Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Int J Mol Sci ; 21(23)2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33255664

ABSTRACT

A microfluidic assay for the detection of promyelocytic leukemia (PML)-retinoic acid receptor α (RARα) fusion protein was developed. This microfluidic-based system can be used for rapid personalized differential diagnosis of acute promyelocyte leukemia (APL) with the aim of early initiation of individualized therapy. The fusion protein PML-RARα occurs in 95% of acute promyelocytic leukemia cases and is considered as diagnostically relevant. The fusion protein is formed as a result of translocation t(15,17) and is detected in the laboratory by fluorescence in situ hybridization (FISH) or reverse transcriptase polymerase chain reaction (RT-PCR). Diagnostic methods require many laboratory steps with specialized staff. The developed microfluidic assay includes a sandwich enzyme-linked immunosorbent assay (ELISA) system for PML-RARα on surface of magnetic microparticles in a microfluidic chip. A rapid detection of PML-RARα in cell lysates is achieved in less than one hour. A biotinylated PML-antibody on the surface of magnetic streptavidin coated microparticles is used as capture antibody. The bound translocation product is detected by a RARα antibody conjugated with horseradish peroxidase and the substrate QuantaRed. The analysis is performed in microfluidic channels which involves automated liquid processing with stringent washing and short incubation times. The results of the developed assay show that cell lysates of PML-RARα-positive cells (NB-4) can be clearly distinguished from PML-RARα-negative cells (HL-60, MV4-11).


Subject(s)
Leukemia, Promyelocytic, Acute/diagnosis , Oncogene Proteins, Fusion/genetics , Promyelocytic Leukemia Protein/genetics , Retinoic Acid Receptor alpha/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/isolation & purification , Granulocyte Precursor Cells/metabolism , Granulocyte Precursor Cells/pathology , Humans , In Situ Hybridization, Fluorescence/methods , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Microfluidics/methods , Oncogene Proteins, Fusion/isolation & purification , Precision Medicine , Translocation, Genetic/genetics
2.
PLoS One ; 9(4): e94993, 2014.
Article in English | MEDLINE | ID: mdl-24740120

ABSTRACT

External signals that are mediated by specific receptors determine stem cell fate. The thrombin receptor PAR1 plays an important role in haemostasis, thrombosis and vascular biology, but also in tumor biology and angiogenesis. Its expression and function in hematopoietic stem cells is largely unknown. Here, we analyzed expression and function of PAR1 in primary hematopoietic cells and their leukemic counterparts. AML patients' blast cells expressed much lower levels of PAR1 mRNA and protein than CD34+ progenitor cells. Constitutive Par1-deficiency in adult mice did not affect engraftment or stem cell potential of hematopoietic cells. To model an AML with Par1-deficiency, we retrovirally introduced the oncogene MLL-AF9 in wild type and Par1-/- hematopoietic progenitor cells. Par1-deficiency did not alter initial leukemia development. However, the loss of Par1 enhanced leukemic stem cell function in vitro and in vivo. Re-expression of PAR1 in Par1-/- leukemic stem cells delayed leukemogenesis in vivo. These data indicate that Par1 contributes to leukemic stem cell maintenance.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Leukemia/genetics , Receptor, PAR-1/genetics , Acute Disease , Animals , Hematopoiesis/genetics , Humans , Immunohistochemistry , Leukemia/metabolism , Leukemia/pathology , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Receptor, PAR-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
3.
Clin Cancer Res ; 20(4): 814-26, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24334763

ABSTRACT

PURPOSE: Cancer cell phenotypes are partially determined by epigenetic specifications, such as DNA methylation. Metastasis development is a late event in cancerogenesis and might be associated with epigenetic alterations. EXPERIMENTAL DESIGN: An in vivo selection approach was used to generate highly aggressive non-small cell lung cancer (NSCLC) cell lines (A549 and HTB56) followed by genome-wide DNA methylation analysis. Furthermore, the therapeutic effects of the epigenetic agent azacytidine on DNA methylation patterns and the in vivo phenotypes were explored. RESULTS: Widespread changes of DNA methylation were observed during development of highly aggressive cell lines. Up to 2.5% of the CpG-rich region was differentially methylated as identified by reduced representation bisulfite sequencing compared with the less aggressive parental cell lines. DNA methyltransferase inhibition by azacytidine reversed the prometastatic phenotype; this was highly associated with the preferential loss of DNA methylation at sites that were hypermethylated during the in vivo selection. Of note, polycomb (PRC2) binding sites were particularly affected by DNA methylation changes after azacytidine exposure that persisted over time. CONCLUSIONS: We could show that metastatic capability of NSCLC is closely associated with DNA methylome alterations. Because inhibition of DNA methyltransferase reversed metastasis-prone phenotype, epigenetic modulation seems to be a potential therapeutic approach to prevent metastasis formation.


