Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Biol Chem ; 290(30): 18343-60, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26037925

ABSTRACT

Hypusine modification of the eukaryotic initiation factor 5A (eIF-5A) is emerging as a crucial regulator in cancer, infections, and inflammation. Although its contribution in translational regulation of proline repeat-rich proteins has been sufficiently demonstrated, its biological role in higher eukaryotes remains poorly understood. To establish the hypusine modification system as a novel platform for therapeutic strategies, we aimed to investigate its functional relevance in mammals by generating and using a range of new knock-out mouse models for the hypusine-modifying enzymes deoxyhypusine synthase and deoxyhypusine hydroxylase as well as for the cancer-related isoform eIF-5A2. We discovered that homozygous depletion of deoxyhypusine synthase and/or deoxyhypusine hydroxylase causes lethality in adult mice with different penetrance compared with haploinsufficiency. Network-based bioinformatic analysis of proline repeat-rich proteins, which are putative eIF-5A targets, revealed that these proteins are organized in highly connected protein-protein interaction networks. Hypusine-dependent translational control of essential proteins (hubs) and protein complexes inside these networks might explain the lethal phenotype observed after deletion of hypusine-modifying enzymes. Remarkably, our results also demonstrate that the cancer-associated isoform eIF-5A2 is dispensable for normal development and viability. Together, our results provide the first genetic evidence that the hypusine modification in eIF-5A is crucial for homeostasis in mammals. Moreover, these findings highlight functional diversity of the hypusine system compared with lower eukaryotes and indicate eIF-5A2 as a valuable and safe target for therapeutic intervention in cancer.


Subject(s)
Lysine/analogs & derivatives , Mixed Function Oxygenases/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Peptide Initiation Factors/metabolism , Animals , Homeostasis/genetics , Humans , Lysine/genetics , Lysine/metabolism , Mice , Mice, Knockout , Mixed Function Oxygenases/metabolism , Neoplasms/genetics , Neoplasms/pathology , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Peptide Initiation Factors/genetics , Protein Biosynthesis , Protein Interaction Maps , Protein Processing, Post-Translational
2.
Dis Model Mech ; 7(8): 963-76, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24832488

ABSTRACT

The central importance of translational control by post-translational modification has spurred major interest in regulatory pathways that control translation. One such pathway uniquely adds hypusine to eukaryotic initiation factor 5A (eIF5A), and thereby affects protein synthesis and, subsequently, cellular proliferation through an unknown mechanism. Using a novel conditional knockout mouse model and a Caenorhabditis elegans knockout model, we found an evolutionarily conserved role for the DOHH-mediated second step of hypusine synthesis in early embryonic development. At the cellular level, we observed reduced proliferation and induction of senescence in 3T3 Dohh-/- cells as well as reduced capability for malignant transformation. Furthermore, mass spectrometry showed that deletion of DOHH results in an unexpected complete loss of hypusine modification. Our results provide new biological insight into the physiological roles of the second step of the hypusination of eIF5A. Moreover, the conditional mouse model presented here provides a powerful tool for manipulating hypusine modification in a temporal and spatial manner, to analyse both how this unique modification normally functions in vivo as well as how it contributes to different pathological conditions.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Embryonic Development , Lysine/analogs & derivatives , Mixed Function Oxygenases/antagonists & inhibitors , 3T3 Cells , Alleles , Animals , Caenorhabditis elegans , Cell Proliferation , Cellular Senescence , Disease Models, Animal , Embryo Loss/metabolism , Embryo Loss/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Knockout Techniques , Hydroxylation , Lysine/metabolism , Mice , Mixed Function Oxygenases/metabolism , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Peptide Initiation Factors/metabolism , Phenotype , Protein Biosynthesis , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins/metabolism , ras Proteins/metabolism , Eukaryotic Translation Initiation Factor 5A
3.
Drug Discov Today Technol ; 11: 89-99, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24847658

