Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Leukemia ; 29(10): 1981-92, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25971362

ABSTRACT

High levels of microRNA-155 (miR-155) are associated with poor outcome in acute myeloid leukemia (AML). In AML, miR-155 is regulated by NF-κB, the activity of which is, in part, controlled by the NEDD8-dependent ubiquitin ligases. We demonstrate that MLN4924, an inhibitor of NEDD8-activating enzyme presently being evaluated in clinical trials, decreases binding of NF-κB to the miR-155 promoter and downregulates miR-155 in AML cells. This results in the upregulation of the miR-155 targets SHIP1, an inhibitor of the PI3K/Akt pathway, and PU.1, a transcription factor important for myeloid differentiation, leading to monocytic differentiation and apoptosis. Consistent with these results, overexpression of miR-155 diminishes MLN4924-induced antileukemic effects. In vivo, MLN4924 reduces miR-155 expression and prolongs the survival of mice engrafted with leukemic cells. Our study demonstrates the potential of miR-155 as a novel therapeutic target in AML via pharmacologic interference with NF-κB-dependent regulatory mechanisms. We show the targeting of this oncogenic microRNA with MLN4924, a compound presently being evaluated in clinical trials in AML. As high miR-155 levels have been consistently associated with aggressive clinical phenotypes, our work opens new avenues for microRNA-targeting therapeutic approaches to leukemia and cancer patients.


Subject(s)
Cyclopentanes/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , Pyrimidines/pharmacology , Tandem Repeat Sequences/genetics , Ubiquitins/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , Animals , Apoptosis/drug effects , Blotting, Western , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , NEDD8 Protein , NF-kappa B/genetics , NF-kappa B/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Leukemia ; 27(10): 1996-2005, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23670294

ABSTRACT

The dismal outcome of blast crisis chronic myelogenous leukemia (CML-BC) patients underscores the need for a better understanding of the mechanisms responsible for the development of drug resistance. Altered expression of the anti-apoptoticBcl-xL has been correlated with BCR-ABL leukemogenesis; however, its involvement in the pathogenesis and evolution of CML has not been formally demonstrated yet. Thus, we generated an inducible mouse model in which simultaneous expression of p210-BCR-ABL1 and deletion of bcl-x occurs within hematopoietic stem and progenitor cells. Absence of Bcl-xL did not affect development of the chronic phase-like myeloproliferative disease, but none of the deficient mice progressed to an advanced phenotype, suggesting the importance of Bcl-xL in survival of progressing early progenitor cells. Indeed, pharmacological antagonism of Bcl-xL, with ABT-263, combined with PP242-induced activation of BAD markedly augmented apoptosis of CML-BC cell lines and primary CD34(+) progenitors but not those from healthy donors, regardless of drug resistance induced by bone marrow stromal cell-generated signals. Moreover, studies in which BAD or Bcl-xL expression was molecularly altered strongly support their involvement in ABT-263/PP242-induced apoptosis of CML-BC progenitors. Thus, suppression of the antiapoptotic potential of Bcl-xL together with BAD activation represents an effective pharmacological approach for patients undergoing blastic transformation.


Subject(s)
Apoptosis/drug effects , Blast Crisis/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Stem Cells/pathology , bcl-X Protein/antagonists & inhibitors , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Blast Crisis/drug therapy , Blast Crisis/genetics , Disease Progression , Female , Flow Cytometry , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/genetics , Humans , Indoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Male , Mice , Mice, Transgenic , Purines/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Sulfonamides/pharmacology , bcl-X Protein/metabolism
3.
Leukemia ; 27(4): 871-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23178755

ABSTRACT

Histone deacetylase (HDAC) inhibitors either alone or in combination with hypomethylating agents have limited clinical effect in acute myeloid leukemia (AML). Previously, we demonstrated that AML patients with higher miR (microRNA)-29b expression had better response to the hypomethylating agent decitabine. Therefore, an increase in miR-29b expression preceding decitabine treatment may provide a therapeutic advantage. We previously showed that miR-29b expression is suppressed by a repressor complex that includes HDACs. Thus, HDAC inhibition may increase miR-29b expression. We hypothesized that priming AML cells with the novel HDAC inhibitor (HDACI) AR-42 would result in increased response to decitabine treatment via upregulation of miR-29b. Here, we show that AR-42 is a potent HDACI in AML, increasing miR-29b levels and leading to downregulation of known miR-29b targets (that is, SP1, DNMT1, DNMT3A and DNMT3B). We then demonstrated that the sequential administration of AR-42 followed by decitabine resulted in a stronger anti-leukemic activity in vitro and in vivo than decitabine followed by AR-42 or either drug alone. These preclinical results with AR-42 priming before decitabine administration represent a promising, novel treatment approach and a paradigm shift with regard to the combination of epigenetic-targeting compounds in AML, where decitabine has been traditionally given before HDACIs.


