messenger genetics, Receptor, Notch3 genetics, Repetitive Sequences, Nucleic Acid genetics, Sequence Alignment, Conserved Sequence genetics, Protein Biosynthesis genetics, Protein Sorting Signals genetics, Receptor-Like Protein Tyrosine Phosphatases, Class 3 genetics
- Abstract
The signal peptides, present at the N-terminus of many proteins, guide the proteins into cell membranes. In some proteins, the signal peptide is with an extended N-terminal region. Previously, it was demonstrated that the N-terminally extended signal peptide of the human PTPRJ contains a cluster of arginine residues, which attenuates translation. The analysis of the mammalian orthologous sequences revealed that this sequence is highly conserved. The PTPRJ transcripts in placentals, marsupials, and monotremes encode a stretch of 10-14 arginine residues, positioned 11-12 codons downstream of the initiating AUG. The remarkable conservation of the repeated arginine residues in the PTPRJ signal peptides points to their key role. Further, the presence of an arginine cluster in the extended signal peptides of other proteins (E3 ubiquitin-protein ligase, NOTCH3) is noted and indicates a more general importance of this cis-acting mechanism of translational suppression., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
6. Mislocalisation of Activated Receptor Tyrosine Kinases - Challenges for Cancer Therapy.
- Author
-
Schmidt-Arras D and Böhmer FD
- Subjects
- Animals, Cell Membrane genetics, Humans, Mutation genetics, Signal Transduction genetics, Neoplasms genetics, Receptor Protein-Tyrosine Kinases genetics
- Abstract
Activating mutations in genes encoding receptor tyrosine kinases (RTKs) mediate proliferation, cell migration, and cell survival, and are therefore important drivers of oncogenesis. Numerous targeted cancer therapies are directed against activated RTKs, including small compound inhibitors, and immunotherapies. It has recently been discovered that not only certain RTK fusion proteins, but also many full-length RTKs harbouring activating mutations, notably RTKs of the class III family, are to a large extent mislocalised in intracellular membranes. Active kinases in these locations cause aberrant activation of signalling pathways. Moreover, low levels of activated RTKs at the cell surface present an obstacle for immunotherapy. We outline here why understanding of the mechanisms underlying mislocalisation will help in improving existing and developing novel therapeutic strategies., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
7. Correction: The acetyltransferase GCN5 maintains ATRA-resistance in non-APL AML.
- Author
-
Kahl M, Brioli A, Bens M, Perner F, Kresinsky A, Schnetzke U, Hinze A, Sbirkov Y, Stengel S, Simonetti G, Martinelli G, Petrie K, Zelent A, Böhmer FD, Groth M, Ernst T, Heidel FH, Scholl S, Hochhaus A, and Schenk T
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
8. A series of novel aryl-methanone derivatives as inhibitors of FMS-like tyrosine kinase 3 (FLT3) in FLT3-ITD-positive acute myeloid leukemia.
- Author
-
Sellmer A, Pilsl B, Beyer M, Pongratz H, Wirth L, Elz S, Dove S, Henninger SJ, Spiekermann K, Polzer H, Klaeger S, Kuster B, Böhmer FD, Fiebig HH, Krämer OH, and Mahboobi S
- Subjects
- Antineoplastic Agents chemical synthesis, Antineoplastic Agents chemistry, Cell Proliferation drug effects, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Humans, Indoles chemical synthesis, Indoles chemistry, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Models, Molecular, Molecular Structure, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors chemistry, Structure-Activity Relationship, Tumor Cells, Cultured, fms-Like Tyrosine Kinase 3 metabolism, Antineoplastic Agents pharmacology, Indoles pharmacology, Protein Kinase Inhibitors pharmacology, fms-Like Tyrosine Kinase 3 antagonists & inhibitors
- Abstract
Mutants of the FLT3 receptor tyrosine kinase (RTK) with duplications in the juxtamembrane domain (FLT3-ITD) act as drivers of acute myeloid leukemia (AML). Potent tyrosine kinase inhibitors (TKi) of FLT3-ITD entered clinical trials and showed a promising, but transient success due to the occurrence of secondary drug-resistant AML clones. A further caveat of drugs targeting FLT3-ITD is the co-targeting of other RTKs which are required for normal hematopoiesis. This is observed quite frequently. Therefore, novel drugs are necessary to treat AML effectively and safely. Recently bis(1H-indol-2-yl)methanones were found to inhibit FLT3 and PDGFR kinases. In order to optimize these agents we synthesized novel derivatives of these methanones with various substituents. Methanone 16 and its carbamate derivative 17b inhibit FLT3-ITD at least as potently as the TKi AC220 (quizartinib). Models indicate corresponding interactions of 16 and quizartinib with FLT3. The activity of 16 is accompanied by a high selectivity for FLT3-ITD., Competing Interests: Declaration of competing interests The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Mahboobi, Siavosh; Sellmer, Andreas; Pongratz, Herwig; Pilsl, Bernardette; Krämer, Oliver; Beyer, Mandy are also the inventors of this entity: PCT Int. Appl. (2019), WO 2019034538 A1 20190221., (Copyright © 2020 Elsevier Masson SAS. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
9. SHP1 regulates a STAT6-ITGB3 axis in FLT3ITD-positive AML cells.
- Author
-
Reich D, Kresinsky A, Müller JP, Bauer R, Kallenbach J, Schnoeder TM, Heidel FH, Fässler R, Mann M, Böhmer FD, and Jayavelu AK
- Subjects
- Animals, Apoptosis, Cell Proliferation, Humans, Integrin beta3 genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Mice, Mice, Inbred C3H, Protein Tyrosine Phosphatase, Non-Receptor Type 6 genetics, Repetitive Sequences, Nucleic Acid, STAT6 Transcription Factor genetics, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, fms-Like Tyrosine Kinase 3 genetics, Gene Expression Regulation, Leukemic, Integrin beta3 metabolism, Leukemia, Myeloid, Acute pathology, Protein Tyrosine Phosphatase, Non-Receptor Type 6 metabolism, STAT6 Transcription Factor metabolism, fms-Like Tyrosine Kinase 3 metabolism
- Published
- 2020
- Full Text
- View/download PDF
10. Wild-type FLT3 and FLT3 ITD exhibit similar ligand-induced internalization characteristics.
- Author
-
Kellner F, Keil A, Schindler K, Tschongov T, Hünninger K, Loercher H, Rhein P, Böhmer SA, Böhmer FD, and Müller JP
- Subjects
- Carcinogenesis, Gene Duplication genetics, Gene Expression Regulation, Neoplastic genetics, Hematopoietic Stem Cells pathology, Humans, Leukemia, Myeloid, Acute pathology, Ligands, Mutation, Tandem Repeat Sequences genetics, Cell Transformation, Neoplastic genetics, Leukemia, Myeloid, Acute genetics, Membrane Proteins genetics, STAT5 Transcription Factor genetics, fms-Like Tyrosine Kinase 3 genetics
- Abstract
Class III receptor tyrosine kinases control the development of hematopoietic stem cells. Constitutive activation of FLT3 by internal tandem duplications (ITD) in the juxtamembrane domain has been causally linked to acute myeloid leukaemia. Oncogenic FLT3 ITD is partially retained in compartments of the biosynthetic route and aberrantly activates STAT5, thereby promoting cellular transformation. The pool of FLT3 ITD molecules in the plasma membrane efficiently activates RAS and AKT, which is likewise essential for cell transformation. Little is known about features and mechanisms of FLT3 ligand (FL)-dependent internalization of surface-bound FLT3 or FLT3 ITD. We have addressed this issue by internalization experiments using human RS4-11 and MV4-11 cells with endogenous wild-type FLT3 or FLT3 ITD expression, respectively, and surface biotinylation. Further, FLT3 wild-type, or FLT3 ITD-GFP hybrid proteins were stably expressed and characterized in 32D cells, and internalization and stability were assessed by flow cytometry, imaging flow cytometry, and immunoblotting. FL-stimulated surface-exposed FLT3 WT or FLT3 ITD protein showed similar endocytosis and degradation characteristics. Kinase inactivation by mutation or FLT3 inhibitor treatment strongly promoted FLT3 ITD surface localization, and attenuated but did not abrogate FL-induced internalization. Experiments with the dynamin inhibitor dynasore suggest that active FLT3 as well as FLT3 ITD is largely endocytosed via clathrin-dependent endocytosis. Internalization of kinase-inactivated molecules occurred through a different yet unidentified mechanism. Our data demonstrate that FLT3 WT and constitutively active FLT3 ITD receptor follow, despite very different biogenesis kinetics, similar internalization and degradation routes., (© 2020 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
11. Loss of PTPRJ/DEP-1 enhances NF2/Merlin-dependent meningioma development.
- Author
-
Waldt N, Scharnetzki D, Kesseler C, Kirches E, Stroscher N, Böhmer FD, and Mawrin C
- Subjects
- Animals, Animals, Newborn, Cell Line, Tumor, Humans, Meningeal Neoplasms genetics, Meningeal Neoplasms pathology, Meningioma genetics, Meningioma pathology, Mice, Mice, Transgenic, Neurofibromin 2 genetics, Receptor-Like Protein Tyrosine Phosphatases, Class 3 deficiency, Receptor-Like Protein Tyrosine Phosphatases, Class 3 genetics, Meningeal Neoplasms metabolism, Meningioma metabolism, Neurofibromin 2 deficiency
- Abstract
Introduction: Meningiomas are common tumors in adults, which develop from the meningeal coverings of the brain and spinal cord. Loss-of-function mutations or deletion of the NF2 gene, resulting in loss of the encoded Merlin protein, lead to Neurofibromatosis type 2 (NF2), but also cause the formation of sporadic meningiomas. It was shown that inactivation of Nf2 in mice caused meningioma formation. Another meningioma tumor-suppressor candidate is the receptor-like density-enhanced phosphatase-1 (DEP-1), encoded by PTPRJ. Loss of DEP-1 enhances meningioma cell motility in vitro and invasive growth in an orthotopic xenograft model. Ptprj-deficient mice develop normally and do not show spontaneous tumorigenesis. Another genetic lesion may be required to interact with DEP-1 loss in meningioma genesis., Methods: In the present study we investigated in vitro and in vivo whether the losses of DEP-1 and Merlin/NF2 may have a combined effect., Results: Human meningioma cells deficient for DEP-1, Merlin/NF2 or both showed no statistically significant changes in cell proliferation, while DEP-1 or DEP1/NF2 deficiency led to moderately increased colony size in clonogenicity assays. In addition, the loss of any of the two genes was sufficient to induce a significant reduction of cell size (p < .05) and profound morphological changes. Most important, in Ptprj knockout mice Cre/lox mediated meningeal Nf2 knockout elicited a four-fold increased rate of meningioma formation within one year compared with mice with Ptprj wild type alleles (25% vs 6% tumor incidence)., Conclusions: Our data suggest that loss of DEP-1 and Merlin/NF2 synergize during meningioma genesis., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
12. Modulation of FLT3 signal transduction through cytoplasmic cysteine residues indicates the potential for redox regulation.