ABSTRACT

Resistance against tyrosine kinase inhibitors (TKIs) represents a relevant clinical problem in treatment of chronic myelogenous leukemia (CML). On the basis of their activity against the spectrum of BCR-ABL mutations that have shown to be the most prominent mechanism of resistance to imatinib, new TKIs have been classified as second generation (such as nilotinib, dasatinib and bosutinib) or third generation (also cover- ing T315I such as ponatinib) TKIs. However, mutations in BCR-ABL only account for about half of the cases of treatment failure under TKI and other mechanisms either rendering the leukemic cells still dependent of BCR-ABL activity or supporting oncogenic properties of the leukemic cells independent of BCR-ABL signaling have been identified. A detailed understanding of the different underlying resistance mechanisms will be the prerequisite to eventually overcome clinical resistance and for the successful use of tailored combinations of targeted inhibitors in the future.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/therapeutic use , Humans
4.
J Proteomics ; 96: 300-13, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24269351

ABSTRACT

We analysed the effects of all-trans retinoic acid (ATRA) on proliferation and changes in the global proteome of the nullipotent human embryonal carcinoma cell line 2102Ep and the pluripotent cell line NTERA2 cl.D1 (NT2). Differentially expressed proteins were assessed by 2D-PAGE and mass spectrometry, followed by verification and analysis of protein modifications of proteins of the retinoid pathway. We established a proteome map of the germ cell tumor (GCT) cell line NT2 showing neuronal differentiation under ATRA treatment for 7days. Using bioinformatic analyses, we identified functional groups of altered proteins and potentially involved pathways, of which changes to the organization of the cytoskeleton and anti-apoptotic effects were the most prominent. Changes observed in the expression of factors involved in the retinoid pathway under ATRA, namely an upregulation of CRBP and CRABP2, were also reflected in GCT tissues of different histologies, providing further insight into factors involved in the differentiation of these pluripotent tumors. BIOLOGICAL SIGNIFICANCE: Treatment of NT2 germ cell tumor cells with all-trans retinoic acid (ATRA) is a model to investigate differentiation. We analysed differentially expressed proteins by 2D-PAGE and mass spectrometry and provide a proteome map of NT2 cells under 7days of ATRA. By bioinformatic analyses, functional groups of proteins and involved pathways like changes to the cytoskeleton and anti-apoptotic effects were identified. Factors involved in the retinoid pathway, in particular upregulation of CRBP, CRABP1 and CRABP2, also showed differential expression in tumors with different histological subtypes, which provides insight into gene regulation under induced and spontaneous differentiation in germ cell tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Embryonal/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/biosynthesis , Protein Processing, Post-Translational/drug effects , Proteome/biosynthesis , Tretinoin/pharmacology , Carcinoma, Embryonal/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Humans , Time Factors , Up-Regulation/drug effects
5.
Kidney Int ; 84(3): 532-44, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23615505

ABSTRACT

The role of parietal epithelial cells (PECs) in glomerular disease is unclear because they also express podocyte proteins under pathophysiological conditions. To help resolve this, we established a novel PEC isolation technique in rats and mice to investigate which regulatory mechanisms lead to podocyte protein expression in PECs. This pure pool of naive PECs was then compared with PECs in primary culture and immortalized PECs in permanent culture. The naive PECs expressed low levels of podocyte-specific mRNA. Accordingly, in crescentic glomerulonephritis, single PECs activated the podocin promoter in vivo. In primary culture, PECs expressed a distinct morphology from podocytes but with high transcript and protein levels of PEC markers. In contrast to naive PECs, cultured PECs also expressed podocyte proteins, and this correlated with reduced proteolytic activity but not with increased transcript levels. Activation of autophagy or proteasomal degradation decreased the levels of podocyte proteins in PECs, whereas inhibition of proteasomal degradation led to the stabilization of podocyte proteins in PECs. Thus, naive PECs express podocyte transcripts physiologically and these podocyte proteins are stable under pathological conditions through decreased proteolysis.