Subject(s)
Azacitidine/analogs & derivatives , Epigenesis, Genetic , Histone Deacetylase Inhibitors/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , MicroRNAs/genetics , Phenylbutyrates/therapeutic use , Animals , Azacitidine/therapeutic use , Blotting, Western , Cell Line, Tumor , Decitabine , Histone Deacetylases/metabolism , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Up-Regulation/drug effects
4.
Leuk Suppl ; 1(Suppl 2): S51-3, 2012 Aug.
Article in English | MEDLINE | ID: mdl-27175251

ABSTRACT

The introduction of Abl tyrosine kinase inhibitors (TKI; that is, imatinib, dasatinib and nilotinib) as front-line therapy completely changed the course of chronic myelogenous leukemia (CML) to the point that most of the TKI-responsive newly diagnosed CML patients can be considered 'clinically' cured and their progression into blast crisis (BC) a rare event. However, a therapy for those patients who transform is still lacking, and TKIs do not eradicate CML at the stem cell level, therefore leaving a reservoir of cancer stem cells in a dormant stage. Thus, it is not surprising that the focus of CML research has shifted significantly toward the dissection of the mechanisms regulating the survival and self-renewal of TKI-resistant Philadelphia-positive leukemic chronic phase and BC stem cells, with the ultimate goal of developing small molecules capable of selectively killing leukemic but not normal hematopoietic stem cells, thereby achieving a 'biological' cure for this disease.

5.
Leukemia ; 23(10): 1708-15, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19641523

ABSTRACT

Ph-positive chronic myeloid leukemia (CML) and Ph-negative chronic myeloproliferative diseases (MPDs), characterized in many cases by the presence of the JAK2(V617F) mutation, have many features in common and yet also show fundamental differences. In this review, we pose five discrete and related questions relevant to both categories of hematological malignancy, namely: What are the mechanisms that underlie disease progression from a relatively benign or chronic phase? By what therapeutic methods might one target residual leukemia stem cells in CML? Is JAK2(V617F) the original molecular event in MPD? What epigenetic events must have a role in dictating disease phenotype in MPDs? And finally, Will the benefits conferred by current or future JAK2(V617F) inhibitors equal or even surpass the clinical success that has resulted from the use of tyrosine kinase inhibitors in CML? These and others questions must be addressed and in some cases should be answered in the foreseeable future.


Subject(s)
Janus Kinase 2/genetics , Leukemia, Myeloid, Chronic-Phase/genetics , Myeloproliferative Disorders/genetics , Philadelphia Chromosome , Chronic Disease , Humans , Myeloproliferative Disorders/classification
6.
Oncogene ; 28(14): 1669-81, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19234487

ABSTRACT

Chronic myelogenous leukemia (CML) patients treated with imatinib mesylate (IM) become drug resistant by mutations within the kinase domain of Bcr-Abl, and by other changes that cause progression to advanced stage (blast crisis) and increased expression of the Lyn tyrosine kinase, the regulation of which is not understood yet. In Bcr-Abl+ cells inhibition of Jak2, a downstream target of Bcr-Abl, by either Jak2 inhibitors or Jak2-specific short interfering RNA (siRNA) reduced the level of the SET protein, and increased PP2A Ser/Thr phosphatase and Shp1 tyrosine phosphatase activities, which led to decreased levels of activated Lyn. Activation of PP2A combined with Jak2 inhibition enhanced the reduction of activated Lyn kinase compared with Jak2 inhibition alone. In contrast, inhibition of either PP2A or Shp1 combined with Jak2 inhibition interfered with the loss of Lyn kinase activation more so than Jak2 inhibition alone, indicating the involvement of PP2A and Shp1 in the inactivation of the Lyn kinase caused by Jak2 inhibition. Inhibition of Jak2 induced apoptosis and reduced colony formation in IM-sensitive and -resistant Bcr-Abl mutant cell lines. Jak2 inhibition also induced apoptosis in CML cells from blast crisis patients but not in normal hematopoietic cells. These results indicate that Lyn is downstream of Jak2, and Jak2 maintains activated Lyn kinase in CML through the SET-PP2A-Shp1 pathway.