Subject(s)
Adenocarcinoma/genetics , DNA-Cytosine Methylases/antagonists & inhibitors , Epigenesis, Genetic , Lung Neoplasms/genetics , Polycomb-Group Proteins/physiology , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/secondary , Animals , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Binding Sites , Cell Line, Tumor , DNA Methylation , DNA-Cytosine Methylases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Phenotype
4.
J Biol Chem ; 286(32): 28210-22, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21540187

ABSTRACT

The cell cycle is driven by the kinase activity of cyclin·cyclin-dependent kinase (CDK) complexes, which is negatively regulated by CDK inhibitor proteins. Recently, we identified INCA1 as an interaction partner and a substrate of cyclin A1 in complex with CDK2. On a functional level, we identified a novel cyclin-binding site in the INCA1 protein. INCA1 inhibited CDK2 activity and cell proliferation. The inhibitory effects depended on the cyclin-interacting domain. Mitogenic and oncogenic signals suppressed INCA1 expression, whereas it was induced by cell cycle arrest. We established a deletional mouse model that showed increased CDK2 activity in spleen with altered spleen architecture in Inca1(-/-) mice. Inca1(-/-) embryonic fibroblasts showed an increase in the fraction of S-phase cells. Furthermore, blasts from acute lymphoid leukemia and acute myeloid leukemia patients expressed significantly reduced INCA1 levels highlighting its relevance for growth control in vivo. Taken together, this study identifies a novel CDK inhibitor with reduced expression in acute myeloid and lymphoid leukemia. The molecular events that control the cell cycle occur in a sequential process to ensure a tight regulation, which is important for the survival of a cell and includes the detection and repair of genetic damage and the prevention of uncontrolled cell division.


Subject(s)
Blast Crisis/metabolism , Carrier Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Neoplasm Proteins/metabolism , Phosphoproteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , S Phase , Signal Transduction , Animals , Blast Crisis/genetics , Blast Crisis/pathology , Carrier Proteins/genetics , Cell Line , Cell Survival/genetics , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Phosphoproteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
5.
Leuk Res ; 35(5): 620-5, 2011 May.
Article in English | MEDLINE | ID: mdl-21176959

ABSTRACT

Epigenetic changes play a crucial role in leukemogenesis. HDACs are frequently recruited to target gene promoters by balanced translocation derived oncogenic fusion proteins. As important epigenetic effector mechanisms, histone deacetylases (HDAC) have emerged as potential therapeutic targets. However, the patterns of HDAC1 localization and the role of HDACs in leukemia pathogenesis remain to be elucidated. Using ChIP-Chip analyses we analyzed HDAC1 deposition patterns at more than 10,000 gene promoters in a large cohort of leukemia patients and CD34+ controls. HDAC1 binding was significantly increased in AML blasts compared to CD34+ progenitor cells at 130 gene promoters whereas decreased binding was observed at 66 gene promoters. Distinct HDAC1 binding patterns occurred in AML subtypes with balanced translocations t(15;17), t(8;21) and inv(16). In addition, a more generalized signature was established, that revealed an AML specific pattern of HDAC1 distribution. Many of the HDAC1-binding altered promoters regulate genes involved in hematopoiesis, transcriptional regulation and signal transduction. HDAC1 binding patterns were associated with patients' event free survival. This is the first study to determine HDAC1 modification patterns in a large number of AML and ALL specimens. Our findings suggest that dyslocalization of HDAC1 is a common feature in AML. Importantly, HDAC1 modifications possess prognostic power for patient survival. Our findings suggest that altered HDAC1 localization is an explanation for the observed benefit of HDAC inhibitors in AML therapy.


Subject(s)
Hematopoiesis/genetics , Histone Deacetylase 1/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Promoter Regions, Genetic , Adolescent , Adult , Aged , Biomarkers, Tumor/metabolism , Child , Child, Preschool , Chromatin/metabolism , Female , Humans , Infant , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Prognosis , Promoter Regions, Genetic/physiology , Protein Binding/physiology , Survival Analysis , Young Adult
6.
Blood ; 116(18): 3564-71, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-20498303