Subject(s)
Desmin/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Podocytes/metabolism , Proteolysis , Sialoglycoproteins/metabolism , Animals , Autophagy/physiology , Cells, Cultured , Disease Models, Animal , Female , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Podocytes/pathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Transcriptome/physiology
6.
PLoS One ; 8(1): e53668, 2013.
Article in English | MEDLINE | ID: mdl-23326482

ABSTRACT

In drug discovery, the characterisation of the precise modes of action (MoA) and of unwanted off-target effects of novel molecularly targeted compounds is of highest relevance. Recent approaches for identification of MoA have employed various techniques for modeling of well defined signaling pathways including structural information, changes in phenotypic behavior of cells and gene expression patterns after drug treatment. However, efficient approaches focusing on proteome wide data for the identification of MoA including interference with mutations are underrepresented. As mutations are key drivers of drug resistance in molecularly targeted tumor therapies, efficient analysis and modeling of downstream effects of mutations on drug MoA is a key to efficient development of improved targeted anti-cancer drugs. Here we present a combination of a global proteome analysis, reengineering of network models and integration of apoptosis data used to infer the mode-of-action of various tyrosine kinase inhibitors (TKIs) in chronic myeloid leukemia (CML) cell lines expressing wild type as well as TKI resistance conferring mutants of BCR-ABL. The inferred network models provide a tool to predict the main MoA of drugs as well as to grouping of drugs with known similar kinase inhibitory activity patterns in comparison to drugs with an additional MoA. We believe that our direct network reconstruction approach, demonstrated on proteomics data, can provide a complementary method to the established network reconstruction approaches for the preclinical modeling of the MoA of various types of targeted drugs in cancer treatment. Hence it may contribute to the more precise prediction of clinically relevant on- and off-target effects of TKIs.


Subject(s)
Models, Biological , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Proteomics/methods , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Benzamides/pharmacology , Benzamides/therapeutic use , Blotting, Western , Cell Line, Tumor , Cluster Analysis , Drug Resistance, Neoplasm/drug effects , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Neoplasm Proteins/metabolism , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use
7.
Leuk Res ; 37(3): 280-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23137523

ABSTRACT

Secondary chromosomal aberrations may contribute to the development of a malignant phenotype in mantle cell lymphoma. Chromosomal band 5p15.33 represents a new recurrent breakpoint in B-cell malignancies. We present a molecular cytogenetic study of 8 mantle cell lymphoma (MCL) cell lines and 23 patients with MCL to determine and characterize novel secondary aberrations. We detected new secondary recurrent rearrangements in all cell lines and in 7 patients and confirmed 5p15.33 as a recurrent breakpoint in 4 cell lines and one patient. Further molecular characterization by flow-FISH and quantitative RT-PCR suggest TERT and CLPTM1L as target genes of 5p15.33 rearrangements.


Subject(s)
Chromosome Banding , Chromosome Breakage , Chromosomes, Human, Pair 5/genetics , Lymphoma, Mantle-Cell/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Telomerase/genetics , Aged , Cell Line, Tumor , DNA Mutational Analysis , Female , Gene Frequency , Genetic Loci/physiology , Humans , In Situ Hybridization, Fluorescence , Lymphoma, Mantle-Cell/epidemiology , Male , Middle Aged , Oncogene Proteins, Fusion/genetics , Prevalence , Reverse Transcriptase Polymerase Chain Reaction , Translocation, Genetic/physiology
8.
Cancer Lett ; 326(2): 161-7, 2012 Dec 30.
Article in English | MEDLINE | ID: mdl-22902505

ABSTRACT

3-Hydroxiglutaric acid (3-OH-GA) is a disease-specific metabolite that accumulates in tissues and body fluids of patients with Glutaric aciduria type I (GAI) and has been associated with vascular abnormalities in these kindreds. Here, we demonstrate that 3-OH-GA also affects the integrity of tumor vessels leading to tumor growth inhibition in a subcutaneous model of human glioblastoma multiforme (GBM). This effect correlated with a marked decrease of VE-Cadherin expression in endothelium of 3-OH-GA-treated tumors. Furthermore, in vitro observations indicated also a direct effect of 3-OH-GA in glioma cells that showed defective mitosis and significant proliferation inhibition. In summary, the GAI-specific metabolite 3-OH-GA significantly inhibited growth of GBM xenografts by affecting the structural integrity of tumor blood vessels and in addition by causing defective mitosis and proliferation inhibition of tumor cells.


