Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 110(14): 5594-9, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23509296

ABSTRACT

MicroRNA-9 (miR-9) is emerging as a critical regulator of organ development and neurogenesis. It is also deregulated in several types of solid tumors; however, its role in hematopoiesis and leukemogenesis is not yet known. Here we show that miR-9 is detected in hematopoietic stem cells and hematopoietic progenitor cells, and that its expression increases during hematopoietic differentiation. Ectopic expression of miR-9 strongly accelerates terminal myelopoiesis and promotes apoptosis in vitro and in vivo. Conversely, in hematopoietic progenitor cells, the inhibition of miR-9 with a miRNA sponge blocks myelopoiesis. Ecotropic viral integration site 1 (EVI1), required for normal embryogenesis, is considered an oncogene because its inappropriate up-regulation induces malignant transformation in solid and hematopoietic cancers. Here we show that EVI1 binds to the promoter of miR-9-3, leading to DNA hypermethylation of the promoter and repression of miR-9. Moreover, miR-9 expression reverses a myeloid differentiation block that is induced by EVI1. Our findings indicate that EVI1, when inappropriately expressed, delays or blocks myeloid differentiation at least in part by DNA hypermethylation and down-regulation of miR-9. It was reported that Forkhead box class O genes (FoxOs) inhibit myeloid differentiation and prevent differentiation of leukemia-initiating cells. Here we identify both FoxO1 and FoxO3 as direct targets of miR-9 in hematopoietic cells and find that up-regulation of FoxO3 inhibits miR-9-induced myelopoiesis. These results reveal a unique role of miR-9 in myelopoiesis and in the pathogenesis of EVI1-induced myeloid neoplasms and provide insights into the epigenetic regulation of miR9 in tumorigenesis.


Subject(s)
DNA Methylation , DNA-Binding Proteins/metabolism , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/metabolism , MicroRNAs/metabolism , Myelopoiesis/physiology , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , Colony-Forming Units Assay , DNA Primers/genetics , Flow Cytometry , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , HEK293 Cells , Humans , MDS1 and EVI1 Complex Locus Protein , Mice , Myelopoiesis/genetics , NIH 3T3 Cells , Proto-Oncogenes , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
2.
J Biol Chem ; 287(27): 22683-90, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22589536

ABSTRACT

The canonical Wnt/ß-catenin signaling is activated during development, tumorigenesis, and in adult homeostasis, yet its role in maintenance of hematopoietic stem/progenitor cells is not firmly established. Here, we demonstrate that conditional expression of an active form of ß-catenin in vivo induces a marked increase in the frequency of apoptosis in hematopoietic stem/progenitor cells (HSCs/HPCs). Activation of Wnt/ß-catenin signaling in HPCs in vitro elevates the activity of caspases 3 and 9 and leads to a loss of mitochondrial membrane potential (ΔΨ(m)), indicating that it induces the intrinsic mitochondrial apoptotic pathway. In vivo, expression of activated ß-catenin in HPCs is associated with down-regulation of Bcl2 and expression of Casp3. Bone marrow transplantation assays reveal that enhanced cell survival by a Bcl2 transgene re-establishes the reconstitution capacity of HSCs/HPCs that express activated ß-catenin. In addition, a Bcl2 transgene prevents exhaustion of these HSCs/HPCs in vivo. Our data suggest that activation of the Wnt/ß-catenin pathway contributes to the defective function of HPCs in part by deregulating their survival.


Subject(s)
Apoptosis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mitochondria/physiology , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Survival/physiology , Cells, Cultured , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , beta Catenin/genetics
3.
PLoS One ; 6(6): e20793, 2011.
Article in English | MEDLINE | ID: mdl-21695170