ABSTRACT

Acute myeloid leukemia (AML) is commonly associated with alterations in transcription factors because of altered expression or gene mutations. These changes might induce leukemia-specific patterns of histone modifications. We used chromatin-immunoprecipitation on microarray to analyze histone 3 lysine 9 trimethylation (H3K9me3) patterns in primary AML (n = 108), acute lymphoid leukemia (n = 28), CD34(+) cells (n = 21) and white blood cells (n = 15) specimens. Hundreds of promoter regions in AML showed significant alterations in H3K9me3 levels. H3K9me3 deregulation in AML occurred preferentially as a decrease in H3K9me3 levels at core promoter regions. The altered genomic regions showed an overrepresentation of cis-binding sites for ETS and cyclic adenosine monophosphate response elements (CREs) for transcription factors of the CREB/CREM/ATF1 family. The decrease in H3K9me3 levels at CREs was associated with increased CRE-driven promoter activity in AML blasts in vivo. AML-specific H3K9me3 patterns were not associated with known cytogenetic abnormalities. But a signature derived from H3K9me3 patterns predicted event-free survival in AML patients. When the H3K9me3 signature was combined with established clinical prognostic markers, it outperformed prognosis prediction based on clinical parameters alone. These findings demonstrate widespread changes of H3K9me3 levels at gene promoters in AML. Signatures of histone modification patterns are associated with patient prognosis in AML.


Subject(s)
Histones/metabolism , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/metabolism , Lysine/metabolism , Adolescent , Antigens, CD34/immunology , Child , Child, Preschool , Disease-Free Survival , Female , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/immunology , Histones/genetics , Humans , Infant , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Male , Methylation , Prognosis , Promoter Regions, Genetic , Protein Binding , Transcription Factors/metabolism , Tumor Cells, Cultured
7.
Blood ; 115(22): 4507-16, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20215640

ABSTRACT

Although the potential role of Pim2 as a cooperative oncogene has been well described in lymphoma, its role in leukemia has remained largely unexplored. Here we show that high expression of Pim2 is observed in patients with acute promyelocytic leukemia (APL). To further characterize the cooperative role of Pim2 with promyelocytic leukemia/retinoic acid receptor alpha (PML/RARalpha), we used a well-established PML-RARalpha (PRalpha) mouse model. Pim2 coexpression in PRalpha-positive hematopoietic progenitor cells (HPCs) induces leukemia in recipient mice after a short latency. Pim2-PRalpha cells were able to repopulate mice in serial transplantations and to induce disease in all recipients. Neither Pim2 nor PRalpha alone was sufficient to induce leukemia upon transplantation in this model. The disease induced by Pim2 overexpression in PRalpha cells contained a slightly higher fraction of immature myeloid cells, compared with the previously described APL disease induced by PRalpha. However, it also clearly resembled an APL-like phenotype and showed signs of differentiation upon all-trans retinoic acid (ATRA) treatment in vitro. These results support the hypothesis that Pim2, which is also a known target of Flt3-ITD (another gene that cooperates with PML-RARalpha), cooperates with PRalpha to induce APL-like disease.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Animals , Base Sequence , Bone Marrow Transplantation , DNA Primers/genetics , Female , Gene Expression , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Leukemia, Promyelocytic, Acute/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Oncogenes , Tretinoin/pharmacology , fms-Like Tyrosine Kinase 3/genetics
8.
Blood ; 115(16): 3185-95, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20053753

ABSTRACT

In a previously developed inducible transgenic mouse model of chronic myeloid leukemia, we now demonstrate that the disease is transplantable using BCR-ABL(+) Lin(-)Sca-1(+)c-kit(+) (LSK) cells. Interestingly, the phenotype is more severe when unfractionated bone marrow cells are transplanted, yet neither progenitor cells (Lin(-)Sca-1(-)c-kit(+)), nor mature granulocytes (CD11b(+)Gr-1(+)), nor potential stem cell niche cells (CD45(-)Ter119(-)) are able to transmit the disease or alter the phenotype. The phenotype is largely independent of BCR-ABL priming before transplantation. However, prolonged BCR-ABL expression abrogates the potential of LSK cells to induce full-blown disease in secondary recipients and increases the fraction of multipotent progenitor cells at the expense of long-term hematopoietic stem cells (LT-HSCs) in the bone marrow. BCR-ABL alters the expression of genes involved in proliferation, survival, and hematopoietic development, probably contributing to the reduced LT-HSC frequency within BCR-ABL(+) LSK cells. Reversion of BCR-ABL, or treatment with imatinib, eradicates mature cells, whereas leukemic stem cells persist, giving rise to relapsed chronic myeloid leukemia on reinduction of BCR-ABL, or imatinib withdrawal. Our results suggest that BCR-ABL induces differentiation of LT-HSCs and decreases their self-renewal capacity.