Subject(s)
Cell Division/drug effects , Glioblastoma/pathology , Glutarates/pharmacology , Neovascularization, Pathologic , Animals , Blood Vessels/drug effects , Cell Line, Tumor , DNA, Complementary , Glioblastoma/blood supply , Humans , Immunohistochemistry , Mice , Mice, Nude , Oligonucleotide Array Sequence Analysis
9.
Mol Cell Proteomics ; 11(11): 1289-305, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22888148

ABSTRACT

Hypusine modification of eukaryotic initiation factor 5A (eIF-5A) represents a unique and highly specific post-translational modification with regulatory functions in cancer, diabetes, and infectious diseases. However, the specific cellular pathways that are influenced by the hypusine modification remain largely unknown. To globally characterize eIF-5A and hypusine-dependent pathways, we used an approach that combines large-scale bioreactor cell culture with tandem affinity purification and mass spectrometry: "bioreactor-TAP-MS/MS." By applying this approach systematically to all four components of the hypusine modification system (eIF-5A1, eIF-5A2, DHS, and DOHH), we identified 248 interacting proteins as components of the cellular hypusine network, with diverse functions including regulation of translation, mRNA processing, DNA replication, and cell cycle regulation. Network analysis of this data set enabled us to provide a comprehensive overview of the protein-protein interaction landscape of the hypusine modification system. In addition, we validated the interaction of eIF-5A with some of the newly identified associated proteins in more detail. Our analysis has revealed numerous novel interactions, and thus provides a valuable resource for understanding how this crucial homeostatic signaling pathway affects different cellular functions.


Subject(s)
Lysine/analogs & derivatives , Protein Interaction Maps , Protein Processing, Post-Translational , Animals , Computational Biology , DNA-Binding Proteins/metabolism , Humans , Lysine/metabolism , Mass Spectrometry , Mice , Mixed Function Oxygenases/metabolism , Multivesicular Bodies/metabolism , NIH 3T3 Cells , Nuclear Proteins/metabolism , Nucleophosmin , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Peptide Fragments/metabolism , Peptide Initiation Factors/metabolism , Protein Transport , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Ribosomal Proteins/metabolism , Subcellular Fractions/metabolism , Eukaryotic Translation Initiation Factor 5A
10.
PLoS One ; 7(8): e43468, 2012.
Article in English | MEDLINE | ID: mdl-22927971

ABSTRACT

Glioblastomas are highly aggressive brain tumors of adults with poor clinical outcome. Despite a broad range of new and more specific treatment strategies, therapy of glioblastomas remains challenging and tumors relapse in all cases. Recent work demonstrated that the posttranslational hypusine modification of the eukaryotic initiation factor 5A (eIF-5A) is a crucial regulator of cell proliferation, differentiation and an important factor in tumor formation, progression and maintenance. Here we report that eIF-5A as well as the hypusine-forming enzymes deoxyhypusine synthase (DHS) and deoxyhypusine hydroxylase (DOHH) are highly overexpressed in glioblastoma patient samples. Importantly, targeting eIF-5A and its hypusine modification with GC7, a specific DHS-inhibitor, showed a strong antiproliferative effect in glioblastoma cell lines in vitro, while normal human astrocytes were not affected. Furthermore, we identified p53 dependent premature senescence, a permanent cell cycle arrest, as the primary outcome in U87-MG cells after treatment with GC7. Strikingly, combined treatment with clinically relevant alkylating agents and GC7 had an additive antiproliferative effect in glioblastoma cell lines. In addition, stable knockdown of eIF-5A and DHS by short hairpin RNA (shRNA) could mimic the antiproliferative effects of GC7. These findings suggest that pharmacological inhibition of eIF-5A may represent a novel concept to treat glioblastomas and may help to substantially improve the clinical course of this tumor entity.


Subject(s)
Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Lysine/analogs & derivatives , Mixed Function Oxygenases/genetics , Molecular Targeted Therapy , Oxidoreductases Acting on CH-NH Group Donors/genetics , Peptide Initiation Factors/genetics , RNA-Binding Proteins/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Carmustine/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glioblastoma/drug therapy , Glioblastoma/enzymology , Glioblastoma/pathology , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Lysine/biosynthesis , Male , Mixed Function Oxygenases/metabolism , Neoplasm Grading , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-NH Group Donors/deficiency , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Peptide Initiation Factors/deficiency , Temozolomide , Eukaryotic Translation Initiation Factor 5A
11.
Invest New Drugs ; 30(6): 2274-83, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22415796