Subject(s)
Apoptosis/drug effects , Chromosomal Proteins, Non-Histone/physiology , Janus Kinase 2/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Phosphatase 2/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , Transcription Factors/physiology , src-Family Kinases/metabolism , Animals , Benzamides , Cyclohexanes/pharmacology , DNA-Binding Proteins , Drug Resistance, Neoplasm , Histone Chaperones , Humans , Imatinib Mesylate , Janus Kinase 2/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Phosphorylation , Piperazines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Tyrphostins/pharmacology
8.
Br J Cancer ; 95(7): 775-81, 2006 Oct 09.
Article in English | MEDLINE | ID: mdl-16953242

ABSTRACT

The deregulated kinase activity of p210-BCR/ABL oncoproteins, hallmark of chronic myelogenous leukaemia (CML), induces and sustains the leukaemic phenotype, and contributes to disease progression. Imatinib mesylate, a BCR/ABL kinase inhibitor, is effective in most of chronic phase CML patients. However, a significant percentage of CML patients develop resistance to imatinib and/or still progresses to blast crisis, a disease stage that is often refractory to imatinib therapy. Furthermore, there is compelling evidence indicating that the CML leukaemia stem cell is also resistant to imatinib. Thus, there is still a need for new drugs that, if combined with imatinib, will decrease the rate of relapse, fully overcome imatinib resistance and prevent blastic transformation of CML. We recently reported that the activity of the tumour suppressor protein phosphatase 2A (PP2A) is markedly inhibited in blast crisis CML patient cells and that molecular or pharmacologic re-activation of PP2A phosphatase led to growth suppression, enhanced apoptosis, impaired clonogenic potential and decreased in vivo leukaemogenesis of imatinib-sensitive and -resistant (T315I included) CML-BC patient cells and/or BCR/ABL+ myeloid progenitor cell lines. Thus, the combination of PP2A phosphatase-activating and BCR/ABL kinase-inhibiting drugs may represent a powerful therapeutic strategy for blast crisis CML patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Blast Crisis/drug therapy , Phosphoprotein Phosphatases/administration & dosage , Piperazines/administration & dosage , Pyrimidines/administration & dosage , Signal Transduction/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols , Benzamides , Drug Resistance, Neoplasm/physiology , Genes, abl/drug effects , Humans , Imatinib Mesylate , Phosphoprotein Phosphatases/metabolism , Protein Phosphatase 2 , Proto-Oncogene Proteins c-bcr/drug effects , Signal Transduction/physiology
10.
Blood ; 96(12): 3915-21, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11090078

ABSTRACT

Growth factor-dependent hematopoietic cell lines expressing the BCR/ABL oncoprotein of the Ph chromosome show growth factor-independent proliferation and resistance to apoptosis. Apoptosis resistance of BCR/ABL-expressing cells may depend on enhanced expression of anti-apoptotic proteins as well as reduced expression and/or inactivation of pro-apoptotic proteins. Compared to myeloid precursor 32Dcl3 cells expressing wild type BCR/ABL, cells expressing a BCR/ABL mutant lacking amino acids 176-426 in the BCR domain (p185 delta BCR) are susceptible to apoptosis induced by interleukin-3 (IL-3) deprivation. These cells exhibited the hypophosphorylated apoptotic BAD and markedly reduced levels of Bcl-2. Upon ectopic expression of Bcl-2, these cells showed no changes in BAD phosphorylation, but they became apoptosis-resistant and proliferated in the absence of IL-3, albeit more slowly than cells expressing wild type BCR/ABL. Moreover, the p185 delta BCR/Bcl-2 double transfectants were leukemogenic when injected into immunodeficient mice, but Bcl-2 expression did not restore the leukemia-inducing effects of p185 delta BCR to the levels of wild type BCR/ABL. Leukemic cells recovered from the spleen of mice injected with p185 delta BCR/Bcl-2 cells did not show rearrangements in the Bcl-2 genomic locus, but they exhibited enhanced proliferation in culture and induced a rapidly fatal disease process when inoculated in secondary recipient mice. Together, these data support the importance of anti-apoptotic pathways for BCR/ABL-dependent leukemogenesis and suggest that Bcl-2 expression promotes secondary changes leading to a more aggressive tumor phenotype. (Blood. 2000;96:3915-3921)