- Author
-
Böhmer A, Barz S, Schwab K, Kolbe U, Gabel A, Kirkpatrick J, Ohlenschläger O, Görlach M, and Böhmer FD
- Subjects
- Cell Line, Cytoplasm metabolism, HEK293 Cells, Humans, Models, Molecular, Oxidation-Reduction, Protein Conformation, Reactive Oxygen Species metabolism, Signal Transduction, fms-Like Tyrosine Kinase 3 genetics, Cyclohexanones pharmacology, Cysteine metabolism, Mutation, fms-Like Tyrosine Kinase 3 chemistry, fms-Like Tyrosine Kinase 3 metabolism
- Abstract
Oxidative modification of cysteine residues has been shown to regulate the activity of several protein-tyrosine kinases. We explored the possibility that Fms-like tyrosine kinase 3 (FLT3), a hematopoietic receptor-tyrosine kinase, is subject to this type of regulation. An underlying rationale was that the FLT3 gene is frequently mutated in Acute Myeloid Leukemia patients, and resulting oncogenic variants of FLT3 with 'internal tandem duplications (FLT3ITD)' drive production of reactive oxygen in leukemic cells. FLT3 was moderately activated by treatment of intact cells with hydrogen peroxide. Conversely, FLT3ITD signaling was attenuated by cell treatments with agents inhibiting formation of reactive oxygen species. FLT3 and FLT3ITD incorporated DCP-Bio1, a reagent specifically reacting with sulfenic acid residues. Mutation of FLT3ITD cysteines 695 and 790 reduced DCP-Bio1 incorporation, suggesting that these sites are subject to oxidative modification. Functional characterization of individual FLT3ITD cysteine-to-serine mutants of all 8 cytoplasmic cysteines revealed phenotypes in kinase activity, signal transduction and cell transformation. Replacement of cysteines 681, 694, 695, 807, 925, and 945 attenuated signaling and blocked FLT3ITD-mediated cell transformation, whereas mutation of cysteine 790 enhanced activity of both FLT3ITD and wild-type FLT3. These effects were not related to altered FLT3ITD dimerization, but likely caused by changed intramolecular interactions. The findings identify the functional relevance of all cytoplasmic FLT3ITD cysteines, and indicate the potential for redox regulation of this clinically important oncoprotein., (Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
13. The acetyltransferase GCN5 maintains ATRA-resistance in non-APL AML.
- Author
-
Kahl M, Brioli A, Bens M, Perner F, Kresinsky A, Schnetzke U, Hinze A, Sbirkov Y, Stengel S, Simonetti G, Martinelli G, Petrie K, Zelent A, Böhmer FD, Groth M, Ernst T, Heidel FH, Scholl S, Hochhaus A, and Schenk T
- Subjects
- Apoptosis, Bone Marrow metabolism, Cell Differentiation, Cell Line, Tumor, Cell Membrane metabolism, Epigenesis, Genetic, Genotype, HEK293 Cells, HL-60 Cells, Histone Demethylases antagonists & inhibitors, Histones chemistry, Humans, Leukocytes, Mononuclear cytology, Drug Resistance, Neoplasm, Leukemia, Myeloid, Acute drug therapy, Leukemia, Promyelocytic, Acute drug therapy, Tretinoin pharmacology, p300-CBP Transcription Factors metabolism
- Abstract
To date, only one subtype of acute myeloid leukemia (AML), acute promyelocytic leukemia (APL) can be effectively treated by differentiation therapy utilizing all-trans retinoic acid (ATRA). Non-APL AMLs are resistant to ATRA. Here we demonstrate that the acetyltransferase GCN5 contributes to ATRA resistance in non-APL AML via aberrant acetylation of histone 3 lysine 9 (H3K9ac) residues maintaining the expression of stemness and leukemia associated genes. We show that inhibition of GCN5 unlocks an ATRA-driven therapeutic response. This response is potentiated by coinhibition of the lysine demethylase LSD1, leading to differentiation in most non-APL AML. Induction of differentiation was not correlated to a specific AML subtype, cytogenetic, or mutational status. Our study shows a previously uncharacterized role of GCN5 in maintaining the immature state of leukemic blasts and identifies GCN5 as a therapeutic target in AML. The high efficacy of the combined epigenetic treatment with GCN5 and LSD1 inhibitors may enable the use of ATRA for differentiation therapy of non-APL AML. Furthermore, it supports a strategy of combined targeting of epigenetic factors to improve treatment, a concept potentially applicable for a broad range of malignancies.
- Published
- 2019
- Full Text
- View/download PDF
14. Lack of CD45 in FLT3-ITD mice results in a myeloproliferative phenotype, cortical porosity, and ectopic bone formation.
- Author
-
Kresinsky A, Schnöder TM, Jacobsen ID, Rauner M, Hofbauer LC, Ast V, König R, Hoffmann B, Svensson CM, Figge MT, Hilger I, Heidel FH, Böhmer FD, and Müller JP
- Subjects
- 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, Bone and Bones, Leukocyte Common Antigens genetics, Myeloproliferative Disorders genetics, Osteoporosis genetics, fms-Like Tyrosine Kinase 3 genetics
- 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.- Published
- 2019
- Full Text
- View/download PDF
15. Loss of DEP-1 (Ptprj) promotes myeloproliferative disease in FLT3 -ITD acute myeloid leukemia.
- Author
-
Kresinsky A, Bauer R, Schnöder TM, Berg T, Meyer D, Ast V, König R, Serve H, Heidel FH, Böhmer FD, and Müller JP
- Subjects
- Animals, Mice, Mice, Knockout, Receptor-Like Protein Tyrosine Phosphatases, Class 3 deficiency, Leukemia, Myeloid, Acute enzymology, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, fms-Like Tyrosine Kinase 3 metabolism
- Published
- 2018
- Full Text
- View/download PDF
16. Hyperglycaemia-induced methylglyoxal accumulation potentiates VEGF resistance of diabetic monocytes through the aberrant activation of tyrosine phosphatase SHP-2/SRC kinase signalling axis.
- Author
-
Dorenkamp M, Müller JP, Shanmuganathan KS, Schulten H, Müller N, Löffler I, Müller UA, Wolf G, Böhmer FD, Godfrey R, and Waltenberger J
- Subjects
- Animals, Chemotaxis, Humans, Mice, Monocytes drug effects, Hyperglycemia pathology, Monocytes pathology, Protein Tyrosine Phosphatase, Non-Receptor Type 11 metabolism, Pyruvaldehyde metabolism, Signal Transduction, Vascular Endothelial Growth Factor A metabolism, src-Family Kinases metabolism
- Abstract
Diabetes mellitus (DM) is a major cardiovascular risk factor contributing to cardiovascular complications by inducing vascular cell dysfunction. Monocyte dysfunction could contribute to impaired arteriogenesis response in DM patients. DM monocytes show blunted chemotactic responses to arteriogenic stimuli, a condition termed as vascular endothelial growth factor (VEGF) resistance. We hypothesize that methylglyoxal (MG), a glucose metabolite, induces monocyte dysfunction and aimed to elucidate the underlying molecular mechanisms. Human monocytes exposed to MG or monocytes from DM patients or mice (db/db) showed VEGF-resistance secondary to a pro-migratory phenotype. Mechanistically, DM conditions or MG exposure resulted in the upregulation of the expression of SHP-2 phosphatase. This led to the enhanced activity of SHP-2 and aided an interaction with SRC kinase. SHP-2 dephosphorylated the inhibitory phosphorylation site of SRC leading to its abnormal activation and phosphorylation of cytoskeletal protein, paxillin. We demonstrated that MG-induced molecular changes could be reversed by pharmacological inhibitors of SHP-2 and SRC and by genetic depletion of SHP-2. Finally, a SHP-2 inhibitor completely reversed the dysfunction of monocytes isolated from DM patients and db/db mice. In conclusion, we identified SHP-2 as a hitherto unknown target for improving monocyte function in diabetes. This opens novel perspectives for treating diabetic complications associated with impaired monocyte function.
- Published
- 2018
- Full Text
- View/download PDF
17. PTPRG and PTPRC modulate nilotinib response in chronic myeloid leukemia cells.
- Author
-
Drube J, Ernst T, Pfirrmann M, Albert BV, Drube S, Reich D, Kresinsky A, Halfter K, Sorio C, Fabisch C, Hochhaus A, and Böhmer FD
- Abstract
The introduction of second-generation tyrosine kinase inhibitors (TKIs) targeting the protein-tyrosine kinase (PTK) BCR-ABL1 has improved treatment response in chronic myeloid leukemia (CML). However, in some patients response still remains suboptimal. Protein-tyrosine phosphatases (PTPs) are natural counter-actors of PTK activity and can affect TKI sensitivity, but the impact of PTPs on treatment response to second-generation TKIs is unknown. We assessed the mRNA expression level of 38 PTPs in 66 newly diagnosed CML patients and analyzed the potential relation with treatment outcome after 9 months of nilotinib medication. A significantly positive association with response was observed for higher PTPN13, PTPRA, PTPRC (also known as CD45), PTPRG, and PTPRM expression. Selected PTPs were then subjected to a functional analysis in CML cell line models using PTP gene knockout by CRISPR/Cas9 technology or PTP overexpression. These analyses revealed PTPRG positively and PTPRC negatively modulating nilotinib response. Consistently, PTPRG negatively and PTPRC positively affected BCR-ABL1 dependent transformation. We identified BCR-ABL1 signaling events, which were affected by modulating PTP levels or nilotinib treatment in the same direction. In conclusion, the PTP status of CML cells is important for the response to second generation TKIs and may help in optimizing therapeutic strategies., Competing Interests: CONFLICTS OF INTEREST A.H. receives research support from Novartis, BMS, Pfizer, Incyte, and MSD. No potential conflict of interest is reported by the other authors.
- Published
- 2018
- Full Text
- View/download PDF
18. Nuclear membrane-localised NOX4D generates pro-survival ROS in FLT3-ITD-expressing AML.
- Author
-
Moloney JN, Jayavelu AK, Stanicka J, Roche SL, O'Brien RL, Scholl S, Böhmer FD, and Cotter TG
- Abstract
Internal tandem duplication of the juxtamembrane domain of FMS-like tyrosine kinase 3 (FLT3-ITD) is the most prevalent genetic aberration present in 20-30% of acute myeloid leukaemia (AML) cases and is associated with a poor prognosis. FLT3-ITD expressing cells express elevated levels of NADPH oxidase 4 (NOX4)-generated pro-survival hydrogen peroxide (H
2 O2 ) contributing to increased levels of DNA oxidation and double strand breaks. NOX4 is constitutively active and has been found to have various isoforms expressed at multiple locations within a cell. The purpose of this study was to investigate the expression, localisation and regulation of NOX4 28 kDa splice variant, NOX4D. NOX4D has previously been shown to localise to the nucleus and nucleolus in various cell types and is implicated in the generation of reactive oxygen species (ROS) and DNA damage. Here, we demonstrate that FLT3-ITD expressing-AML patient samples as well as -cell lines express the NOX4D isoform resulting in elevated H2 O2 levels compared to FLT3-WT expressing cells, as quantified by flow cytometry. Cell fractionation indicated that NOX4D is nuclear membrane-localised in FLT3-ITD expressing cells. Treatment of MV4-11 cells with receptor trafficking inhibitors, tunicamycin and brefeldin A, resulted in deglycosylation of NOX4 and NOX4D. Inhibition of the FLT3 receptor revealed that the FLT3-ITD oncogene is responsible for the production of NOX4D-generated H2 O2 in AML. We found that inhibition of the PI3K/AKT and STAT5 pathways resulted in down-regulation of NOX4D-generated pro-survival ROS. Taken together these findings indicate that nuclear membrane-localised NOX4D-generated pro-survival H2 O2 may be contributing to genetic instability in FLT3-ITD expressing AML., Competing Interests: CONFLICTS OF INTEREST The authors declare that they have no conflicts of interest.- Published
- 2017
- Full Text
- View/download PDF
19. Synergistic killing of FLT3ITD-positive AML cells by combined inhibition of tyrosine-kinase activity and N-glycosylation.
- Author
-
Tsitsipatis D, Jayavelu AK, Müller JP, Bauer R, Schmidt-Arras D, Mahboobi S, Schnöder TM, Heidel F, and Böhmer FD
- Subjects
- 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, Antineoplastic Agents pharmacology, Gene Duplication, Leukemia, Myeloid, Acute genetics, Protein Kinase Inhibitors pharmacology, Tandem Repeat Sequences, fms-Like Tyrosine Kinase 3 genetics
- 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.
- Published
- 2017
- Full Text
- View/download PDF
20. The protein-tyrosine phosphatase DEP-1 promotes migration and phagocytic activity of microglial cells in part through negative regulation of fyn tyrosine kinase.