ABSTRACT

Effective inhibition of BCR-ABL tyrosine kinase activity with Imatinib represents a breakthrough in the treatment of patients with chronic myeloid leukemia (CML). However, more than 30 % of patients with CML in chronic phase do not respond adequately to Imatinib and the drug seems not to affect the quiescent pool of BCR-ABL positive leukemic stem and progenitor cells. Therefore, despite encouraging clinical results, Imatinib can still not be considered a curative treatment option in CML. We recently reported downregulation of eukaryotic initiation factor 5A (eIF5A) in Imatinib treated K562 cells. Furthermore, the inhibition of eIF5A by siRNA in combination with Imatinib has been shown to exert synergistic cytotoxic effects on BCR-ABL positive cell lines. Based on the structure of known deoxyhypusine synthase (DHS) inhibitors such as CNI-1493, a drug design approach was applied to develop potential compounds targeting DHS. Here we report the biological evaluation of selected novel (DHSI-15) as compared to established (CNI-1493, deoxyspergualin) DHS inhibitors. We show that upon the compounds tested, DHSI-15 and deoxyspergualin exert strongest antiproliferative effects on BCR-ABL cells including Imatinib resistant mutants. However, this effect did not seem to be restricted to BCR-ABL positive cell lines or primary cells. Both compounds are able to induce apoptosis/necrosis during long term incubation of BCR-ABL positive BA/F3 derivates. Pharmacological synergism can be observed for deoxyspergualin and Imatinib, but not for DHSI-15 and Imatinib. Finally we show that deoxyspergualin is able to inhibit proliferation of CD34+ progenitor cells from CML patients. We conclude that inhibition of deoxyhypusine synthase (DHS) can be supportive for the anti-proliferative treatment of leukemia and merits further investigation including other cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Animals , Antigens, CD34 , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Guanidines/pharmacology , Humans , Hydrazones/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mice
12.
Mol Cancer ; 10: 129, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21999842

ABSTRACT

BACKGROUND: We have previously reported significant downregulation of ubiquitin carboxyl-terminal hydrolase 1 (UCHL1) in prostate cancer (PCa) compared to the surrounding benign tissue. UCHL1 plays an important role in ubiquitin system and different cellular processes such as cell proliferation and differentiation. We now show that the underlying mechanism of UCHL1 downregulation in PCa is linked to its promoter hypermethylation. Furthermore, we present evidences that UCHL1 expression can affect the behavior of prostate cancer cells in different ways. RESULTS: Methylation specific PCR analysis results showed a highly methylated promoter region for UCHL1 in 90% (18/20) of tumor tissue compared to 15% (3/20) of normal tissues from PCa patients. Pyrosequencing results confirmed a mean methylation of 41.4% in PCa whereas only 8.6% in normal tissues. To conduct functional analysis of UCHL1 in PCa, UCHL1 is overexpressed in LNCaP cells whose UCHL1 expression is normally suppressed by promoter methylation and found that UCHL1 has the ability to decrease the rate of cell proliferation and suppresses anchorage-independent growth of these cells. In further analysis, we found evidence that exogenous expression of UCHL1 suppress LNCaP cells growth probably via p53-mediated inhibition of Akt/PKB phosphorylation and also via accumulation of p27kip1 a cyclin dependant kinase inhibitor of cell cycle regulating proteins. Notably, we also observed that exogenous expression of UCHL1 induced a senescent phenotype that was detected by using the SA-ß-gal assay and might be due to increased p14ARF, p53, p27kip1 and decreased MDM2. CONCLUSION: From these results, we propose that UCHL1 downregulation via promoter hypermethylation plays an important role in various molecular aspects of PCa biology, such as morphological diversification and regulation of proliferation.