Subject(s)
Proto-Oncogene Proteins c-bcl-2/pharmacology , Animals , Apoptosis/drug effects , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cell Line , Cell Transformation, Neoplastic/drug effects , Fusion Proteins, bcr-abl/adverse effects , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Experimental/etiology , Mice , Mice, SCID , Mutation , Neoplasm Transplantation , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Transfection , bcl-Associated Death Protein
11.
Mol Cell Biol ; 20(16): 6159-69, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10913197

ABSTRACT

The DNA binding activity of FUS (also known as TLS), a nuclear pro-oncogene involved in multiple translocations, is regulated by BCR-ABL in a protein kinase CbetaII (PKCbetaII)-dependent manner. We show here that in normal myeloid progenitor cells FUS, although not visibly ubiquitinated, undergoes proteasome-dependent degradation, whereas in BCR-ABL-expressing cells, degradation is suppressed by PKCbetaII phosphorylation. Replacement of serine 256 with the phosphomimetic aspartic acid prevents proteasome-dependent proteolysis of FUS, while the serine-256-to-alanine FUS mutant is unstable and susceptible to degradation. Ectopic expression of the phosphomimetic S256D FUS mutant in granulocyte colony-stimulating factor-treated 32Dcl3 cells induces massive apoptosis and inhibits the differentiation of the cells escaping cell death, while the degradation-prone S256A mutant has no effect on either survival or differentiation. FUS proteolysis is induced by c-Jun, is suppressed by BCR-ABL or Jun kinase 1, and does not depend on c-Jun transactivation potential, ubiquitination, or its interaction with Jun kinase 1. In addition, c-Jun-induced FUS proteasome-dependent degradation is enhanced by heterogeneous nuclear ribonucleoprotein (hnRNP) A1 and depends on the formation of a FUS-Jun-hnRNP A1-containing complex and on lack of PKCbetaII phosphorylation at serine 256 but not on FUS ubiquitination. Thus, novel mechanisms appear to be involved in the degradation of FUS in normal myeloid cells; moreover, the ability of the BCR-ABL oncoprotein to suppress FUS degradation by the induction of posttranslational modifications might contribute to the phenotype of BCR-ABL-expressing hematopoietic cells.


Subject(s)
Cysteine Endopeptidases/genetics , Fusion Proteins, bcr-abl/genetics , Genes, jun , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Multienzyme Complexes/genetics , Protein Kinase C/genetics , Ribonucleoproteins/genetics , Signal Transduction , Animals , Apoptosis , Cell Line , Cysteine Endopeptidases/metabolism , Enzyme Activation , Fusion Proteins, bcr-abl/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoproteins , Mice , Multienzyme Complexes/metabolism , Phosphorylation , Proteasome Endopeptidase Complex , Protein Kinase C/metabolism , RNA-Binding Protein FUS , Ribonucleoproteins/metabolism , Signal Transduction/genetics
12.
Exp Cell Res ; 257(2): 265-71, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10837140

ABSTRACT

DRnm23 belongs to a multigene family which includes nm23-H1, the first bona fide metastasis suppressor gene, nm23-H2, nm23-H4, and nm23-H5. Like nm23-H1, nm23-H2, and nm23-H4, DRnm23 possesses nucleoside diphosphate kinase (NDPK) activity. Upon overexpression in myeloid precursor 32Dcl3 cells, DRnm23 inhibits granulocytic differentiation and promotes apoptosis. Two specific mutants of DRnm23 (H134Q and S136P), at residues required for the NDPK activity, inhibit differentiation and promote apoptosis of 32Dcl3 cells. By contrast, substitution of serine 61 with proline (S61P) or deletion of the RGD domain (DeltaRGD) abrogates the effects of wild-type DRnm23. Like wild-type DRnm23, all four mutants show a predominantly mitochondrial subcellular localization. These studies indicate that the enzymatic activity of DRnm23 is not required for the effects observed in 32Dcl3 cells. Moreover, the inability of the S61P and DeltaRGD DRnm23 mutants to inhibit differentiation and promote apoptosis may be due to defective protein-protein interactions at the mitochondria, the predominant site of DRnm23 subcellular localization.