- Author
-
Schneble N, Müller J, Kliche S, Bauer R, Wetzker R, Böhmer FD, Wang ZQ, and Müller JP
- Subjects
- Animals, Animals, Newborn, Cell Line, Transformed, Cell Movement genetics, Cells, Cultured, Cerebral Cortex cytology, Immunoprecipitation, Mice, Mice, Inbred C57BL, Mice, Knockout, Phagocytosis physiology, Proto-Oncogene Proteins c-fyn genetics, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Receptor-Like Protein Tyrosine Phosphatases, Class 3 genetics, Receptor-Like Protein Tyrosine Phosphatases, Class 3 metabolism, Cell Movement physiology, Gene Expression Regulation genetics, Microglia physiology, Phagocytosis genetics, Proto-Oncogene Proteins c-fyn metabolism
- Abstract
Microglia cells are brain macrophages whose proper functioning is essential for maintenance and repair processes of the central nervous system (CNS). Migration and phagocytosis are critical aspects of microglial activity. By using genetically modified cell lines and knockout mice we demonstrate here that the receptor protein-tyrosine phosphatase (PTP) DEP-1 (also known as PTPRJ or CD148) acts as a positive regulator of both processes in vitro and in vivo. Notably, reduced microglial migration was detectable in brains of Ptprj
-/- mice using a wounding assay. Mechanistically, density-enhanced phosphatase-1 (DEP-1) may in part function by inhibiting the activity of the Src family kinase Fyn. In the microglial cell line BV2 DEP-1 depletion by shRNA-mediated knockdown resulted in enhanced phosphorylation of the Fyn activating tyrosine (Tyr420 ) and elevated specific Fyn-kinase activity in immunoprecipitates. Moreover, Fyn mRNA and protein levels were reduced in DEP-1 deficient microglia cells. Consistent with a negative regulatory role of Fyn for microglial functions, which is inhibited by DEP-1, microglial cells from Fyn-/- mice exhibited elevated migration and phagocytosis. Enhanced microglia migration to a site of injury was also observed in Fyn-/- mice in vivo. Taken together our data revealed a previously unrecognized role of DEP-1 and suggest the existence of a potential DEP-1-Fyn axis in the regulation of microglial functions. GLIA 2017;65:416-428., (© 2016 Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
21. NOX-driven ROS formation in cell transformation of FLT3-ITD-positive AML.
- Author
-
Jayavelu AK, Moloney JN, Böhmer FD, and Cotter TG
- Subjects
- Animals, DNA Damage, Humans, Leukemia, Myeloid, Acute pathology, Oxidation-Reduction, Protein Tyrosine Phosphatases metabolism, Signal Transduction, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, NADPH Oxidases metabolism, Reactive Oxygen Species metabolism, Tandem Repeat Sequences, fms-Like Tyrosine Kinase 3 genetics
- Abstract
In different types of myeloid leukemia, increased formation of reactive oxygen species (ROS) has been noted and associated with aspects of cell transformation, including the promotion of leukemic cell proliferation and migration, as well as DNA damage and accumulation of mutations. Work reviewed in this article has revealed the involvement of NADPH oxidase (NOX)-derived ROS downstream of oncogenic protein-tyrosine kinases in both processes, and the related pathways have been partially identified. FMS-like tyrosine kinase 3 with internal tandem duplications (FLT3-ITD), an important oncoprotein in a subset of acute myeloid leukemias, causes activation of AKT and, subsequently, stabilization of p22
phox , a regulatory subunit for NOX1-4. This process is linked to ROS formation and DNA damage. Moreover, FLT3-ITD signaling through STAT5 enhances expression of NOX4, ROS formation, and inactivation of the protein-tyrosine phosphatase DEP-1/PTPRJ, a negative regulator of FLT3 signaling, by reversible oxidation of its catalytic cysteine residue. Genetic inactivation of NOX4 restores DEP-1 activity and attenuates cell transformation by FLT3-ITD in vitro and in vivo. Future work is required to further explore these mechanisms and their causal involvement in leukemic cell transformation, which may result in the identification of novel candidate targets for therapy., (Copyright © 2016 ISEH - International Society for Experimental Hematology. Published by Elsevier Inc. All rights reserved.)- Published
- 2016
- Full Text
- View/download PDF
22. Proteinase-activated receptor 2 (PAR2) in hepatic stellate cells - evidence for a role in hepatocellular carcinoma growth in vivo.
- Author
-
Mußbach F, Ungefroren H, Günther B, Katenkamp K, Henklein P, Westermann M, Settmacher U, Lenk L, Sebens S, Müller JP, Böhmer FD, and Kaufmann R
- Subjects
- Animals, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular metabolism, Cell Line, Tumor, Cell Movement, Hepatic Stellate Cells cytology, Humans, Liver Neoplasms genetics, Liver Neoplasms metabolism, Mice, Mice, SCID, Neoplasm Transplantation, Proteomics methods, RNA Interference, Signal Transduction, Angiogenesis Inducing Agents metabolism, Carcinoma, Hepatocellular pathology, Liver Neoplasms pathology, Receptor, PAR-2 genetics, Receptor, PAR-2 metabolism, Receptors, G-Protein-Coupled genetics, Receptors, G-Protein-Coupled metabolism
- Abstract
Background: Previous studies have established that proteinase-activated receptor 2 (PAR2) promotes migration and invasion of hepatocellular carcinoma (HCC) cells, suggesting a role in HCC progression. Here, we assessed the impact of PAR2 in HCC stromal cells on HCC growth using LX-2 hepatic stellate cells (HSCs) and Hep3B cells as model., Methods: PAR2 expression and function in LX-2 cells was analysed by RT-PCR, confocal immunofluorescence, electron microscopy, and [Ca(2+)]i measurements, respectively. The impact of LX-2-expressed PAR2 on tumour growth in vivo was monitored using HCC xenotransplantation experiments in SCID mice, in which HCC-like tumours were induced by coinjection of LX-2 cells and Hep3B cells. To characterise the effects of PAR2 activation in LX-2 cells, various signalling pathways were analysed by immunoblotting and proteome profiler arrays., Results: Following verification of functional PAR2 expression in LX-2 cells, in vivo studies showed that these cells promoted tumour growth and angiogenesis of HCC xenografts in mice. These effects were significantly reduced when F2RL1 (encoding PAR2) was downregulated by RNA interference (RNAi). In vitro studies confirmed these results demonstrating RNAi mediated inhibition of PAR2 attenuated Smad2/3 activation in response to TGF-β1 stimulation in LX-2 cells and blocked the pro-mitotic effect of LX-2 derived conditioned medium on Hep3B cells. Furthermore, PAR2 stimulation with trypsin or a PAR2-selective activating peptide (PAR2-AP) led to activation of different intracellular signalling pathways, an increased secretion of pro-angiogenic and pro-mitotic factors and proteinases, and an enhanced migration rate across a collagen-coated membrane barrier. Silencing F2RL1 by RNAi or pharmacological inhibition of Src, hepatocyte growth factor receptor (Met), platelet-derived growth factor receptor (PDGFR), p42/p44 mitogen activated protein kinase (MAPK) or matrix-metalloproteinases (MMPs) blocked PAR2-AP-induced migration., Conclusion: PAR2 in HSCs plays a crucial role in promoting HCC growth presumably by mediating migration and secretion of pro-angiogenic and pro-mitotic factors. Therefore, PAR2 in stromal HSCs may have relevance as a therapeutic target of HCC.
- Published
- 2016
- Full Text
- View/download PDF
23. NOX4-driven ROS formation mediates PTP inactivation and cell transformation in FLT3ITD-positive AML cells.
- Author
-
Jayavelu AK, Müller JP, Bauer R, Böhmer SA, Lässig J, Cerny-Reiterer S, Sperr WR, Valent P, Maurer B, Moriggl R, Schröder K, Shah AM, Fischer M, Scholl S, Barth J, Oellerich T, Berg T, Serve H, Frey S, Fischer T, Heidel FH, and Böhmer FD
- Subjects
- Animals, Cells, Cultured, Humans, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, NADPH Oxidase 4, NADPH Oxidases genetics, Receptor-Like Protein Tyrosine Phosphatases, Class 3 analysis, Tandem Repeat Sequences, fms-Like Tyrosine Kinase 3 analysis, Cell Transformation, Neoplastic, Leukemia, Myeloid, Acute pathology, NADPH Oxidases physiology, Protein Tyrosine Phosphatases metabolism, Reactive Oxygen Species metabolism, fms-Like Tyrosine Kinase 3 physiology
- Abstract
Activating mutations of FMS-like tyrosine kinase 3 (FLT3), notably internal tandem duplications (ITDs), are associated with a grave prognosis in acute myeloid leukemia (AML). Transforming FLT3ITD signal transduction causes formation of reactive oxygen species (ROS) and inactivation of the protein-tyrosine phosphatase (PTP) DEP-1/PTPRJ, a negative regulator of FLT3 signaling. Here we addressed the underlying mechanisms and biological consequences. NADPH oxidase 4 (NOX4) messenger RNA and protein expression was found to be elevated in FLT3ITD-positive cells and to depend on FLT3ITD signaling and STAT5-mediated activation of the NOX4 promoter. NOX4 knockdown reduced ROS levels, restored DEP-1 PTP activity and attenuated FLT3ITD-driven transformation. Moreover, Nox4 knockout (Nox4(-/-)) murine hematopoietic progenitor cells were refractory to FLT3ITD-mediated transformation in vitro. Development of a myeloproliferative-like disease (MPD) caused by FLT3ITD-transformed 32D cells in C3H/HeJ mice, and of a leukemia-like disease in mice transplanted with MLL-AF9/ FLT3ITD-transformed murine hematopoietic stem cells were strongly attenuated by NOX4 downregulation. NOX4-targeting compounds were found to counteract proliferation of FLT3ITD-positive AML blasts and MPD development in mice. These findings reveal a previously unrecognized mechanism of oncoprotein-driven PTP oxidation, and suggest that interference with FLT3ITD-STAT5-NOX4-mediated overproduction of ROS and PTP inactivation may have therapeutic potential in a subset of AML.
- Published
- 2016
- Full Text
- View/download PDF
24. In Situ Proximity Ligation Assay (In Situ PLA) to Assess PTP-Protein Interactions.
- Author
-
Koch S, Helbing I, Böhmer SA, Hayashi M, Claesson-Welsh L, Söderberg O, and Böhmer FD
- Subjects
- Animals, COS Cells, Cell Line, Chlorocebus aethiops, HEK293 Cells, Human Umbilical Vein Endothelial Cells, Humans, Optical Imaging methods, Protein Interaction Maps, Receptor, TIE-2 metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism, fms-Like Tyrosine Kinase 3 metabolism, Protein Interaction Mapping methods, Protein Tyrosine Phosphatases metabolism
- Abstract
Spatiotemporal aspects of protein-tyrosine phosphatase (PTP) activity and interaction partners for many PTPs are elusive. We describe here an elegant and relatively simple method, in situ proximity ligation assay (in situ PLA), which can be used to address these issues. The possibility to detect endogenous unmodified proteins in situ and to visualize individual interactions with spatial resolution is the major advantage of this technique. We provide protocols suitable to monitor association of the transmembrane PTPs PTPRJ/DEP-1/CD148 and PTPRB/VE-PTP with their substrates, the receptor tyrosine kinases FMS-like tyrosine kinase 3 (FLT3/CD135), and Tie2 and vascular endothelial growth factor receptor 2 (VEGFR2), respectively. Detailed description of method development and reagents as well as highlighting of critical factors will enable the reader to apply the method successfully to other PTP-protein interactions.
- Published
- 2016
- Full Text
- View/download PDF
25. High REDOX RESPONSIVE TRANSCRIPTION FACTOR1 Levels Result in Accumulation of Reactive Oxygen Species in Arabidopsis thaliana Shoots and Roots.