Subject(s)
DNA Methylation , Gene Silencing , Promoter Regions, Genetic , Prostatic Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Cell Adhesion , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Cellular Senescence , Cyclin A/genetics , Cyclin A/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Male , Phosphorylation , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Sequence Analysis, DNA , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitination
13.
J Exp Med ; 208(11): 2163-74, 2011 Oct 24.
Article in English | MEDLINE | ID: mdl-21911423

ABSTRACT

Chronic myeloid leukemia (CML) is induced by the oncogenic BCR-ABL1 tyrosine kinase and can be effectively treated for many years with tyrosine kinase inhibitors (TKIs). However, unless CML patients receive life-long TKI treatment, leukemia will eventually recur; this is attributed to the failure of TKI treatment to eradicate leukemia-initiating cells (LICs). Recent work demonstrated that FoxO factors are critical for maintenance of CML-initiating cells; however, the mechanism of FoxO-dependent leukemia initiation remained elusive. Here, we identified the BCL6 protooncogene as a critical effector downstream of FoxO in self-renewal signaling of CML-initiating cells. BCL6 represses Arf and p53 in CML cells and is required for colony formation and initiation of leukemia. Importantly, peptide inhibition of BCL6 in human CML cells compromises colony formation and leukemia initiation in transplant recipients and selectively eradicates CD34(+) CD38(-) LICs in patient-derived CML samples. These findings suggest that pharmacological inhibition of BCL6 may represent a novel strategy to eradicate LICs in CML. Clinical validation of this concept could limit the duration of TKI treatment in CML patients, which is currently life-long, and substantially decrease the risk of blast crisis transformation.


Subject(s)
Cell Survival , DNA-Binding Proteins/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Neoplastic Stem Cells/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Antigens, CD34/metabolism , Benzamides , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neoplasm Transplantation , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-6 , Pyrimidines/therapeutic use , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
14.
Nature ; 473(7347): 384-8, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21593872

ABSTRACT

Tyrosine kinase inhibitors (TKIs) are widely used to treat patients with leukaemia driven by BCR-ABL1 (ref. 1) and other oncogenic tyrosine kinases. Recent efforts have focused on developing more potent TKIs that also inhibit mutant tyrosine kinases. However, even effective TKIs typically fail to eradicate leukaemia-initiating cells (LICs), which often cause recurrence of leukaemia after initially successful treatment. Here we report the discovery of a novel mechanism of drug resistance, which is based on protective feedback signalling of leukaemia cells in response to treatment with TKI. We identify BCL6 as a central component of this drug-resistance pathway and demonstrate that targeted inhibition of BCL6 leads to eradication of drug-resistant and leukaemia-initiating subclones.


Subject(s)
DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/antagonists & inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , ADP-Ribosylation Factor 1/metabolism , Animals , Cell Survival/drug effects , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-bcl-6 , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
15.
PLoS One ; 6(4): e19164, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21541334

ABSTRACT

The success of Imatinib (IM) therapy in chronic myeloid leukemia (CML) is compromised by the development of IM resistance and by a limited IM effect on hematopoietic stem cells. Danusertib (formerly PHA-739358) is a potent pan-aurora and ABL kinase inhibitor with activity against known BCR-ABL mutations, including T315I. Here, the individual contribution of both signaling pathways to the therapeutic effect of Danusertib as well as mechanisms underlying the development of resistance and, as a consequence, strategies to overcome resistance to Danusertib were investigated. Starting at low concentrations, a dose-dependent inhibition of BCR-ABL activity was observed, whereas inhibition of aurora kinase activity required higher concentrations, pointing to a therapeutic window between the two effects. Interestingly, the emergence of resistant clones during Danusertib exposure in vitro occurred considerably less frequently than with comparable concentrations of IM. In addition, Danusertib-resistant clones had no mutations in BCR-ABL or aurora kinase domains and remained IM-sensitive. Overexpression of Abcg2 efflux transporter was identified and functionally validated as the predominant mechanism of acquired Danusertib resistance in vitro. Finally, the combined treatment with IM and Danusertib significantly reduced the emergence of drug resistance in vitro, raising hope that this drug combination may also achieve more durable disease control in vivo.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Benzamides/pharmacology , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/metabolism , Neoplasm Proteins/genetics , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Apoptosis/drug effects , Aurora Kinases , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Clone Cells , Drug Resistance, Neoplasm/genetics , Drug Synergism , Fusion Proteins, bcr-abl/antagonists & inhibitors , Gene Expression Regulation, Leukemic/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mutation/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Piperazines/pharmacology , Polyploidy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects
16.
Exp Hematol ; 39(1): 66-76.e1-2, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20940029