Subject(s)
Cell Differentiation/genetics , Cell Transformation, Neoplastic/pathology , Fusion Proteins, bcr-abl/physiology , Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Animals , Cell Separation , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Flow Cytometry , Genes, abl/physiology , Hematopoietic Stem Cell Transplantation , Mice , Mice, Transgenic , Neoplasm Staging , Neoplasm Transplantation , Reverse Transcriptase Polymerase Chain Reaction
9.
Blood ; 114(19): 4197-208, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19734451

ABSTRACT

Somatic mutations of Kit have been found in leukemias and gastrointestinal stromal tumors. The proto-oncogene c-Cbl negatively regulates Kit and Flt3 by its E3 ligase activity and acts as a scaffold. We recently identified the first c-Cbl mutation in human disease in an acute myeloid leukemia patient, called Cbl-R420Q. Here we analyzed the role of Cbl mutants on Kit-mediated transformation. Coexpression of Cbl-R420Q or Cbl-70Z with Kit induced cytokine-independent proliferation, survival, and clonogenic growth. Primary murine bone marrow retrovirally transduced with c-Cbl mutants and transplanted into mice led to a generalized mastocytosis, a myeloproliferative disease, and myeloid leukemia. Overexpression of these Cbl mutants inhibited stem cell factor (SCF)-induced ubiquitination and internalization of Kit. Both Cbl mutants enhanced the basal activation of Akt and prolonged the ligand-dependent activation. Importantly, transformation was observed also with kinase-dead forms of Kit and Flt3 in the presence of Cbl-70Z, but not in the absence of Kit or Flt3, suggesting a mechanism dependent on receptor tyrosine kinases, but independent of their kinase activity. Instead, transformation depends on the Src family kinase Fyn, as c-Cbl coimmunoprecipitated with Fyn and inhibition abolished transformation. These findings may explain primary resistance to tyrosine kinase inhibitors targeted at receptor tyrosine kinases.


Subject(s)
Mastocytosis/genetics , Mutation , Myeloproliferative Disorders/genetics , Proto-Oncogene Proteins c-cbl/genetics , Animals , Bone Marrow Transplantation , COS Cells , Cell Transformation, Neoplastic , Chlorocebus aethiops , Disease Models, Animal , Female , Humans , Ligands , Mastocytosis/etiology , Mastocytosis/metabolism , Mastocytosis/pathology , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction , Ubiquitination
10.
Int J Oncol ; 33(6): 1215-21, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19020754

ABSTRACT

Wnt signalling regulates proliferation, self renewal and cell fate. Aberrant Wnt signalling is thought to contribute to AML pathogenesis by enhancing self renewal. Herein, we provide evidence for increased expression of Frizzled-4, a receptor for Wnt ligands, in primary AML blasts compared to normal bone marrow on the protein level. In addition, Frizzled-4 is highly expressed in human CD34 positive cells as well as in lineage negative sorted mouse bone marrow cells. Functionally, Frizzled-4 expression modulates apoptosis and enhances Wnt3a induced beta-catenin stability in myeloid progenitor cells. Frizzled-4-dependent beta-catenin stabilization is dkk-1 sensitive, implicating a specific Wnt-ligand/Frizzled-receptor interaction. These findings indicate enhanced sensitivity of AML blasts for Wnt-ligands and suggest an additional mechanism of Wnt signalling activation in the pathogenesis of AML.


Subject(s)
Frizzled Receptors/metabolism , Leukemia, Myeloid, Acute/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Wnt Proteins/metabolism , Animals , Apoptosis , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cell Lineage , Cell Survival , Frizzled Receptors/genetics , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Receptors, G-Protein-Coupled/genetics , Time Factors , Transduction, Genetic , Up-Regulation , Wnt Proteins/genetics , Wnt3 Protein , Wnt3A Protein , beta Catenin/metabolism
11.
Int J Hematol ; 88(2): 174-180, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18668305