Subject(s)
Apoptosis , Granulocytes/cytology , Hematopoietic Stem Cells/cytology , Nucleoside-Diphosphate Kinase/metabolism , Animals , Cell Differentiation , Cell Line , Cell Survival , Gene Expression , Hematopoietic Stem Cells/enzymology , Mice , Mutagenesis , Nucleoside-Diphosphate Kinase/genetics , Subcellular Fractions , Transfection
13.
Exp Cell Res ; 256(1): 248-56, 2000 Apr 10.
Article in English | MEDLINE | ID: mdl-10739671

ABSTRACT

The preferential expression of the protooncogene c-myb in hematopoietic cells is in part regulated by a mechanism of transcriptional block in the first intron. By electrophoresis mobility shift assays using probes corresponding to different segments of the putative human c-myb intron 1 transcription pause region and nuclear extracts from myeloid leukemia HL 60 and fibroblast WI 38 cells, we detected a HL-60-specific DNA-protein complex with a 123-bp fragment containing binding sites for the interferon regulatory factors (IRFs) nuclear proteins. Formation of the DNA-protein complex was abrogated by competition with an oligomer containing the wild-type, but not the mutated, IRF binding site and the complex was specifically supershifted by the anti-IRF-1 or the anti-IRF-2 antibody. Moreover, in vitro translated IRF-1 or IRF-2 protein did interact with the 123-bp c-myb intron 1 fragment. Upon TPA-induced differentiation, c-myb expression was readily down-modulated in parental HL 60 cells, but not in cells transfected with an antisense IRF-1 plasmid. Moreover, chloramphenicol acetyltransferase activity driven by a c-myb promoter containing the entire intron 1 was suppressed upon IRF-1, but not IRF-2 expression. Together, these results are consistent with the existence of a functional relationship between IRF-1 and c-myb in which IRF-1 negatively regulates c-myb expression at the transcriptional level by a mechanism that may depend on the interaction of IRF-1 with a segment of the c-myb gene implicated in transcription pausing.


Subject(s)
DNA-Binding Proteins/metabolism , Genes, myb , Introns , Phosphoproteins/metabolism , Repressor Proteins , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Chloramphenicol O-Acetyltransferase/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation/drug effects , Genomic Library , HL-60 Cells , Humans , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-2 , Mice , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Proto-Oncogene Proteins c-myb/genetics , Recombinant Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transfection
14.
Cancer Res ; 59(10): 2451-6, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10344757

ABSTRACT

Transcriptional regulators of the Myb family play important roles in cell proliferation, differentiation, and survival. To investigate the role of Myb proteins in the regulation of apoptosis, we studied the apoptotic response of interleukin 2-dependent CTLL-2 cells stably transfected with B-Myb. B-Myb-overexpressing cells showed a diminished cytokine dependence and were resistant to apoptosis induced by doxorubicin, ceramide, and dexamethasone. Overexpression of B-Myb was associated with enhanced expression of bcl-2, which was dependent, at least in part, on increased transcription. In transient transfection assays in T-lymphoblastic cells, B-Myb was able to stimulate the promoter activity of the bcl-2 5' flanking region linked to the chloramphenicol acetyltransferase reporter gene. A segment of the bcl-2 promoter (nucleotides +34 to +58 relative to the transcription initiation site) contained a putative Myb-binding site and was shown to specifically interact with B-Myb and to confer B-Myb responsiveness to a bcl-2/chloramphenicol acetyltransferase reporter construct. These results indicate that B-Myb promotes T cells survival by enhancing the expression of bcl-2 and identify bcl-2 as a B-Myb target gene regulated in a DNA binding-dependent manner.