ABSTRACT

OBJECTIVE: In chronic myeloid leukemia (CML), increased cellular turnover of hematopoietic cells driven by the oncogene BCR-ABL leads to accelerated telomere shortening despite increased telomerase activity. It has been postulated that shortened telomeres, particularly in the context of increased telomerase activity, might facilitate accumulation of genetic aberrations and, consequently, disease progression from chronic phase to accelerated phase and blast crisis. Therefore, inhibition of telomerase might be a promising approach in CML therapy. MATERIAL AND METHODS: To investigate the therapeutic potential of telomerase inhibition in this model disorder, we used a small molecule telomerase inhibitor, BIBR1532 as well as expression of a dominant-negative mutant of hTERT (DNhTERT-IRES-GFP) in the p53-negative CML blast crisis cell line K562 and characterized the effects in long-term culture. Furthermore, we expressed an inducible p53 construct (vector pBabe-p53ER(tam)) via retroviral transduction in cells with critically short telomeres and in cells with a normal telomere length to explain the role of the tumor suppressor in response to critical telomere shortening in BCR-ABL-positive cells. RESULTS: BIBR1532-treated bulk cultures did not show altered growth kinetics despite significant telomere shortening to a critical length of approximately 5 kb. In comparison, DNhTERT-expressing clones either lost telomere length, leading to a significant but transient slow down in proliferation but eventually all escaped senescence/crisis (group I) or, alternatively, remained virtually unaffected despite measurable telomerase inhibition (group II). Further analyses of group I clones revealed impaired DNA damage response and an accumulation of dicentric chromosomes. However, upon restoration of p53 in telomerase-negative K562 clones with critically short telomeres, immediate reinduction of apoptosis and complete eradication of cells was observed, whereas vector control cells continued to escape from crisis. CONCLUSIONS: These results suggest that the success of strategies aimed at telomerase inhibition in CML is highly dependent on the presence of functional p53 and should be explored preferentially in chronic phase CML.


Subject(s)
Genes, abl , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Telomerase/antagonists & inhibitors , Tumor Suppressor Protein p53/physiology , Apoptosis , Blotting, Western , Genomic Instability , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Telomere
17.
Neoplasia ; 11(9): 934-44, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19724687

ABSTRACT

Patients with advanced stages of hepatocellular carcinoma (HCC) face a poor prognosis. Although encouraging clinical results have been obtained with multikinase inhibitor sorafenib, the development of improved therapeutic strategies for HCC remains an urgent goal. Aurora kinases are key regulators of the cell cycle, and their uncontrolled expression promotes aneuploidy and tumor development. In tissue microarray analyses, we detected aurora-A kinase expression in all of the examined 93 human HCC samples, whereas aurora-B kinase expression levels significantly correlated with the proliferation index of HCCs. In addition, two human HCC cell lines (Huh-7 and HepG2) were tested positive for aurora-A and -B and revealed Ser10 phosphorylation of histone H3, indicating an increased aurora-B kinase activity. The antiproliferative features of a novel aurora kinase inhibitor, PHA-739358, currently under investigation in phase 2 clinical trials for other solid tumors, were examined in vitro and in vivo. At concentrations exceeding 50 nM, PHA-739358 completely suppressed tumor cell proliferation in cell culture experiments and strongly decreased histone H3 phosphorylation. Cell cycle inhibition and endoreduplication were observed at 50 nM, whereas higher concentrations led to a complete G(2)/M-phase arrest. In vivo, administration of PHA-739358 resulted in significant tumor growth inhibition at a well-tolerated dose. In combination with sorafenib, additive effects were observed. Remarkably, when tumors restarted to grow under sorafenib monotherapy, subsequent treatment with PHA-739358 induced tumor shrinkage by up to 81%. Thus, targeting aurora kinases with PHA-739358 is a promising therapeutic strategy administered alone or in combination with sorafenib for patients with advanced stages of HCC.