ABSTRACT

The Wnt-signaling pathway plays a critical role in directing cell fate during embryogenesis and also in the pathogenesis of cancer. In leukemia, it is well described that activating internal tandem duplications (ITD) mutations in receptor tyrosine kinases like cKit or Flt3 confer to the pathogenesis of cancer. Here, we analyzed whether Wnt-signaling plays a role in cKit-ITD mediated transformation. Stably transfected 32D cells with cKit-ITD cells had higher beta-Catenin protein levels compared to the cKit-WT. Analysis of beta-Catenin mRNA and protein levels revealed that beta-Catenin was regulated at post-transcriptional level in cKit-ITD as well as Flt3-ITD compared to the wildtype. Signaling analyses revealed higher-phosphorylation of GSK3beta by oncogenic cKit-ITD. Moreover, activation of Wnt signaling was confirmed by constitutive activation of c-myc luciferase by cKit-ITD cells. Importantly, using dominant negative TCF4, we show that activation of Wnt signaling plays an important role in cKit mediated transformation of myeloid cells. Application of specific receptor tyrosine kinase inhibitors for Flt3 or cKit result in a decrease of beta-Catenin that underwent with a decrease of GSK3beta phosphorylation, suggesting an indirect mechanism of beta-Catenin regulation by oncogenic receptor tyrosine kinases in both ITD mutations. Our study shows the importance of activation of Wnt signaling in leukemia and suggests as attractive target for future therapeutical approaches.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute , Piperazines/pharmacology , Proto-Oncogene Proteins c-kit/metabolism , Pyrimidines/pharmacology , Wnt Proteins/metabolism , Animals , Benzamides , Cell Line , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Leukemic , Glycogen Synthase Kinase 3/metabolism , Humans , Imatinib Mesylate , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Phosphorylation , Proto-Oncogene Proteins c-kit/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Tandem Repeat Sequences , Transfection , Wnt Proteins/genetics , beta Catenin/genetics , beta Catenin/metabolism , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
12.
Blood ; 110(3): 1004-12, 2007 08 01.
Article in English | MEDLINE | ID: mdl-17446348

ABSTRACT

In acute myeloid leukemia (AML), mutational activation of the receptor tyrosine kinase (RTK) Flt3 is frequently involved in leukemic transformation. However, little is known about a possible role of highly expressed wild-type Flt3 in AML. The proto-oncogene c-Cbl is an important regulator of RTK signaling, acting through its ubiquitin ligase activity and as a platform for several signaling adaptor molecules. Here, we analyzed the role of c-Cbl in Flt3 signal transduction and myeloid transformation. C-Cbl physically interacted with Flt3 and was tyrosine phosphorylated in the presence of Flt3-ligand (FL). Overexpression of a dominant-negative form of c-Cbl (Cbl-70Z) inhibited FL-induced Flt3 ubiquitylation and internalization, indicating involvement of c-Cbl in Flt3 signaling. DNA sequencing of AML bone marrow revealed a case with a c-Cbl point mutation (Cbl-R420Q). Cbl-R420Q inhibited Flt3 internalization and ubiquitylation. Coexpression of Cbl-R420Q or Cbl-70Z with Flt3 induced cytokine-independent growth and survival of 32Dcl3 cells in the absence of FL. Also, the mutant Cbl proteins altered the amplitude and duration of Flt3-dependent signaling events. Our results indicate an important role of Cbl proteins in Flt3 signal modulation. Also, the data suggest a novel mechanism of leukemic transformation in AML by mutational inactivation of negative RTK regulators.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Leukemia, Myeloid, Acute/metabolism , Point Mutation , Proto-Oncogene Proteins c-cbl/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , COS Cells , Cell Survival , Cell Transformation, Neoplastic/genetics , Chlorocebus aethiops , Gene Expression Regulation, Leukemic , Genes, Dominant , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Proto-Oncogene Mas , Proto-Oncogene Proteins c-cbl/genetics , Signal Transduction , fms-Like Tyrosine Kinase 3/genetics
13.
Cancer Genet Cytogenet ; 174(1): 16-23, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17350462

ABSTRACT

Lipocalin 24p3 plays a direct role in iron transport and regulates the levels of important proteins of the iron metabolism. Iron-loaded 24p3 binds to its specific receptor (24p3R) on the cell surface. Upon binding to its receptor, 24p3 is internalized into the cell, where it releases its bound iron. Iron-free 24p3 can withdraw iron from inside the cell to the outside by a reverse mechanism. We analyzed the role of the murine 24p3 gene Lcn2 (alias 24p3) as a target of the Wnt pathway. In cells with activated Wnt pathway, the levels of 24p3 protein and RNA were decreased. The withdrawal of iron led to 24p3 downregulation, and iron addition to iron-deprived cells induced 24p3 expression. Despite its strong inhibitory effect on 24p3 expression, Wnt pathway activation had no effect on the intracellular iron level. In cells with nonactivated Wnt pathway, we found an as yet unidentified transcript of 24p3R. Our results indicate independent regulation of 24p3 expression by the Wnt pathway and by the intracellular iron level. Differential splicing of the 24p3R transcript, depending on the activation state of the Wnt pathway, may modify the function of 24p3.