- Author
-
Matsuo M, Johnson JM, Hieno A, Tokizawa M, Nomoto M, Tada Y, Godfrey R, Obokata J, Sherameti I, Yamamoto YY, Böhmer FD, and Oelmüller R
- Subjects
- Acetylcysteine pharmacology, Alternaria physiology, Arabidopsis microbiology, Arabidopsis Proteins genetics, Base Sequence, Cell Death drug effects, Ditiocarb pharmacology, Gene Expression Regulation, Plant drug effects, Genes, Plant, Light, Molecular Sequence Data, Pathogen-Associated Molecular Pattern Molecules metabolism, Phenotype, Plant Diseases genetics, Plant Diseases microbiology, Plant Roots drug effects, Plant Roots genetics, Plant Roots radiation effects, Plant Shoots drug effects, Plant Shoots radiation effects, Plants, Genetically Modified, Promoter Regions, Genetic genetics, Seedlings drug effects, Seedlings genetics, Seedlings microbiology, Seedlings radiation effects, Stress, Physiological drug effects, Stress, Physiological genetics, Stress, Physiological radiation effects, Transcription Factors genetics, Arabidopsis metabolism, Arabidopsis Proteins metabolism, Plant Roots metabolism, Plant Shoots metabolism, Reactive Oxygen Species metabolism, Transcription Factors metabolism
- Abstract
Redox Responsive Transcription Factor1 (RRTF1) in Arabidopsis is rapidly and transiently upregulated by H2O2, as well as biotic- and abiotic-induced redox signals. RRTF1 is highly conserved in angiosperms, but its physiological role remains elusive. Here we show that inactivation of RRTF1 restricts and overexpression promotes reactive oxygen species (ROS) accumulation in response to stress. Transgenic lines overexpressing RRTF1 are impaired in root and shoot development, light sensitive, and susceptible to Alternaria brassicae infection. These symptoms are diminished by the beneficial root endophyte Piriformospora indica, which reduces ROS accumulation locally in roots and systemically in shoots, and by antioxidants and ROS inhibitors that scavenge ROS. More than 800 genes were detected in mature leaves and seedlings of transgenic lines overexpressing RRTF1; ∼ 40% of them have stress-, redox-, ROS-regulated-, ROS-scavenging-, defense-, cell death- and senescence-related functions. Bioinformatic analyses and in vitro DNA binding assays demonstrate that RRTF1 binds to GCC-box-like sequences in the promoter of RRTF1-responsive genes. Upregulation of RRTF1 by stress stimuli and H2O2 requires WRKY18/40/60. RRTF1 is co-regulated with the phylogenetically related RAP2.6, which contains a GCC-box-like sequence in its promoter, but transgenic lines overexpressing RAP2.6 do not accumulate higher ROS levels. RRTF1 also stimulates systemic ROS accumulation in distal non-stressed leaves. We conclude that the elevated levels of the highly conserved RRTF1 induce ROS accumulation in response to ROS and ROS-producing abiotic and biotic stress signals., (Copyright © 2015 The Author. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
26. Proteinase-activated receptor 1- and 4-promoted migration of Hep3B hepatocellular carcinoma cells depends on ROS formation and RTK transactivation.
- Author
-
Mußbach F, Henklein P, Westermann M, Settmacher U, Böhmer FD, and Kaufmann R
- Subjects
- Blotting, Western, Cell Line, Tumor, Cell Movement drug effects, Gene Expression Regulation, Neoplastic, Humans, Receptor Protein-Tyrosine Kinases metabolism, Receptor, PAR-1 metabolism, Signal Transduction, Apoptosis Regulatory Proteins metabolism, Carcinoma, Hepatocellular metabolism, Liver Neoplasms metabolism, Oligopeptides metabolism, Reactive Oxygen Species metabolism, Receptor Protein-Tyrosine Kinases genetics, Transcriptional Activation
- Abstract
Purpose: There is growing evidence for a role of proteinase-activated receptors (PARs), a subfamily of G protein-coupled receptors, in cancer. We have previously shown that PAR1 and PAR4 are able to promote the migration of hepatocellular carcinoma (HCC) cells suggesting a function in HCC progression. In this study, we assessed the underlying signalling mechanisms., Methods: Using Hep3B liver carcinoma cells, RTK activation was assessed by Western blot employing phospho-RTK specific antibodies, ROS level were estimated by H2DCF-DA using confocal laser scanning microscopy, and measurement of PTP activity was performed in cell lysates using 6,8-difluoro-4-methylumbelliferyl phosphate (DiFMUP) as a substrate., Results: Thrombin, the PAR1 selective agonist peptide TFLLRN-NH2 (PAR1-AP), and the PAR4 selective agonist peptide, AYPGKF-NH2 (PAR4-AP), induced a significant increase in Hep3B cell migration that could be blocked by inhibitors targeting formation of reactive oxygen species (ROS), or activation of hepatocyte-growth factor receptor (Met), or platelet-derived growth factor receptor (PDGFR), respectively. The involvement of these intracellular effectors in PAR1/4-initiated migratory signalling was further supported by the findings that individual stimulation of Hep3B cells with the PAR1-AP and the PAR4-AP induced an increase in ROS production and the transactivation of Met and PDGFR. In addition, PAR1- and PAR4-mediated inhibition of total PTP activity and specifically PTP1B. ROS inhibition by N-acetyl-L-cysteine prevented the inhibition of PTP1B phosphatase activity induced by PAR1-AP and the PAR4-AP, but had no effect on PAR1/4-mediated activation of Met and PDGFR in Hep3B cells., Conclusions: Collectively, our data indicate that PAR1 and PAR4 activate common promigratory signalling pathways in Hep3B liver carcinoma cells including activation of the receptor tyrosine kinases Met and PDGFR, the formation of ROS and the inactivation of PTP1B. However, PAR1/4-triggered Met and PDGFR transactivation seem to be mediated independently from the ROS-PTP1B signalling module.
- Published
- 2015
- Full Text
- View/download PDF
27. Deficiency of the protein-tyrosine phosphatase DEP-1/PTPRJ promotes matrix metalloproteinase-9 expression in meningioma cells.
- Author
-
Petermann A, Stampnik Y, Cui Y, Morrison H, Pachow D, Kliese N, Mawrin C, and Böhmer FD
- Subjects
- Analysis of Variance, Cell Line, Tumor, Cytokines metabolism, Gene Expression Regulation, Neoplastic drug effects, Glioma pathology, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Humans, Intercellular Signaling Peptides and Proteins pharmacology, Matrix Metalloproteinase 2 genetics, Matrix Metalloproteinase 2 metabolism, Matrix Metalloproteinase 9 genetics, Neurofibromatosis 2 genetics, Neurofibromatosis 2 metabolism, RNA, Messenger metabolism, Receptor-Like Protein Tyrosine Phosphatases, Class 3 deficiency, Receptor-Like Protein Tyrosine Phosphatases, Class 3 genetics, Transfection, Gene Expression Regulation, Neoplastic genetics, Glioma metabolism, Matrix Metalloproteinase 9 metabolism
- Abstract
Brain-invasive growth of a subset of meningiomas is associated with less favorable prognosis. The molecular mechanisms causing invasiveness are only partially understood, however, the expression of matrix metalloproteinases (MMPs) has been identified as a contributing factor. We have previously found that loss of density enhanced phosphatase-1 (DEP-1, also designated PTPRJ), a transmembrane protein-tyrosine phosphatase, promotes meningioma cell motility and invasive growth in an orthotopic xenotransplantation model. We have now analyzed potential alterations of the expression of genes involved in motility control, caused by DEP-1 loss in meningioma cell lines. DEP-1 depleted cells exhibited increased expression of mRNA encoding MMP-9, and the growth factors EGF and FGF-2. The increase of MMP-9 expression in DEP-1 depleted cells was also readily detectable at the protein level by zymography. MMP-9 upregulation was sensitive to chemical inhibitors of growth factor signal transduction. Conversely, MMP-9 mRNA levels could be stimulated with growth factors (e.g. EGF) and inflammatory cytokines (e.g. TNFα). Increase of MMP-9 expression by DEP-1 depletion, or growth factor/cytokine stimulation qualitatively correlated with increased invasiveness in vitro scored as transmigration through matrigel-coated membranes. The studies suggest induction of MMP-9 expression promoted by DEP-1 deficiency, or potentially by growth factors and inflammatory cytokines, as a mechanism contributing to meningioma brain invasiveness.
- Published
- 2015
- Full Text
- View/download PDF
28. Enhanced insulin signaling in density-enhanced phosphatase-1 (DEP-1) knockout mice.
- Author
-
Krüger J, Brachs S, Trappiel M, Kintscher U, Meyborg H, Wellnhofer E, Thöne-Reineke C, Stawowy P, Östman A, Birkenfeld AL, Böhmer FD, and Kappert K
- Abstract
Objective: Insulin resistance can be triggered by enhanced dephosphorylation of the insulin receptor or downstream components in the insulin signaling cascade through protein tyrosine phosphatases (PTPs). Downregulating density-enhanced phosphatase-1 (DEP-1) resulted in an improved metabolic status in previous analyses. This phenotype was primarily caused by hepatic DEP-1 reduction., Methods: Here we further elucidated the role of DEP-1 in glucose homeostasis by employing a conventional knockout model to explore the specific contribution of DEP-1 in metabolic tissues. Ptprj (-/-) (DEP-1 deficient) and wild-type C57BL/6 mice were fed a low-fat or high-fat diet. Metabolic phenotyping was combined with analyses of phosphorylation patterns of insulin signaling components. Additionally, experiments with skeletal muscle cells and muscle tissue were performed to assess the role of DEP-1 for glucose uptake., Results: High-fat diet fed-Ptprj (-/-) mice displayed enhanced insulin sensitivity and improved glucose tolerance. Furthermore, leptin levels and blood pressure were reduced in Ptprj (-/-) mice. DEP-1 deficiency resulted in increased phosphorylation of components of the insulin signaling cascade in liver, skeletal muscle and adipose tissue after insulin challenge. The beneficial effect on glucose homeostasis in vivo was corroborated by increased glucose uptake in skeletal muscle cells in which DEP-1 was downregulated, and in skeletal muscle of Ptprj (-/-) mice., Conclusion: Together, these data establish DEP-1 as novel negative regulator of insulin signaling.
- Published
- 2015
- Full Text
- View/download PDF
29. Re-evaluation of cytostatic therapies for meningiomas in vitro.
- Author
-
Wilisch-Neumann A, Pachow D, Wallesch M, Petermann A, Böhmer FD, Kirches E, and Mawrin C
- Subjects
- Apoptosis, Cell Line, Tumor, DNA Methylation, DNA Modification Methylases genetics, DNA Repair Enzymes genetics, Dacarbazine analogs & derivatives, Dacarbazine pharmacology, Drug Resistance, Neoplasm, Drug Screening Assays, Antitumor, Erlotinib Hydrochloride, Humans, Losartan pharmacology, Metformin pharmacology, Mifepristone pharmacology, Neurofibromin 2 genetics, Promoter Regions, Genetic, Quinazolines pharmacology, Radiation Tolerance, Tamoxifen pharmacology, Temozolomide, Tumor Suppressor Proteins genetics, Verapamil pharmacology, Antineoplastic Agents pharmacology, Hydroxyurea pharmacology, Meningeal Neoplasms drug therapy, Meningioma drug therapy
- Abstract
Purpose: The purpose was to re-evaluate in cell culture models the therapeutic usefulness of some discussed chemotherapies or targeted therapies for meningiomas with a special emphasis on the role of the neurofibromatosis type 2 (NF2) tumor suppressor, which had been neglected so far. In addition, the study intended to evaluate a potential benefit from a treatment with drugs which are well established in other fields of medicine and have been linked recently with tumor disease by epidemiological studies., Methods: Meningioma cell lines corresponding to various subtypes and pairs of syngenic meningioma cell lines with or without shRNA-induced NF2 knockdown were analyzed for their dose-dependent response to the drugs in microtiter tetrazolium assays, BrdU assays and for selected cases in ELISAs measuring nucleosome liberation to specifically separate cell death from pure inhibition of cell proliferation., Results: We confirmed a moderate efficacy of hydroxyurea (HU) in clinically relevant concentrations. Under appropriate dosing, we neither detected major responses to the alkylating compound temozolomide nor to various drugs targeting membrane receptors or enzymes (tamoxifen, erlotinib, mifepristone, losartan, metformin and verapamil). Only concentrations far beyond achievable serum levels generated significant effects with the exception of losartan, which showed no effects at all. Chemosensitivity varied markedly among meningioma cell lines. Importantly, cells with NF2 loss exhibited a significantly higher induction of cell death by HU., Conclusions: Alternative chemotherapeutic or targeted approaches besides HU have still to be evaluated in further studies, and the role of NF2 must be taken into account.