Subject(s)
Apoptosis/drug effects , Cell Cycle Proteins , Ceramides/pharmacology , DNA-Binding Proteins/physiology , Dexamethasone/pharmacology , Doxorubicin/pharmacology , Gene Expression Regulation , Genes, bcl-2 , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Trans-Activators/physiology , Animals , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/genetics , Genes, Reporter , Mice , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , T-Lymphocytes/metabolism , Transcription, Genetic , Transfection
15.
Proc Natl Acad Sci U S A ; 96(6): 3092-7, 1999 Mar 16.
Article in English | MEDLINE | ID: mdl-10077642

ABSTRACT

Ectopic expression of decorin in a wide variety of transformed cells results in growth arrest and the inability to generate tumors in nude mice. This process is caused by a decorin-mediated activation of the epidermal growth factor receptor, which leads to a sustained induction of endogenous p21(WAF1/CIP1) (the cyclin-dependent kinase inhibitor p21) and growth arrest. However, mice harboring a targeted disruption of the decorin gene do not develop spontaneous tumors. To test the role of decorin in tumorigenesis, we generated mice lacking both decorin and p53, an established tumor-suppressor gene. Mice lacking both genes showed a faster rate of tumor development and succumbed almost uniformly to thymic lymphomas within 6 months [mean survival age (T50) approximately 4 months]. Mice harboring one decorin allele and no p53 gene developed the same spectrum of tumors as the double knockout animals, but had a survival rate similar to the p53 null animals (T50 approximately 6 months). Ectopic expression of decorin in thymic lymphoma cells isolated from double mutant animals markedly suppressed their colony-forming ability. When these lymphoma cells were cocultured with fibroblasts derived from either wild-type or decorin null embryos, the cells grew faster in the absence of decorin. Moreover, exogenous decorin proteoglycan or its protein core significantly retarded their growth in vitro. These results indicate that the lack of decorin is permissive for lymphoma tumorigenesis in a mouse model predisposed to cancer and suggest that germ-line mutations in decorin and p53 may cooperate in the transformation of lymphocytes and ultimately lead to a more aggressive phenotype by shortening the tumor latency.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/pathology , Proteoglycans/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Division/genetics , Cell Line , Decorin , Extracellular Matrix Proteins , Germ-Line Mutation , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology
16.
EMBO J ; 17(15): 4442-55, 1998 Aug 03.
Article in English | MEDLINE | ID: mdl-9687511

ABSTRACT

The leukemogenic potential of BCR/ABL oncoproteins depends on their tyrosine kinase activity and involves the activation of several downstream effectors, some of which are essential for cell transformation. Using electrophoretic mobility shift assays and Southwestern blot analyses with a double-stranded oligonucleotide containing a zinc finger consensus sequence, we identified a 68 kDa DNA-binding protein specifically induced by BCR/ABL. The peptide sequence of the affinity-purified protein was identical to that of the RNA-binding protein FUS (also called TLS). Binding activity of FUS required a functional BCR/ABL tyrosine kinase necessary to induce PKCbetaII-dependent FUS phosphorylation. Moreover, suppression of PKCbetaII activity in BCR/ABL-expressing cells by treatment with the PKCbetaII inhibitor CGP53353, or by expression of a dominant-negative PKCbetaII, markedly impaired the ability of FUS to bind DNA. Suppression of FUS expression in myeloid precursor 32Dcl3 cells transfected with a FUS antisense construct was associated with upregulation of the granulocyte-colony stimulating factor receptor (G-CSFR) and downregulation of interleukin-3 receptor (IL-3R) beta-chain expression, and accelerated G-CSF-stimulated differentiation. Downregulation of FUS expression in BCR/ABL-expressing 32Dcl3 cells was associated with suppression of growth factor-independent colony formation, restoration of G-CSF-induced granulocytic differentiation and reduced tumorigenic potential in vivo. Together, these results suggest that FUS might function as a regulator of BCR/ABL leukemogenesis, promoting growth factor independence and preventing differentiation via modulation of cytokine receptor expression.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Leukemia, Myeloid/genetics , Proto-Oncogenes/physiology , Ribonucleoproteins/genetics , Translocation, Genetic , Amino Acid Sequence , Animals , Cell Differentiation , Cell Division , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/physiology , Gene Expression Regulation, Neoplastic , Growth Substances/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Heterogeneous-Nuclear Ribonucleoproteins , Humans , Leukemia, Myeloid/enzymology , Leukemia, Myeloid/etiology , Mice , Mice, Inbred ICR , Mice, SCID , Molecular Sequence Data , Phosphorylation , Protein Kinase C/physiology , Protein-Tyrosine Kinases/biosynthesis , RNA-Binding Protein FUS , Ribonucleoproteins/biosynthesis , Ribonucleoproteins/metabolism , Signal Transduction , Tumor Cells, Cultured
17.
Blood ; 91(6): 1934-46, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9490676