Subject(s)
Benzamides/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/therapeutic use , Xenograft Model Antitumor Assays , Animals , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Aurora Kinase A , Aurora Kinase B , Aurora Kinases , Benzenesulfonates/therapeutic use , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , G2 Phase/drug effects , Humans , Immunoenzyme Techniques , In Vitro Techniques , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Niacinamide/analogs & derivatives , Phenylurea Compounds , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Sorafenib , Tissue Array Analysis
18.
Hematol Oncol ; 27(3): 123-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19569255

ABSTRACT

Telomeres are specialized structures localized at the end of human chromosomes. Due to the end replication problem, each cell division results in a loss of telomeric repeats in normal somatic cells. In germ line and stem cells, the multicomponent enzyme telomerase maintains the length of telomere repeats. However, elevated telomerase activity has also been reported in the majority of solid tumours as well as in acute and chronic leukaemia. Chronic myeloid leukaemia (CML) serves as a model disease to study telomere biology in clonal myeloproliferative disorders. In CML, telomere shortening correlates with disease stage, duration of chronic phase (CP), prognosis measured by the Hasford risk score and the response to disease-modifying therapeutics such as the tyrosine kinase inhibitor Imatinib. In addition, telomerase activity (TA) is already increased in CP CML and further upregulated with disease progression to accelerated phase and blast crisis (BC). Furthermore, a correlation of TA with increased genetic instability as well as a shorter survival of the patients has been reported. Here, we review the current state of knowledge of the role of telomere and telomerase biology in CML and discuss the possible impact of novel treatment approaches.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Telomerase/antagonists & inhibitors , Telomerase/metabolism , Telomere/pathology , Animals , Disease Progression , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Telomerase/genetics
19.
Cancer Res ; 66(6): 2881-4, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16540631

ABSTRACT

Cellular senescence, a permanent cell cycle arrest, is considered a safeguard mechanism that may prevent aged or abnormal cells from further expansion. Although the term "replicative senescence" stands for the widely accepted model of a terminal growth arrest due to telomere attrition, the significance of "oncogene-inducible senescence" remained an issue of debate over the years. A number of recent studies now show the effect of this acute and telomere-independent form of senescence as a tumor-protective, fail-safe mechanism in vivo that shares conceptual and possibly therapeutic similarities with the genetically encoded apoptosis machinery.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cellular Senescence/genetics , Oncogenes/physiology , Animals , Humans , Precancerous Conditions/genetics
20.
Nature ; 436(7051): 660-5, 2005 Aug 04.
Article in English | MEDLINE | ID: mdl-16079837

ABSTRACT

Acute induction of oncogenic Ras provokes cellular senescence involving the retinoblastoma (Rb) pathway, but the tumour suppressive potential of senescence in vivo remains elusive. Recently, Rb-mediated silencing of growth-promoting genes by heterochromatin formation associated with methylation of histone H3 lysine 9 (H3K9me) was identified as a critical feature of cellular senescence, which may depend on the histone methyltransferase Suv39h1. Here we show that Emicro-N-Ras transgenic mice harbouring targeted heterozygous lesions at the Suv39h1, or the p53 locus for comparison, succumb to invasive T-cell lymphomas that lack expression of Suv39h1 or p53, respectively. By contrast, most N-Ras-transgenic wild-type ('control') animals develop a non-lymphoid neoplasia significantly later. Proliferation of primary lymphocytes is directly stalled by a Suv39h1-dependent, H3K9me-related senescent growth arrest in response to oncogenic Ras, thereby cancelling lymphomagenesis at an initial step. Suv39h1-deficient lymphoma cells grow rapidly but, unlike p53-deficient cells, remain highly susceptible to adriamycin-induced apoptosis. In contrast, only control, but not Suv39h1-deficient or p53-deficient, lymphomas senesce after drug therapy when apoptosis is blocked. These results identify H3K9me-mediated senescence as a novel Suv39h1-dependent tumour suppressor mechanism whose inactivation permits the formation of aggressive but apoptosis-competent lymphomas in response to oncogenic Ras.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cellular Senescence , Genes, ras/genetics , Lymphoma/genetics , Lymphoma/pathology , ADP-Ribosylation Factors/metabolism , Animals , Apoptosis/drug effects , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/metabolism , Cellular Senescence/drug effects , Cellular Senescence/genetics , Chromosomal Instability/genetics , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Heterochromatin/genetics , Heterochromatin/metabolism , Lymphoma/metabolism , Methylation , Methyltransferases/deficiency , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Transgenic , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transgenes/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...