- Published
- 2014
- Full Text
- View/download PDF
30. Protein tyrosine phosphatases as wardens of STAT signaling.
- Author
-
Böhmer FD and Friedrich K
- Abstract
Signaling by signal transducers and activators of transcription (STATs) is controlled at many levels of the signaling cascade. Protein tyrosine phosphatases (PTPs) regulate STAT activation at several layers, including direct pSTAT dephosphorylation in both cytoplasm and nucleus. Despite the importance of this regulation mode, many aspects are still incompletely understood, e.g., the identity of PTPs acting on certain members of the STAT family. After a brief introduction into the STAT and PTP families, we discuss here the current knowledge on PTP mediated regulation of STAT activity, focusing on the interaction of individual STATs with specific PTPs. Finally, we highlight open questions and propose important tasks of future research.
- Published
- 2014
- Full Text
- View/download PDF
31. A genome-wide RNAi screen identifies proteins modulating aberrant FLT3-ITD signaling.
- Author
-
Caldarelli A, Müller JP, Paskowski-Rogacz M, Herrmann K, Bauer R, Koch S, Heninger AK, Krastev D, Ding L, Kasper S, Fischer T, Brodhun M, Böhmer FD, and Buchholz F
- Subjects
- Animals, Base Sequence, DNA Primers, HEK293 Cells, Humans, Male, Mice, Mice, Inbred C3H, Real-Time Polymerase Chain Reaction, STAT5 Transcription Factor metabolism, Genome, Proteins physiology, RNA Interference, Signal Transduction physiology, fms-Like Tyrosine Kinase 3 metabolism
- Abstract
Fms-like tyrosine kinase-3 is a commonly mutated gene in acute myeloid leukemia, with about one-third of patients carrying an internal-tandem duplication of the juxtamembrane domain in the receptor (FLT3-ITD). FLT3-ITD exhibits altered signaling quality, including aberrant activation of STAT5. To identify genes affecting FLT3-ITD-mediated STAT5 signaling, we performed an esiRNA-based RNAi screen utilizing a STAT5-driven reporter assay. Knockdowns that caused reduced FLT3-ITD-mediated STAT5 signaling were enriched for genes encoding proteins involved in protein secretion and intracellular protein transport, indicating that modulation of protein transport processes could potentially be used to reduce constitutive STAT5 signaling in FLT3-ITD-positive cells. The relevance of KDELR1, a component involved in the Golgi-ER retrograde transport, was further analyzed. In FLT3-ITD-expressing leukemic MV4-11 cells, downregulation of KDELR1 resulted in reduced STAT5 activation, proliferation and colony-forming capacity. Stable shRNA-mediated depletion of KDELR1 in FLT3-ITD-expressing 32D cells likewise resulted in reduced STAT5 signaling and cell proliferation. Importantly, these cells also showed a reduced capacity to generate a leukemia-like disease in syngeneic C3H/HeJ mice. Together our data suggest intracellular protein transport as a potential target for FLT3-ITD driven leukemias, with KDELR1 emerging as a positive modulator of oncogenic FLT3-ITD activity.
- Published
- 2013
- Full Text
- View/download PDF
32. A quantitative assessment of costimulation and phosphatase activity on microclusters in early T cell signaling.
- Author
-
Witsenburg JJ, Glauner H, Müller JP, Groenewoud JM, Roth G, Böhmer FD, Adjobo-Hermans MJ, and Brock R
- Subjects
- CD28 Antigens metabolism, Costimulatory and Inhibitory T-Cell Receptors metabolism, Gene Knockdown Techniques, Humans, Jurkat Cells, Phospholipase C gamma metabolism, Phosphorylation, Protein Tyrosine Phosphatase, Non-Receptor Type 11 genetics, Receptors, Antigen, T-Cell metabolism, T-Lymphocytes metabolism, Tyrosine metabolism, Costimulatory and Inhibitory T-Cell Receptors physiology, Phosphoric Monoester Hydrolases metabolism, Receptors, Antigen, T-Cell physiology, Signal Transduction, T-Lymphocytes physiology
- Abstract
T cell signaling is triggered through stimulation of the T cell receptor and costimulatory receptors. Receptor activation leads to the formation of membrane-proximal protein microclusters. These clusters undergo tyrosine phosphorylation and organize multiprotein complexes thereby acting as molecular signaling platforms. Little is known about how the quantity and phosphorylation levels of microclusters are affected by costimulatory signals and the activity of specific signaling proteins. We combined micrometer-sized, microcontact printed, striped patterns of different stimuli and simultaneous analysis of different cell strains with image processing protocols to address this problem. First, we validated the stimulation protocol by showing that high expression levels CD28 result in increased cell spreading. Subsequently, we addressed the role of costimulation and a specific phosphotyrosine phosphatase in cluster formation by including a SHP2 knock-down strain in our system. Distinguishing cell strains using carboxyfluorescein succinimidyl ester enabled a comparison within single samples. SHP2 exerted its effect by lowering phosphorylation levels of individual clusters while CD28 costimulation mainly increased the number of signaling clusters and cell spreading. These effects were observed for general tyrosine phosphorylation of clusters and for phosphorylated PLCγ1. Our analysis enables a clear distinction between factors determining the number of microclusters and those that act on these signaling platforms.
- Published
- 2013
- Full Text
- View/download PDF
33. Features of Ras activation by a mislocalized oncogenic tyrosine kinase: FLT3 ITD signals through K-Ras at the plasma membrane of acute myeloid leukemia cells.
- Author
-
Köthe S, Müller JP, Böhmer SA, Tschongov T, Fricke M, Koch S, Thiede C, Requardt RP, Rubio I, and Böhmer FD
- Subjects
- Animals, Cell Growth Processes physiology, Cell Membrane genetics, Cell Membrane metabolism, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Genes, ras, Humans, Leukemia, Myeloid, Acute genetics, Mice, Phosphorylation, Signal Transduction, Tandem Repeat Sequences, Tumor Cells, Cultured, fms-Like Tyrosine Kinase 3 genetics, ras Proteins genetics, Leukemia, Myeloid, Acute metabolism, fms-Like Tyrosine Kinase 3 metabolism, ras Proteins metabolism
- Abstract
FMS-like tyrosine kinase 3 with internal tandem duplication (FLT3 ITD) is an important oncoprotein in acute myeloid leukemia (AML). Owing to its constitutive kinase activity FLT3 ITD partially accumulates at endomembranes, a feature shared with other disease-associated, mutated receptor tyrosine kinases. Because Ras proteins also transit through endomembranes we have investigated the possible existence of an intracellular FLT3-ITD/Ras signaling pathway by comparing Ras signaling of FLT3 ITD with that of wild-type FLT3. Ligand stimulation activated both K- and N-Ras in cells expressing wild-type FLT3. Live-cell Ras-GTP imaging revealed ligand-induced Ras activation at the plasma membrane (PM). FLT3-ITD-dependent constitutive activation of K-Ras and N-Ras was also observed primarily at the PM, supporting the view that the PM-resident pool of FLT3 ITD engaged the Ras/Erk pathway in AML cells. Accordingly, specific interference with FLT3-ITD/Ras signaling at the PM using PM-restricted dominant negative K-RasS17N potently inhibited cell proliferation and promoted apoptosis. In conclusion, Ras signaling is crucial for FLT3-ITD-dependent cell transformation and FLT3 ITD addresses PM-bound Ras despite its pronounced mislocalization to endomembranes.
- Published
- 2013
- Full Text
- View/download PDF
34. The integrin inhibitor cilengitide affects meningioma cell motility and invasion.
- Author
-
Wilisch-Neumann A, Kliese N, Pachow D, Schneider T, Warnke JP, Braunsdorf WE, Böhmer FD, Hass P, Pasemann D, Helbing C, Kirches E, and Mawrin C
- Subjects
- Animals, Cell Cycle drug effects, Cell Line, Tumor, Cell Survival drug effects, Disease Models, Animal, Humans, Immunohistochemistry, Integrins metabolism, Meningeal Neoplasms drug therapy, Meningeal Neoplasms mortality, Meningeal Neoplasms radiotherapy, Meningioma drug therapy, Meningioma mortality, Meningioma radiotherapy, Mice, Neoplasm Invasiveness, Neurofibromin 2 genetics, Neurofibromin 2 metabolism, Receptors, Vitronectin metabolism, Snake Venoms administration & dosage, Tumor Burden drug effects, Cell Movement drug effects, Integrins antagonists & inhibitors, Meningeal Neoplasms metabolism, Meningeal Neoplasms pathology, Meningioma metabolism, Meningioma pathology, Snake Venoms pharmacology
- Abstract
Purpose: Meningiomas are frequent intracranial or spinal neoplasms, which recur frequently and can show aggressive clinical behaviour. We elucidated the impact of the integrin inhibitor cilengitide on migration, proliferation, and radiosensitization of meningioma cells., Experimental Design: We analyzed integrin expression in tissue microarrays of human meningiomas and the antimeningioma properties of cilengitide in cell cultures, subcutaneous and intracranial nude mouse models by measuring tumor volumes and survival times., Results: αvβ5 was the predominantly expressed integrin heterodimer in meningiomas, whereas αvβ3 was mainly detected in tumor blood vessels. Application of up to 100 μg/mL cilengitide resulted in only mildly reduced proliferation/survival of meningioma cell lines. Effects on cell survival could be enhanced by irradiation. One μg/mL cilengitide was sufficient to significantly inhibit meningioma cell migration and invasion in vitro. A daily dosage of 75 mg/kg did neither affect tumor volumes nor overall survival (P = 0.813, log-rank test), but suppressed brain invasion in a significant fraction of treated animals. A combination of 75 mg/kg cilengitide daily and irradiation (2 × 5 Gy) led to a 67% reduction of MRI-estimated tumor volumes in the intracranial model (P < 0.01), whereas the corresponding reduction reached by irradiation alone was only 55% (P < 0.05)., Conclusions: These data show that a monotherapy with cilengitide is not likely to achieve major responses in rapidly growing malignant meningiomas, although brain invasion may be reduced because of the strong antimigratory properties of the drug. The combination with radiotherapy warrants further attention., (©2013 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
35. Ras palmitoylation is necessary for N-Ras activation and signal propagation in growth factor signalling.
- Author
-
Song SP, Hennig A, Schubert K, Markwart R, Schmidt P, Prior IA, Böhmer FD, and Rubio I
- Subjects
- Amino Acid Substitution, Animals, Cell Line, Cells, Cultured, Chlorocebus aethiops, Embryo, Mammalian cytology, Enzyme Activation, GTP Phosphohydrolases genetics, Humans, Lipoylation, Membrane Proteins agonists, Membrane Proteins genetics, Mice, Mice, Knockout, Mutant Proteins agonists, Mutant Proteins metabolism, Protein Processing, Post-Translational, Protein Transport, Recombinant Proteins agonists, Recombinant Proteins metabolism, ras Proteins genetics, Cell Membrane metabolism, Down-Regulation, Epidermal Growth Factor metabolism, GTP Phosphohydrolases metabolism, Membrane Proteins metabolism, Palmitic Acid metabolism, Signal Transduction, ras Proteins metabolism
- Abstract
Ras GTPases undergo post-translational modifications that govern their subcellular trafficking and localization. In particular, palmitoylation of the Golgi tags N-Ras and H-Ras for exocytotic transport and residency at the PM (plasma membrane). Following depalmitoylation, PM-Ras redistributes to all subcellular membranes causing an accumulation of palmitate-free Ras at endomembranes, including the Golgi and endoplasmic reticulum. Palmitoylation is unanimously regarded as a critical modification at the crossroads of Ras activity and trafficking control, but its precise relevance to native wild-type Ras function in growth factor signalling is unknown. We show in the present study by use of palmitoylation-deficient N-Ras mutants and via the analysis of palmitate content of agonist-activated GTP-loaded N-Ras that only palmitoylated N-Ras becomes activated by agonists. In line with an essential role of palmitoylation in Ras activation, dominant-negative RasS17N loses its blocking potency if rendered devoid of palmitoylation. Live-cell Ras-GTP imaging shows that N-Ras activation proceeds only at the PM, consistent with activated N-Ras-GTP being palmitoylated. Finally, palmitoylation-deficient N-Ras does not sustain EGF (epidermal growth factor) or serum-elicited mitogenic signalling, confirming that palmitoylation is essential for signal transduction by N-Ras. These findings document that N-Ras activation proceeds at the PM and suggest that depalmitoylation, by removing Ras from the PM, may contribute to the shutdown of Ras signalling.