Subject(s)
Acute-Phase Proteins/genetics , Acute-Phase Proteins/metabolism , Iron/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Wnt Proteins/metabolism , Animals , Base Sequence , DNA, Complementary/genetics , Gene Expression Regulation/drug effects , Iron Chelating Agents/pharmacology , Lipocalin-2 , Lipocalins , Mice , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits
14.
Blood ; 110(1): 370-4, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17356133

ABSTRACT

Mutations in the receptor tyrosine kinase Flt3 represent a very common genetic lesion in acute myeloid leukemia (AML). Internal tandem duplication (ITD) mutations clustered in the juxtamembrane domain are the most frequent and best characterized mutations found in Flt3. Oncogenic activation of Flt3 by ITD mutations is known to activate aberrant signaling including activation of STAT5 and repression of myeloid transcription factors Pu.1 and c/EBP-alpha. However, the mechanisms of STAT5 activation by Flt3-ITD remain unclear. Using small molecule inhibitors and cell lines deficient for Src family kinases or Jak2 or Tyk2, here we show that Flt3-ITD-induced STAT5 activation is independent of Src or Jak kinases. Also, overexpression of SOCS1, an inhibitor of Jak kinases, inhibited IL-3- but not Flt3-ITD-mediated STAT5 activation. Furthermore, in vitro kinase assays revealed that STAT5 is a direct target of Flt3. Taken together, our data provide the mechanistic basis of STAT5 activation by Flt3-ITD.


Subject(s)
STAT5 Transcription Factor/metabolism , Tandem Repeat Sequences , fms-Like Tyrosine Kinase 3/genetics , Animals , Cell Line , Janus Kinase 2 , Mice , Mice, Knockout , Mutation , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , TYK2 Kinase
15.
J Leukoc Biol ; 79(6): 1306-13, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16565323

ABSTRACT

The Wnt-signaling pathway plays a critical role in directing cell fate during embryogenesis. Several lines of evidence also suggest a role in inflammatory processes. Here, we analyzed whether Wnt signaling plays a role in leukocyte inflammatory responses. Monocytes from healthy donors expressed different Frizzled receptors, which are ligands for the Wnt molecules. Activation of the Wnt/beta-catenin pathway by LiCl or Wnt3a increased beta-catenin protein levels in monocytes but not in granulocytes. It is interesting that the activation of Wnt/beta-catenin signaling via Wnt3a in monocytes resulted in a decrease in migration through an endothelial layer (human dermal microvascular endothelial cell-1). Further experiments revealed that the decrease in transendothelial migration was associated with specific monocyte adherence to endothelial cells after Wnt exposure. The specificity was verified by a lack of Wnt3a-induced adhesion to fibronectin, laminin, or collagen compared with endothelial interaction. Analysis of the distribution of beta-catenin revealed a Wnt3a-induced increase of beta-catenin in the cytoplasm. Wnt3a exposure did not result in any activation of the classical Wnt-target gene c-myc or a Wnt-target gene involved in cell adhesion (Connexin43). Our study implicates for the first time a role of canonical Wnt signaling in inflammatory processes in monocytes.


Subject(s)
Endothelium, Vascular/cytology , Monocytes/physiology , Signal Transduction/physiology , Wnt Proteins/physiology , beta Catenin/physiology , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Cell Nucleus/chemistry , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cytoplasm/chemistry , Frizzled Receptors/biosynthesis , Frizzled Receptors/genetics , Frizzled Receptors/physiology , Granulocytes/drug effects , HL-60 Cells/drug effects , Humans , Lithium Chloride/pharmacology , Mice , Monocytes/drug effects , Recombinant Fusion Proteins/pharmacology , Wnt Proteins/genetics , Wnt Proteins/pharmacology , Wnt3 Protein , Wnt3A Protein , beta Catenin/biosynthesis , beta Catenin/genetics
16.
Expert Opin Emerg Drugs ; 11(1): 153-65, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16503833

ABSTRACT

Acute myeloid leukaemia (AML) is characterised by the infiltration of the bone marrow with highly proliferative leukaemic cells that stop to differentiate at different stages of myeloid development and carry survival advantages. Conventionally, AML is treated with aggressive cytotoxic therapy, in eligible patients followed by allogeneic bone marrow transplantation. However, despite this aggressive treatment, many patients relapse and eventually die from the disease. Activating mutations in the coding sequence of the receptor tyrosine kinase Flt3 are found in leukaemic blasts from approximately 30% of AML patients. The mutations have been described to severely alter the signalling properties of this receptor and to have transforming activity in cell-line models and in primary mouse bone marrow. The prognosis of patients harbouring the most common Flt3 mutations tends to be worse than that of comparable patients without the mutations. Thus, Flt3 seems a promising target for therapeutic intervention. Several small molecules that inhibit Flt3 kinase activity are being evaluated for the treatment of AML in clinical trials. This review article discusses the signal transduction and biological function of Flt3 and its mutations in normal and malignant haematopoiesis and recent progress in drug development aiming at the inhibition of Flt3 kinases.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/therapeutic use , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Carbazoles/therapeutic use , Clinical Trials as Topic , Drug Evaluation, Preclinical , Drug Therapy, Combination , Furans , Humans , Indoles/pharmacology , Indoles/therapeutic use , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Mutation , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Pyrroles/therapeutic use , Signal Transduction/drug effects , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Staurosporine/therapeutic use , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
17.
FEBS J ; 272(7): 1600-15, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15794748