ABSTRACT

Kit is a tyrosine kinase receptor that plays an important role in human hematopoietic cell growth. The promoter elements that modulate the gene's expression have not been extensively studied. Because of c-kit's acknowledged importance in hematopoiesis, we sought to address this issue in more detail. To perform these studies we analyzed a human c-kit 5' flanking fragment approximately 1 kilobase in length. Deletion constructs showed a region approximately 139 nucleotides upstream from the translation initiation site that was critical for promoter activity. A region containing a potential silencing element was also identified. Sequence analysis indicated several potential Myb- and Ets-binding sites. The functional significance of these sites was explored by showing that both wild-type Myb and Ets-2 protein, but not a DNA binding-deficient Myb mutant protein, bound to distinct 5' flanking fragments that included these sites. Furthermore, binding of recombinant Myb and Ets-2 protein to these fragments could be competed with an excess of double stranded oligodeoxynucleotides containing canonical, but not mutated, Myb- or Ets-binding sites. We also showed that the 5' flanking region of c-kit exhibited promoter activity in nonhematopoietic cells only when the cells were transfected with c-myb or ets-2 expression vectors. Moreover, Myb and Ets-2 coexpression in such cells augmented transactivation of c-kit promoter constructs in comparison to that observed in cells transfected with either construct alone. Promoter constructs lacking various Myb and Ets sites deleted were much less effective in this same system. Finally, Myb and Ets-2 mRNA expression was detected in CD34+, Kit low as well as CD34+, Kit bright cells. In aggregate, these data further define the human c-kit promoter's functional anatomy and suggest that Myb and Ets proteins play an important, perhaps cooperative, role in regulating expression of this critical hematopoietic cell receptor.


Subject(s)
DNA-Binding Proteins , Gene Expression Regulation/physiology , Proto-Oncogene Proteins c-kit/biosynthesis , Proto-Oncogene Proteins/physiology , Repressor Proteins , Trans-Activators/physiology , Transcription Factors , 3T3 Cells , Animals , Base Sequence , Binding Sites , CHO Cells , COS Cells , Cricetinae , Cricetulus , Humans , Leukemia, Erythroblastic, Acute/pathology , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Proto-Oncogene Protein c-ets-2 , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-myb , Sequence Deletion , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured
18.
EMBO J ; 16(20): 6151-61, 1997 Oct 15.
Article in English | MEDLINE | ID: mdl-9321394

ABSTRACT

The BCR/ABL oncogenic tyrosine kinase activates phosphatidylinositol 3-kinase (PI-3k) by a mechanism that requires binding of BCR/ABL to p85, the regulatory subunit of PI-3k, and an intact BCR/ABL SH2 domain. SH2 domain BCR/ABL mutants deficient in PI-3k activation failed to stimulate Akt kinase, a recently identified PI-3k downstream effector with oncogenic potential, but did activate p21 RAS and p70 S6 kinase. The PI-3k/Akt pathway is essential for BCR/ABL leukemogenesis as indicated by experiments demonstrating that wortmannin, a PI-3k specific inhibitor at low concentrations, suppressed BCR/ABL-dependent colony formation of murine marrow cells, and that a kinase-deficient Akt mutant with dominant-negative activity inhibited BCR/ABL-dependent transformation of murine bone marrow cells in vitro and suppressed leukemia development in SCID mice. In complementation assays using mouse marrow progenitor cells, the ability of transformation-defective SH2 domain BCR/ABL mutants to induce growth factor-independent colony formation and leukemia in SCID mice was markedly enhanced by expression of constitutively active Akt. In retrovirally infected mouse marrow cells, the BCR/ABL mutant lacking the SH2 domain was unable to upregulate the expression of c-Myc and Bcl-2; in contrast, expression of a constitutively active Akt mutant induced Bcl-2 and c-Myc expression, and stimulated the transcription activation function of c-Myc. Together, these data demonstrate the requirement for the BCR/ABL SH2 domain in PI-3k activation and document the essential role of the PI-3k/Akt pathway in BCR/ABL leukemogenesis.