- Published
- 2013
- Full Text
- View/download PDF
36. Association of the protein-tyrosine phosphatase DEP-1 with its substrate FLT3 visualized by in situ proximity ligation assay.
- Author
-
Böhmer SA, Weibrecht I, Söderberg O, and Böhmer FD
- Subjects
- Animals, Apoptosis, COS Cells, Cell Proliferation, Chlorocebus aethiops, Fluorescent Antibody Technique, Indirect, Humans, Kinetics, Membrane Proteins physiology, Microscopy, Fluorescence, Oxidation-Reduction, Protein Interaction Mapping, Receptor-Like Protein Tyrosine Phosphatases, Class 3 metabolism, Signal Transduction, fms-Like Tyrosine Kinase 3 antagonists & inhibitors, fms-Like Tyrosine Kinase 3 metabolism
- Abstract
Protein-tyrosine phosphatases (PTPs) are important regulators of signal transduction processes. Essential for the functional characterization of PTPs is the identification of their physiological substrates, and an important step towards this goal is the demonstration of a physical interaction. The association of PTPs with their cellular substrates is, however, often transient and difficult to detect with unmodified proteins at endogenous levels. Density-enhanced phosphatase-1 (DEP-1/PTPRJ) is a regulator of hematopoietic cell functions, and a candidate tumor suppressor. However, association of DEP-1 with any of its proposed substrates at endogenous levels has not yet been shown. We have previously obtained functional and biochemical evidence for a direct interaction of DEP-1 with the hematopoietic receptor-tyrosine kinase Fms-like tyrosine kinase-3 (FLT3). In the current study we have used the method of in situ proximity ligation assay (in situ PLA) to validate this interaction at endogenous levels, and to further characterize it. In situ PLA readily detected association of endogenous DEP-1 and FLT3 in the human acute monocytic leukemia cell line THP-1, which was enhanced by FLT3 ligand (FL) stimulation in a time-dependent manner. Association peaked between 10 and 20 min of stimulation and returned to basal levels at 30 min. This time course was similar to the time course of FLT3 autophosphorylation. FLT3 kinase inhibition and DEP-1 oxidation abrogated association. Consistent with a functional role of DEP-1-FLT3 interaction, stable knockdown of DEP-1 in THP-1 cells enhanced FL-induced ERK1/2 activation. These findings support that FLT3 is a bona fide substrate of DEP-1 and that interaction occurs mainly via an enzyme-substrate complex formation triggered by FLT3 ligand stimulation.
- Published
- 2013
- Full Text
- View/download PDF
37. Knockout of Density-Enhanced Phosphatase-1 impairs cerebrovascular reserve capacity in an arteriogenesis model in mice.
- Author
-
Hackbusch D, Dülsner A, Gatzke N, Krüger J, Hillmeister P, Nagorka S, Blaschke F, Ritter Z, Thöne-Reineke C, Böhmer FD, Buschmann I, and Kappert K
- Subjects
- Animals, Becaplermin, Brain blood supply, Brain physiology, Carotid Artery, Common growth & development, Carotid Artery, Common surgery, Cells, Cultured, Circle of Willis growth & development, Circle of Willis surgery, Humans, Mice, Mice, Knockout, Proto-Oncogene Proteins c-sis metabolism, Receptor-Like Protein Tyrosine Phosphatases, Class 3 metabolism, Signal Transduction, Gene Expression Regulation, Neovascularization, Physiologic genetics, Proto-Oncogene Proteins c-sis biosynthesis, Receptor-Like Protein Tyrosine Phosphatases, Class 3 genetics
- Abstract
Collateral growth, arteriogenesis, represents a proliferative mechanism involving endothelial cells, smooth muscle cells, and monocytes/macrophages. Here we investigated the role of Density-Enhanced Phosphatase-1 (DEP-1) in arteriogenesis in vivo, a protein-tyrosine-phosphatase that has controversially been discussed with regard to vascular cell biology. Wild-type C57BL/6 mice subjected to permanent left common carotid artery occlusion (CCAO) developed a significant diameter increase in distinct arteries of the circle of Willis, especially in the anterior cerebral artery. Analyzing the impact of loss of DEP-1 function, induction of collateralization was quantified after CCAO and hindlimb femoral artery ligation comparing wild-type and DEP-1(-/-) mice. Both cerebral collateralization assessed by latex perfusion and peripheral vessel growth in the femoral artery determined by microsphere perfusion and micro-CT analysis were not altered in DEP-1(-/-) compared to wild-type mice. Cerebrovascular reserve capacity, however, was significantly impaired in DEP-1(-/-) mice. Cerebrovascular transcriptional analysis of proarteriogenic growth factors and receptors showed specifically reduced transcripts of PDGF-B. SiRNA knockdown of DEP-1 in endothelial cells in vitro also resulted in significant PDGF-B downregulation, providing further evidence for DEP-1 in PDGF-B gene regulation. In summary, our data support the notion of DEP-1 as positive functional regulator in vascular cerebral arteriogenesis, involving differential PDGF-B gene expression.
- Published
- 2013
- Full Text
- View/download PDF
38. Breakdown of the FLT3-ITD/STAT5 axis and synergistic apoptosis induction by the histone deacetylase inhibitor panobinostat and FLT3-specific inhibitors.
- Author
-
Pietschmann K, Bolck HA, Buchwald M, Spielberg S, Polzer H, Spiekermann K, Bug G, Heinzel T, Böhmer FD, and Krämer OH
- Subjects
- Benzothiazoles chemistry, Benzothiazoles pharmacology, Caspases metabolism, Cell Line, Drug Synergism, Gene Knockdown Techniques, Histone Deacetylase Inhibitors chemistry, Humans, Hydroxamic Acids chemistry, Indoles chemistry, Leukemia, Myeloid, Acute pathology, Panobinostat, Phenylurea Compounds chemistry, Phenylurea Compounds pharmacology, Protein Kinase Inhibitors chemistry, Protein Kinase Inhibitors pharmacology, Protein Stability drug effects, Proteolysis drug effects, Signal Transduction drug effects, Staurosporine analogs & derivatives, Staurosporine chemistry, Staurosporine pharmacology, fms-Like Tyrosine Kinase 3 metabolism, Apoptosis drug effects, Gene Duplication, Histone Deacetylase Inhibitors pharmacology, Hydroxamic Acids pharmacology, Indoles pharmacology, STAT5 Transcription Factor metabolism, fms-Like Tyrosine Kinase 3 antagonists & inhibitors, fms-Like Tyrosine Kinase 3 genetics
- Abstract
Activating mutations of the class III receptor tyrosine kinase FLT3 are the most frequent molecular aberration in acute myeloid leukemia (AML). Mutant FLT3 accelerates proliferation, suppresses apoptosis, and correlates with poor prognosis. Therefore, it is a promising therapeutic target. Here, we show that RNA interference against FLT3 with an internal tandem duplication (FLT3-ITD) potentiates the efficacy of the histone deacetylase inhibitor (HDACi) panobinostat (LBH589) against AML cells expressing FLT3-ITD. Similar to RNA interference, tyrosine kinase inhibitors (TKI; AC220/cpd.102/PKC412) in combination with LBH589 exhibit superior activity against AML cells. Median dose-effect analyses of drug-induced apoptosis rates of AML cells (MV4-11 and MOLM-13) revealed combination index (CI) values indicating strong synergism. AC220, the most potent and FLT3-specific TKI, shows highest synergism with LBH589 in the low nanomolar range. A 4-hour exposure to LBH589 + AC220 already generates more than 50% apoptosis after 24 hours. Different cell lines lacking FLT3-ITD as well as normal peripheral blood mononuclear cells are not significantly affected by LBH589 + TKI, showing the specificity of this treatment regimen. Immunoblot analyses show that LBH589 + TKI induce apoptosis via degradation of FLT3-ITD and its prosurvival target STAT5. Previously, we showed the LBH589-induced proteasomal degradation of FLT3-ITD. Here, we show that activated caspase-3 also contributes to the degradation of FLT3-ITD and that STAT5 is a direct target of this protease. Our data strongly emphasize HDACi/TKI drug combinations as promising modality for the treatment of FLT3-ITD-positive AMLs., (©2012 AACR.)
- Published
- 2012
- Full Text
- View/download PDF
39. Expression of protein-tyrosine phosphatases in Acute Myeloid Leukemia cells: FLT3 ITD sustains high levels of DUSP6 expression.
- Author
-
Arora D, Köthe S, van den Eijnden M, Hooft van Huijsduijnen R, Heidel F, Fischer T, Scholl S, Tölle B, Böhmer SA, Lennartsson J, Isken F, Müller-Tidow C, and Böhmer FD
- Abstract
Protein-tyrosine phosphatases (PTPs) are important regulators of cellular signaling and changes in PTP activity can contribute to cell transformation. Little is known about the role of PTPs in Acute Myeloid Leukemia (AML). The aim of this study was therefore to establish a PTP expression profile in AML cells and to explore the possible role of FLT3 ITD (Fms-like tyrosine kinase 3 with internal tandem duplication), an important oncoprotein in AML for PTP gene expression. PTP mRNA expression was analyzed in AML cells from patients and in cell lines using a RT-qPCR platform for detection of transcripts of 92 PTP genes. PTP mRNA expression was also analyzed based on a public microarray data set for AML patients. Highly expressed PTPs in AML belong to all PTP subfamilies. Very abundantly expressed PTP genes include PTPRC, PTPN2, PTPN6, PTPN22, DUSP1, DUSP6, DUSP10, PTP4A1, PTP4A2, PTEN, and ACP1. PTP expression was further correlated with the presence of FLT3 ITD, focusing on a set of highly expressed dual-specificity phosphatases (DUSPs). Elevated expression of DUSP6 in patients harboring FLT3 ITD was detected in this analysis. The mechanism and functional role of FLT3 ITD-mediated upregulation of DUSP6 was then explored using pharmacological inhibitors of FLT3 ITD signal transduction and si/shRNA technology in human and murine cell lines. High DUSP6 expression was causally associated with the presence of FLT3 ITD and dependent on FLT3 ITD kinase activity and ERK signaling. DUSP6 depletion moderately increased ERK1/2 activity but attenuated FLT3 ITD-dependent cell proliferation of 32D cells. In conclusion, DUSP6 may play a contributing role to FLT3 ITD-mediated cell transformation.
- Published
- 2012
- Full Text
- View/download PDF
40. Cell transformation by FLT3 ITD in acute myeloid leukemia involves oxidative inactivation of the tumor suppressor protein-tyrosine phosphatase DEP-1/ PTPRJ.