ABSTRACT

The Wnt pathway controls biological processes via the regulation of target gene expression. The expression of direct Wnt target genes, e.g. cyclin D1 and MYC, is activated by the transcription factor TCF, which binds to specific sequence motifs in the promoter. Indirect target genes are regulated via transcription regulators, which are targets of the Wnt pathway. As an example, MYC regulates the MYC interacting zinc finger protein-1 (MIZ-1), which is able to inhibit the expression of the indirect target p21WAF1. We intended to identify new Wnt target genes and to get a deeper insight into the regulatory mechanisms of Wnt target gene expression. For this we analyzed the differential expression pattern of Wnt-1 activated cells by microarray analysis. We identified 43 sequences including eight expressed sequence tags (ESTs), which showed increased transcript levels, and 104 sequences including 19 ESTs with decreased RNA levels. Northern blot and real-time quantitative PCR analysis of the differential expression levels of 15 genes confirmed the differential expression trends of eight candidate genes. When the Wnt pathway was regulated at the lower level of glycogen synthase kinase-3 beta (GSK-3 beta) or adenomatous polyposis coli (APC), we detected discrepant expression trends. We compared the number of binding sites of transcription factors in the genomic regions of all candidate target genes with the number of sites in control genes. We found that the genomic regions of the down-regulated genes include an increased number of putative MIZ-1 binding sites. Our study introduces several new Wnt target genes and provides indications that the specific gene expression pattern depends on the type of the activation trigger or the level of interference with the Wnt pathway. Furthermore, our data indicate that a high proportion of Wnt target genes are regulated by indirect mechanisms.


Subject(s)
Gene Expression Regulation/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Promoter Regions, Genetic , Animals , Data Interpretation, Statistical , Intercellular Signaling Peptides and Proteins/genetics , Mice , Models, Genetic , Oligonucleotide Array Sequence Analysis , Wnt Proteins , Wnt1 Protein
18.
Blood ; 106(1): 265-73, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15769897

ABSTRACT

Activating mutations of Flt3 are found in approximately one third of patients with acute myeloid leukemia (AML) and are an attractive drug target. Two classes of Flt3 mutations occur: internal tandem duplications (ITDs) in the juxtamembrane and point mutations in the tyrosine kinase domain (TKD). We and others have shown that Flt3-ITD induced aberrant signaling including strong activation of signal transducer and activator of transcription 5 (STAT5) and repression of CCAAT/estradiol-binding protein alpha (c/EBPalpha) and Pu.1. Here, we compared the signaling properties of Flt3-ITD versus Flt3-TKD in myeloid progenitor cells. We demonstrate that Flt3-TKD mutations induced autonomous growth of 32D cells in suspension cultures. However, in contrast to Flt3-ITD and similar to wild-type Flt3 (Flt3-WT), Flt3-TKD cannot support colony formation in semisolid media. Also, in contrast to Flt3-ITD, neither Flt3-WT nor Flt3-TKD induced activation or induction of STAT5 target genes. Flt3-TKD also failed to repress c/EBPalpha and Pu.1. No significant differences were observed in receptor autophosphorylation and the phosphorylation of Erk-1 and -2, Akt, and Shc. Importantly, TKD but not ITD mutations were a log power more sensitive toward the tyrosine kinase inhibitor protein kinase C 412 (PKC412) than Flt3-WT. In conclusion, Flt3-ITD and Flt3-TKD mutations display differences in their signaling properties that could have important implications for their transforming capacity and for the design of mutation-specific therapeutic approaches.