Subject(s)
Bone Marrow Cells , Cell Transformation, Neoplastic/genetics , Fusion Proteins, bcr-abl/genetics , Leukemia, Experimental/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Animals , Bone Marrow/pathology , Enzyme Activation , Genes, bcl-2 , Genes, myc , Leukemia, Experimental/etiology , Leukemia, Experimental/pathology , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , Proto-Oncogene Proteins c-akt , Signal Transduction , Spleen/pathology
19.
Blood ; 90(5): 1828-39, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9292515

ABSTRACT

Numerous transcription factors allow hematopoietic cells to respond to lineage- and stage-specific cytokines and/or to act as their effectors. The transcription factors PU.1 and c-Myb are essential for hematopoiesis, most likely acting at distinct stages of differentiation, but sharing a common set of target genes. To determine whether PU.1 and c-Myb are functionally interrelated, murine bone marrow (BM) cells and 32Dcl3 murine myeloid precursor cells were infected with a retrovirus carrying a PU.1 cDNA and assessed for myeloid colony formation and for granulocytic differentiation, respectively. Compared with noninfected normal BM cells or to cells infected with an empty virus, hematopoietic precursor cells expressing PU.1 formed an increased number of interleukin-3 (IL-3) and granulocyte colony-stimulating factor (G-CSF)-stimulated colonies. Moreover, granulocytic differentiation of 32Dcl3 cells constitutively expressing PU.1 was accelerated, as indicated by morphology and by expression of differentiation markers. Downregulation of c-Myb protein levels by expression of an antisense c-myb construct was also associated with a faster kinetics of 32Dcl3 granulocytic differentiation. Sequence analysis of the 5' flanking region of the c-myb gene revealed a consensus PU box at position +16 to +21 able to specifically interact in electrophoretic mobility shift assays with either bacterially synthesized PU.1 protein or whole cell extracts from differentiated 32Dcl3 cells. Transient expression of PU.1 in cotransfection assays in different cell lines resulted in inhibition of chloramphenicol acetyl transferase activity driven by different segments of the c-myb promoter. Moreover, such an effect was dependent on an intact PU box. Thus, the ability of PU.1 to potentiate terminal myeloid differentiation appears to involve downregulation of c-myb expression, an essential step during differentiation of hematopoietic precursor cells.


Subject(s)
Bone Marrow Cells , Granulocytes/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/pharmacology , Trans-Activators/genetics , Trans-Activators/pharmacology , Animals , Bone Marrow/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Gene Expression Regulation/drug effects , Granulocytes/drug effects , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Mice , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-myb
20.
Proc Natl Acad Sci U S A ; 94(8): 3966-71, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-9108088

ABSTRACT

In vitro, uniformly modified oligonucleotide N3'-->P5' phosphoramidates are apparently more potent antisense agents than phosphorothioate derivatives. To determine whether such compounds are also effective in vivo, severe combined immunodeficiency mice injected with HL-60 myeloid leukemia cells were treated systemically with equal doses of either phosphoramidate or phosphorothioate c-myc antisense or mismatched oligonucleotides. Compared with mice treated with mismatched oligodeoxynucleotides, the peripheral blood leukemic load of mice treated with the antisense sequences was markedly reduced, and such effects were associated with significantly prolonged survival of the antisense-treated mice. Moreover, with each of three different treatment schedules (100, 300, or 900 microg/day for 6 consecutive days), survival of the phosphoramidate-treated mice was significantly longer than that of the phosphorothioate-treated mice. Both phosphoramidate and phosphorothioate oligonucleotides were efficiently taken up by leukemic cells in vivo and were capable of specifically down-regulating c-Myc expression. Moreover, tissue distribution of the phosphoramidate derivatives was undistinguishable from that of the phosphorothioate derivatives. Collectively, these studies suggest that phosphoramidate oligonucleotides can serve as potent and specific antisense agents in the treatment of human leukemia and probably of other malignancies.


Subject(s)
Genes, myc , Leukemia, Experimental/therapy , Oligonucleotides, Antisense/therapeutic use , Animals , Humans , Leukemia, Experimental/genetics , Male , Mice , Mice, Inbred ICR , Mice, SCID
SELECTION OF CITATIONS
SEARCH DETAIL
...