- Author
-
Godfrey R, Arora D, Bauer R, Stopp S, Müller JP, Heinrich T, Böhmer SA, Dagnell M, Schnetzke U, Scholl S, Östman A, and Böhmer FD
- Subjects
- Animals, Cell Line, Tumor, Genes, Tumor Suppressor drug effects, HEK293 Cells, Humans, Mice, Mice, Inbred C3H, Oxidants pharmacology, Oxidation-Reduction drug effects, Reactive Oxygen Species pharmacology, Receptor-Like Protein Tyrosine Phosphatases, Class 3 antagonists & inhibitors, Receptor-Like Protein Tyrosine Phosphatases, Class 3 genetics, Receptor-Like Protein Tyrosine Phosphatases, Class 3 metabolism, Tandem Repeat Sequences genetics, Transfection, Cell Transformation, Neoplastic genetics, Leukemia, Myeloid, Acute genetics, fms-Like Tyrosine Kinase 3 genetics
- Abstract
Signal transduction of FMS-like tyrosine kinase 3 (FLT3) is regulated by protein-tyrosine phosphatases (PTPs). We recently identified the PTP DEP-1/CD148/PTPRJ as a novel negative regulator of FLT3. This study addressed the role of DEP-1 for regulation of the acute myeloid leukemia (AML)-related mutant FLT3 internal tandem duplication (ITD) protein. Our experiments revealed that DEP-1 was expressed but dysfunctional in cells transformed by FLT3 ITD. This was caused by enzymatic inactivation of DEP-1 through oxidation of the DEP-1 catalytic cysteine. In intact cells, including primary AML cells, FLT3 ITD kinase inhibition reactivated DEP-1. DEP-1 reactivation was also achieved by counteracting the high levels of reactive oxygen species (ROS) production detected in FLT3 ITD-expressing cell lines by inhibition of reduced NAD phosphate (NADPH)-oxidases, or by overexpression of catalase or peroxiredoxin-1 (Prx-1). Interference with ROS production in 32D cells inhibited cell transformation by FLT3 ITD in a DEP-1-dependent manner, because RNAi-mediated depletion of DEP-1 partially abrogated the inhibitory effect of ROS quenching. Reactivation of DEP-1 by stable overexpression of Prx-1 extended survival of mice in the 32D cell/C3H/HeJ mouse model of FLT3 ITD-driven myeloproliferative disease. The study thus uncovered DEP-1 oxidation as a novel event contributing to cell transformation by FLT3 ITD.
- Published
- 2012
- Full Text
- View/download PDF
41. Ponatinib may overcome resistance of FLT3-ITD harbouring additional point mutations, notably the previously refractory F691I mutation.
- Author
-
Zirm E, Spies-Weisshart B, Heidel F, Schnetzke U, Böhmer FD, Hochhaus A, Fischer T, and Scholl S
- Subjects
- Apoptosis drug effects, Apoptosis genetics, Cell Line, Cell Proliferation drug effects, Drug Resistance, Neoplasm genetics, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Myeloid Cell Leukemia Sequence 1 Protein, Phosphorylation drug effects, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, Signal Transduction drug effects, fms-Like Tyrosine Kinase 3 antagonists & inhibitors, Antineoplastic Agents pharmacology, Imidazoles pharmacology, Point Mutation, Protein Kinase Inhibitors pharmacology, Pyridazines pharmacology, fms-Like Tyrosine Kinase 3 genetics
- Abstract
Fms-like tyrosine kinase (FLT3) mutations are the most frequent mutations in patients with acute myeloid leukaemia (AML) that confer a poor prognosis. Constitutively active FLT3-ITD (internal tandem duplications) mutations define a promising target for therapeutic approaches using small molecule inhibitors. However, several point mutations of the FLT3 tyrosine kinase domain (FLT3-TKD) have been identified to mediate resistance towards FLT3 tyrosine kinase inhibitors (FLT3-TKI), including secondary mutations of FLT3. We investigated the cellular effects of the recently characterised FLT3-TKI ponatinib (AP24534) on murine myeloid cells transfected with FLT3-ITD with or without additional point mutations of the FLT3-TKD including the (so far) multi-resistant F691I mutation. Ponatinib effectively induced apoptosis not only in the parental FLT3-ITD cell line but also in all stably transfected subclones harbouring additional FLT3-TKD point mutations (N676D, F691I or G697R). These observations correlated with a strong inhibition of FLT3-ITD and its downstream targets STAT5, AKT and ERK1/2 upon ponatinib incubation, as determined by Western blotting. We conclude that ponatinib represents a promising FLT3-TKI that should be further investigated in clinical trials. The targeted therapy of FLT3-ITD-positive AML with ponatinib might be associated with a lower frequency of secondary resistance caused by acquired FLT3-TKD mutations., (© 2012 Blackwell Publishing Ltd.)
- Published
- 2012
- Full Text
- View/download PDF
42. Novel inhibitors of epidermal growth factor receptor: (4-(Arylamino)-7H-pyrrolo[2,3-d]pyrimidin-6-yl)(1H-indol-2-yl)methanones and (1H-indol-2-yl)(4-(phenylamino)thieno[2,3-d]pyrimidin-6-yl)methanones.
- Author
-
Beckers T, Sellmer A, Eichhorn E, Pongratz H, Schächtele C, Totzke F, Kelter G, Krumbach R, Fiebig HH, Böhmer FD, and Mahboobi S
- Subjects
- Antineoplastic Agents chemical synthesis, Antineoplastic Agents pharmacology, Cell Line, Tumor, Drug Screening Assays, Antitumor, ErbB Receptors metabolism, Humans, Indoles chemical synthesis, Indoles pharmacology, Phosphorylation drug effects, Pyrimidines chemical synthesis, Pyrimidines pharmacology, Quinazolines chemical synthesis, Quinazolines chemistry, Quinazolines pharmacology, Structure-Activity Relationship, Antineoplastic Agents chemistry, ErbB Receptors antagonists & inhibitors, Indoles chemistry, Pyrimidines chemistry
- Abstract
Several members of the quinazoline class of known tyrosine kinase inhibitors are approved anticancer agents, often showing selectivity for receptors of the HER/ErbB-family. Combining structural elements of this class with the bisindolylmethanone-structure led to a series of novel compounds. These compounds inhibited EGFR in the nanomolar range. Moreover, inhibition of EGFR autophosphorylation in intact A431 cells was shown, with IC(50) values ranging form 0.3-1μM for compound 42, and 0.1-0.3μM for 45. In a panel of 42 human tumor cell lines the sensitivity profile of the novel compounds was shown to be similar to that of the quinazoline class of tyrosine kinase inhibitors lapatinib and erlotinib (Tarceva®)., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
43. Regulation of protein tyrosine phosphatases by reversible oxidation.
- Author
-
Ostman A, Frijhoff J, Sandin A, and Böhmer FD
- Subjects
- Animals, Humans, Oxidation-Reduction, Reactive Nitrogen Species metabolism, Reactive Oxygen Species metabolism, Signal Transduction, Protein Tyrosine Phosphatases metabolism
- Abstract
Oxidation of the catalytic cysteine of protein-tyrosine phosphatases (PTP), which leads to their reversible inactivation, has emerged as an important regulatory mechanism linking cellular tyrosine phosphorylation and signalling by reactive-oxygen or -nitrogen species (ROS, RNS). This review focuses on recent findings about the involved pathways, enzymes and biochemical mechanisms. Both the general cellular redox state and extracellular ligand-stimulated ROS production can cause PTP oxidation. Members of the PTP family differ in their intrinsic susceptibility to oxidation, and different types of oxidative modification of the PTP catalytic cysteine can occur. The role of PTP oxidation for physiological signalling processes as well as in different pathologies is described on the basis of well-investigated examples. Criteria to establish the causal involvement of PTP oxidation in a given process are proposed. A better understanding of mechanisms leading to selective PTP oxidation in a cellular context, and finding ways to pharmacologically modulate these pathways are important topics for future research.
- Published
- 2011
- Full Text
- View/download PDF
44. Loss of the protein-tyrosine phosphatase DEP-1/PTPRJ drives meningioma cell motility.
- Author
-
Petermann A, Haase D, Wetzel A, Balavenkatraman KK, Tenev T, Gührs KH, Friedrich S, Nakamura M, Mawrin C, and Böhmer FD
- Subjects
- Animals, Cell Adhesion genetics, Cell Line, Tumor, Gene Knockdown Techniques, Humans, Immunoblotting, Immunohistochemistry, Loss of Heterozygosity, Meningeal Neoplasms genetics, Meningeal Neoplasms pathology, Meningioma genetics, Meningioma pathology, Mice, Mice, Nude, Paxillin metabolism, Polymerase Chain Reaction, RNA, Small Interfering, Receptor-Like Protein Tyrosine Phosphatases, Class 3 genetics, Receptor-Like Protein Tyrosine Phosphatases, Class 3 metabolism, Signal Transduction physiology, Transfection, Cell Movement genetics, Meningeal Neoplasms metabolism, Meningioma metabolism, Neoplasm Invasiveness genetics
- Abstract
DEP-1/PTPRJ is a transmembrane protein-tyrosine phosphatase which has been proposed as a suppressor of epithelial tumors. We have found loss of heterozygosity (LOH) of the PTPRJ gene and loss of DEP-1 protein expression in a subset of human meningiomas. RNAi-mediated suppression of DEP-1 in DEP-1 positive meningioma cell lines caused enhanced motility and colony formation in semi-solid media. Cells devoid of DEP-1 exhibited enhanced signaling of endogenous platelet-derived growth factor (PDGF) receptors, and reduced paxillin phosphorylation upon seeding. Moreover, DEP-1 loss caused diminished adhesion to different matrices, and impaired cell spreading. DEP-1-deficient meningioma cells exhibited invasive growth in an orthotopic xenotransplantation model in nude mice, indicating that elevated motility translates into a biological phenotype in vivo. We propose that negative regulation of PDGF receptor signaling and positive regulation of adhesion signaling by DEP-1 cooperate in inhibition of meningioma cell motility, and possibly tumor invasiveness. These phenotypes of DEP-1 loss reveal functions of DEP-1 in adherent cells, and may be more generally relevant for tumorigenesis., (© 2010 The Authors. Brain Pathology © 2010 International Society of Neuropathology.)
- Published
- 2011
- Full Text
- View/download PDF
45. Protein-tyrosine phosphatase DEP-1 controls receptor tyrosine kinase FLT3 signaling.
- Author
-
Arora D, Stopp S, Böhmer SA, Schons J, Godfrey R, Masson K, Razumovskaya E, Rönnstrand L, Tänzer S, Bauer R, Böhmer FD, and Müller JP
- Subjects
- Amino Acid Substitution, Animals, Cell Line, Tumor, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Enzyme Activation physiology, Gene Knockdown Techniques, HEK293 Cells, Humans, Leukemia genetics, Leukemia metabolism, Male, Mice, Mutation, Missense, Phosphorylation, Receptor-Like Protein Tyrosine Phosphatases, Class 3 genetics, Receptor-Like Protein Tyrosine Phosphatases, Class 3 metabolism, STAT5 Transcription Factor genetics, STAT5 Transcription Factor metabolism, fms-Like Tyrosine Kinase 3 genetics, Signal Transduction physiology, fms-Like Tyrosine Kinase 3 metabolism
- Abstract
Fms-like tyrosine kinase 3 (FLT3) plays an important role in hematopoietic differentiation, and constitutively active FLT3 mutant proteins contribute to the development of acute myeloid leukemia. Little is known about the protein-tyrosine phosphatases (PTP) affecting the signaling activity of FLT3. To identify such PTP, myeloid cells expressing wild type FLT3 were infected with a panel of lentiviral pseudotypes carrying shRNA expression cassettes targeting different PTP. Out of 20 PTP tested, expressed in hematopoietic cells, or presumed to be involved in oncogenesis or tumor suppression, DEP-1 (PTPRJ) was identified as a PTP negatively regulating FLT3 phosphorylation and signaling. Stable 32D myeloid cell lines with strongly reduced DEP-1 levels showed site-selective hyperphosphorylation of FLT3. In particular, the sites pTyr-589, pTyr-591, and pTyr-842 involved in the FLT3 ligand (FL)-mediated activation of FLT3 were hyperphosphorylated the most. Similarly, acute depletion of DEP-1 in the human AML cell line THP-1 caused elevated FLT3 phosphorylation. Direct interaction of DEP-1 and FLT3 was demonstrated by "substrate trapping" experiments showing association of DEP-1 D1205A or C1239S mutant proteins with FLT3 by co-immunoprecipitation. Moreover, activated FLT3 could be dephosphorylated by recombinant DEP-1 in vitro. Enhanced FLT3 phosphorylation in DEP-1-depleted cells was accompanied by enhanced FLT3-dependent activation of ERK and cell proliferation. Stable overexpression of DEP-1 in 32D cells and transient overexpression with FLT3 in HEK293 cells resulted in reduction of FL-mediated FLT3 signaling activity. Furthermore, FL-stimulated colony formation of 32D cells expressing FLT3 in methylcellulose was induced in response to shRNA-mediated DEP-1 knockdown. This transforming effect of DEP-1 knockdown was consistent with a moderately increased activation of STAT5 upon FL stimulation but did not translate into myeloproliferative disease formation in the 32D-C3H/HeJ mouse model. The data indicate that DEP-1 is negatively regulating FLT3 signaling activity and that its loss may contribute to but is not sufficient for leukemogenic cell transformation.