Subject(s)
Leukemia, Myeloid/metabolism , Myeloid Cells/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Staurosporine/analogs & derivatives , Acute Disease , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Cell Line , DNA-Binding Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/pharmacology , Milk Proteins/metabolism , Muridae , Mutagenesis, Site-Directed , Myeloid Cells/cytology , Phosphorylation , Point Mutation , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , STAT5 Transcription Factor , Staurosporine/pharmacology , Tandem Repeat Sequences , Trans-Activators/metabolism , Transcription Factors/metabolism , fms-Like Tyrosine Kinase 3
19.
Blood ; 105(9): 3699-706, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15650056

ABSTRACT

Activating Flt3 mutations occur in about 30% of patients with acute myeloid leukemia (AML), often as in-frame internal tandem duplication (ITD) at the juxtamembrane domain of the receptor. These mutations transform hematopoietic cell lines and primary mouse bone marrow. Here, we analyzed the interaction between oncogenic Flt3-ITD mutations and the Wingless-type (Wnt) signaling pathway in the myeloid progenitor cell line 32D. Microarray analyses revealed higher mRNA expression of Frizzled-4, a receptor for Wnt ligands in 32D/Flt3-ITD cells. Findings were verified by quantitative realtime reverse transcription-polymerase chain reaction (RT-PCR) and on the protein level. Compared with 32D/Flt3-WT (wild-type) cells, 32D/Flt3-ITD cells also showed greatly enhanced beta-catenin protein levels, irrespective of their exposure to Wnt3a, a ligand inducing the canonical Wnt signal transduction pathway. In addition, 5 of 7 AML samples with Flt3-ITD mutations expressed high beta-catenin protein levels, whereas patients with wild-type Flt3 did not. Also, Flt3-ITD induced enhanced T-cell factor (TCF)-dependent transcriptional activity and the induction of the Wnt target gene c-myc. In the presence of Flt3-WT or Flt3-ITD signaling, Wnt3a slightly increased 32D cell proliferation. However, transfection experiments with dominant-negative (dn) TCF4 revealed a strong dependence of Flt3-ITD-mediated clonogenic growth on TCF activity. Taken together, our results indicate that Flt3-ITD and Wnt-dependent signaling pathways synergize in myeloid transformation.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Leukemia/pathology , Mutation , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Tandem Repeat Sequences , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cytoskeletal Proteins/analysis , DNA-Binding Proteins , Humans , Leukemia/etiology , Leukemia/genetics , Mice , Myeloid Progenitor Cells , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , TCF Transcription Factors , Trans-Activators/analysis , Transcription Factor 7-Like 2 Protein , Transcription Factors , Wnt Proteins , beta Catenin , fms-Like Tyrosine Kinase 3
20.
Blood ; 105(5): 2107-14, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15536149

ABSTRACT

Activating fetal liver tyrosine kinase 3 (Flt3) mutations represent the most common genetic aberrations in acute myeloid leukemia (AML). Most commonly, they occur as internal tandem duplications in the juxtamembrane domain (Flt3-ITD) that transform myeloid cells in vitro and in vivo and that induce aberrant signaling and biologic functions. We identified RGS2, a regulator of G-protein signaling, as a gene specifically repressed by Flt3-ITD. Here we demonstrate an important role of RGS2 in Flt3-ITD-mediated transformation. RGS2 was repressed after forced expression of activating Flt3 mutations in 2 myeloid cell lines (32Dcl3 and NB4). Furthermore, RGS2 was repressed in Flt3-mutation-positive AML cases in comparison to Flt3-mutation-negative cases, especially in Flt3-ITD-positive cases with a high ITD-to-wild-type (WT) ratio. Coexpression of RGS2 with Flt3-ITD inhibited Flt3-ITD-induced autonomous proliferation and clonal growth of 32D cells. RGS2 also inhibited Flt3-ITD-induced phosphorylation of Akt and glycogen synthase kinase beta (Gsk3-beta) without influencing signal transducer and activator of transcription 5 (STAT5) activation. In addition, RGS2 reinduced the expression of Flt3-ITD-repressed CCAAT/enhancer-binding protein alpha (c/EBPalpha) and antagonized the Flt3-ITD-induced differentiation block in 32D cells. Expression analyses in myeloid cell lines revealed induction of RGS2 during granulocytic but not during monocytic differentiation. Taken together, RGS2 is a novel mediator of myeloid differentiation, and its repression is an important event in Flt3-ITD-induced transformation.


Subject(s)
Cell Transformation, Neoplastic , Leukemia, Myeloid/genetics , Mutation , Myeloid Cells/pathology , Proto-Oncogene Proteins/genetics , RGS Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Acute Disease , Cell Differentiation , Cell Line , Cell Proliferation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HL-60 Cells , Humans , Leukemia, Myeloid/etiology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Repressor Proteins , Tandem Repeat Sequences , fms-Like Tyrosine Kinase 3
SELECTION OF CITATIONS
SEARCH DETAIL
...