- Published
- 2011
- Full Text
- View/download PDF
46. EGF/TGFβ1 co-stimulation of oral squamous cell carcinoma cells causes an epithelial-mesenchymal transition cell phenotype expressing laminin 332.
- Author
-
Richter P, Umbreit C, Franz M, Berndt A, Grimm S, Uecker A, Böhmer FD, Kosmehl H, and Berndt A
- Subjects
- Carcinoma, Squamous Cell metabolism, Cell Differentiation, Cell Line, Tumor, Cell Transformation, Neoplastic metabolism, Cells, Cultured, Epidermal Growth Factor physiology, Epithelial Cells metabolism, Epithelial Cells pathology, Extracellular Matrix Proteins metabolism, Humans, Laminin metabolism, Mouth Neoplasms metabolism, Neoplasm Invasiveness, Vimentin metabolism, Kalinin, Carcinoma, Squamous Cell pathology, Cell Adhesion Molecules metabolism, Cell Transformation, Neoplastic pathology, Mouth Neoplasms pathology, Transforming Growth Factor beta1 physiology
- Abstract
Epithelial-mesenchymal transition (EMT) is suggested to be crucial for the development of an invasive and metastatic carcinoma cell phenotype. Therefore, the definition of this phenotype is of great clinical interest. We recently evidenced vimentin positive cells in oral squamous cell carcinoma (OSCC) invasive front expressing laminin γ2 chain mRNA implicating an EMT origin of these cells. To further elucidate the nature of these cells, we have investigated the relation between EMT criteria and laminin-332 expression in a cell culture model of transforming growth factor beta-1 (TGFβ1)/epithelial growth factor (EGF) long time co-stimulation. We demonstrate that in contrast to TGFβ1 or EGF alone, co-stimulation induces phenotype transition in OSCC cells which fulfils the criteria of EMT in terms of vimentin up-regulation and E-cadherin down-regulation on protein level as well as cell scattering. Furthermore, cells displayed a strongly enhanced invasiveness and adhesion to type I-IV collagens. Phenotype transition is accompanied by an enhanced expression of laminin-332, especially of its γ2 chain. We further analyse the expression of extracellular matrix related genes by RT-PCR profiling. With respect to strongly enhanced proteins, data confirm the EMT phenotype of co-stimulated OSCC cells and expression of laminin-332. Furthermore, alpha catenin, collagen type 16, the integrin α7 and β1 chains, and MMP11 are suggested as candidates with potential role in EMT in OSCC. In summary we are able to show that EMT in OSCC is mediated by multiple growth factors and is accompanied by laminin γ2 chain up-regulation evidencing the existence of an intermediate Vim(+) /Ln332(+) EMT phenotype as seen in situ., (© 2010 John Wiley & Sons A/S.)
- Published
- 2011
- Full Text
- View/download PDF
47. Chimeric tyrosine kinase-HDAC inhibitors as antiproliferative agents.
- Author
-
Uecker A, Sicker M, Beckers T, Mahboobi S, Hägerstrand D, Ostman A, and Böhmer FD
- Subjects
- Antineoplastic Agents chemical synthesis, Benzamides, Cell Line, Tumor, Cell Proliferation drug effects, Drug Evaluation, Preclinical, Histone Deacetylase Inhibitors chemical synthesis, Humans, Imatinib Mesylate, Piperazines therapeutic use, Protein Kinase Inhibitors chemical synthesis, Pyrimidines therapeutic use, Receptors, Platelet-Derived Growth Factor drug effects, Receptors, Platelet-Derived Growth Factor metabolism, Antineoplastic Agents therapeutic use, Histone Deacetylase Inhibitors therapeutic use, Protein Kinase Inhibitors therapeutic use, Protein-Tyrosine Kinases antagonists & inhibitors
- Abstract
Combined treatment with tyrosine kinase inhibitors (TKi) and additional drugs is emerging as a promising strategy for cancer therapy. TKi and histone-deacetylase inhibitors (HDI) are two classes of anti-tumor agents with distant mechanisms of action. We have designed and synthesized chimeric compounds, which comprise structural elements of the TKi imatinib, and of prototypical HDI compounds. These compounds retain TKi activity similar to imatinib, exemplified by the inhibition of the platelet-derived growth factor receptor, and c-Kit kinase in intact cells. In addition, the chimeric compounds have in vitro and cellular HDI activity, and potently inhibit growth of cancer cell lines, including that of imatinib-resistant cell lines. Chimeric molecules with combined TKi and HDI activity may simplify combination treatment and be applicable to overcome clinical resistance to TKi single-agent therapy.
- Published
- 2010
- Full Text
- View/download PDF
48. Ubiquitin conjugase UBCH8 targets active FMS-like tyrosine kinase 3 for proteasomal degradation.
- Author
-
Buchwald M, Pietschmann K, Müller JP, Böhmer FD, Heinzel T, and Krämer OH
- Subjects
- Blotting, Western, Cell Line, Cell Separation, Flow Cytometry, Histone Deacetylase Inhibitors pharmacology, Humans, Hydrolysis, Immunoprecipitation, Mutation, Phosphorylation, Tyrosine metabolism, fms-Like Tyrosine Kinase 3 genetics, Proteasome Endopeptidase Complex metabolism, Ubiquitin-Conjugating Enzymes metabolism, fms-Like Tyrosine Kinase 3 metabolism
- Abstract
The class III receptor tyrosine kinase FMS-like tyrosine kinase 3 (FLT3) regulates normal hematopoiesis and immunological functions. Nonetheless, constitutively active mutant FLT3 (FLT3-ITD) causally contributes to transformation and is associated with poor prognosis of acute myeloid leukemia (AML) patients. Histone deacetylase inhibitors (HDACi) can counteract deregulated gene expression profiles and decrease oncoprotein stability, which renders them candidate drugs for AML treatment. However, these drugs have pleiotropic effects and it is often unclear how they correct oncogenic transcriptomes and proteomes. We report here that treatment of AML cells with the HDACi LBH589 induces the ubiquitin-conjugating enzyme UBCH8 and degradation of FLT3-ITD. Gain- and loss-of-function approaches show that UBCH8 and the ubiquitin-ligase SIAH1 physically interact with and target FLT3-ITD for proteasomal degradation. These ubiquitinylating enzymes though have a significantly lesser effect on wild-type FLT3. Furthermore, physiological and pharmacological stimulation of FLT3 phosphorylation, inhibition of FLT3-ITD autophosphorylation and analysis of kinase-inactive FLT3-ITD revealed that tyrosine phosphorylation determines degradation of FLT3 and FLT3-ITD by the proteasome. These results provide novel insights into antileukemic activities of HDACi and position UBCH8, which have been implicated primarily in processes in the nucleus, as a previously unrecognized important modulator of FLT3-ITD stability and leukemic cell survival.
- Published
- 2010
- Full Text
- View/download PDF
49. Stent-based release of a selective PDGF-receptor blocker from the bis-indolylmethanon class inhibits restenosis in the rabbit animal model.
- Author
-
Jandt E, Mutschke O, Mahboobi S, Uecker A, Platz R, Berndt A, Böhmer FD, Figulla HR, and Werner GS
- Subjects
- Animals, Coronary Restenosis drug therapy, Coronary Restenosis pathology, Female, Iliac Artery metabolism, Iliac Artery pathology, Rabbits, Receptors, Platelet-Derived Growth Factor metabolism, Coronary Restenosis prevention & control, Disease Models, Animal, Drug-Eluting Stents, Iliac Artery drug effects, Indoles administration & dosage, Receptors, Platelet-Derived Growth Factor antagonists & inhibitors
- Abstract
Long-term success of modern therapies for myocardial ischemia is limited by restenosis, with proliferation and migration of vascular smooth muscle cells (VSMC) as key events. Since findings in recent years indicate, that the Platelet Derived Growth Factor (PDGF) is an important selective factor in mitogenic and motogenic pathways of VSMC, different concepts for reducing restenosis by inhibiting PDGF signaling have been investigated, with local delivery of small receptor kinase inhibitors looking most promising. We tested the stent-based delivery of the PDGF-receptor inhibitor D-65495, a bis(1H-2-indolyl)methanone, in the rabbit iliac artery model of restenosis. New Zealand white rabbits underwent balloon dilation of iliac arteries for implantation of D-65495-coated or non-coated (solvent, either DMSO or 90%THF / 10% DMSO) coronary stents. After 4 weeks stents were removed and neointima formation in medial and proximal/ distal stent sections was histomorphometrically and immunohistochemically analyzed. Arteries with D-65495 eluting stents showed an up to 50% reduced restenosis compared to control stents. Also, the neointimal area was reduced, but there were no significant differences in injury score. Importantly, endothelialization was similar for control stents as well as for D-65495-coated stents, suggesting absence of a general cytostatic effect of the inhibitor. The impact of D-65495 on PDGF-receptor signaling in the vessel wall was indirectly assessed by immunohistochemical staining for activated protein kinase Akt, and PCNA as a proliferation marker and revealed some reduction for the inhibitor-treated vessels. In conclusion, the application of D-65495 caused a significant decrease in neointima formation, further supporting the concept of using locally released PDGF-receptor kinase inhibitors as anti-restenotic agents., (2009 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
50. Mislocalized activation of oncogenic RTKs switches downstream signaling outcomes.
- Author
-
Choudhary C, Olsen JV, Brandts C, Cox J, Reddy PN, Böhmer FD, Gerke V, Schmidt-Arras DE, Berdel WE, Müller-Tidow C, Mann M, and Serve H
- Subjects
- Amino Acid Sequence, Animals, Apoptosis drug effects, Brefeldin A pharmacology, Cell Compartmentation drug effects, Endoplasmic Reticulum drug effects, Endoplasmic Reticulum enzymology, Enzyme Activation drug effects, HeLa Cells, Humans, Isotope Labeling, Mice, Mitogen-Activated Protein Kinases metabolism, Molecular Sequence Data, Phosphatidylinositol 3-Kinases metabolism, Phosphoproteins chemistry, Phosphoproteins metabolism, Phosphorylation drug effects, Protein Structure, Tertiary, Proteomics, Proto-Oncogene Proteins c-kit metabolism, Receptor Protein-Tyrosine Kinases chemistry, STAT5 Transcription Factor metabolism, Sequence Deletion, Tunicamycin pharmacology, fms-Like Tyrosine Kinase 3 chemistry, fms-Like Tyrosine Kinase 3 metabolism, Oncogenes, Receptor Protein-Tyrosine Kinases metabolism, Signal Transduction drug effects
- Abstract
Inappropriate activation of oncogenic kinases at intracellular locations is frequently observed in human cancers, but its effects on global signaling are incompletely understood. Here, we show that the oncogenic mutant of Flt3 (Flt3-ITD), when localized at the endoplasmic reticulum (ER), aberrantly activates STAT5 and upregulates its targets, Pim-1/2, but fails to activate PI3K and MAPK signaling. Conversely, membrane targeting of Flt3-ITD strongly activates the MAPK and PI3K pathways, with diminished phosphorylation of STAT5. Global phosphoproteomics quantified 12,186 phosphorylation sites, confirmed compartment-dependent activation of these pathways and discovered many additional components of Flt3-ITD signaling. The differential activation of Akt and Pim kinases by ER-retained Flt3-ITD helped to identify their putative targets. Surprisingly, we find spatial regulation of tyrosine phosphorylation patterns of the receptor itself. Thus, intracellular activation of RTKs by oncogenic mutations in the biosynthetic route may exploit cellular architecture to initiate aberrant signaling cascades, thus evading negative regulation.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.