ABSTRACT

EVI1 has pleiotropic functions during murine embryogenesis and its targeted disruption leads to prenatal death by severely affecting the development of virtually all embryonic organs. However, its functions in adult tissues are still unclear. When inappropriately expressed, EVI1 becomes one of the most aggressive oncogenes associated with human hematopoietic and solid cancers. The mechanisms by which EVI1 transforms normal cells are unknown, but we showed recently that EVI1 indirectly upregulates self-renewal and cell-cycling genes by inappropriate methylation of CpG dinucleotides in the regulatory regions of microRNA-124-3 (miR-124-3), leading to the repression of this small gene that controls normal differentiation and cell cycling of somatic cells. We used the regulatory regions of miR-124-3 as a read-out system to investigate how EVI1 induces de novo methylation of DNA. Here we show that EVI1 physically interacts with DNA methyltransferases 3a and 3b (Dnmt3a/b), which are the only de novo DNA methyltransferases identified to date in mouse and man, and that it forms an enzymatically active protein complex that induces de novo DNA methylation in vitro. This protein complex targets and binds to a precise region of miR-124-3 that is necessary for repression of a reporter gene by EVI1. Based on our findings, we propose that in cooperation with Dnmt3a/b EVI1 regulates the methylation of DNA as a sequence-specific mediator of de novo DNA methylation and that inappropriate EVI1 expression contributes to carcinogenesis through improper DNA methylation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Catalytic Domain , DNA Methyltransferase 3A , DNA-Binding Proteins/chemistry , Genome/genetics , Humans , Immunoprecipitation , MDS1 and EVI1 Complex Locus Protein , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , NIH 3T3 Cells , Oncogene Proteins/chemistry , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogenes , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/chemistry , Zinc Fingers , DNA Methyltransferase 3B
4.
Blood ; 117(24): 6498-508, 2011 Jun 16.
Article in English | MEDLINE | ID: mdl-21518930

ABSTRACT

The transcription factor (TF) RUNX1 cooperates with lineage-specifying TFs (eg, PU.1/SPI1) to activate myeloid differentiation genes, such as macrophage and granulocyte macrophage colony-stimulating factor receptors (MCSFR and GMCSFR). Disruption of cooperative gene activation could contribute to aberrant repression of differentiation genes and leukemogenesis initiated by mutations and translocations of RUNX1. To investigate the mechanisms underlying cooperative gene activation, the effects of Runx1 deficiency were examined in an in vitro model of Pu.1-driven macrophage differentiation and in primary cells. Runx1 deficiency decreased Pu.1-mediated activation of Mcsfr and Gmcsfr, accompanied by decreased histone acetylation at the Mcsfr and Gmcsfr promoters, and increased endogenous corepressor (Eto2, Sin3A, and Hdac2) coimmunoprecipitation with Pu.1. In cotransfection experiments, corepressors were excluded from a multiprotein complex containing full-length RUNX1 and PU.1. However, corepressors interacted with PU.1 if wild-type RUNX1 was replaced with truncated variants associated with leukemia. Histone deacetylase (HDAC) enzyme activity is a major component of corepressor function. HDAC inhibition using suberoylanilide hydroxamic acid or MS-275 significantly increased MCSFR and GMCSFR expression in leukemia cell lines that express PU.1 and mutated or translocated RUNX1. RUNX1 deficiency is associated with persistent corepressor interaction with PU.1. Thus, inhibiting HDAC can partly compensate for the functional consequences of RUNX1 deficiency.


Subject(s)
Co-Repressor Proteins/metabolism , Core Binding Factor Alpha 2 Subunit/physiology , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Cell Line, Tumor , Co-Repressor Proteins/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Humans , Mice , Mice, Transgenic , Microarray Analysis , NIH 3T3 Cells , Protein Binding/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Transfection
5.
Blood ; 116(26): 6023-6, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-20861460

ABSTRACT

Recently, Dawson et al identified a previously unrecognized nuclear role of JAK2 in the phosphorylation of histone H3 in hematopoietic cell lines. We searched nuclear JAK2 in total bone marrow (BM) cells and in 4 sorted BM cell populations (CD34(+), CD15(+), CD41(+), and CD71(+)) of 10 myeloproliferative neoplasia (MPN) patients with JAK2V617F mutation and 5 patients with wild-type JAK2 MPN. Confocal immunofluorescent images and Western blot analyses of nuclear and cytoplasmic fractions found nuclear JAK2 in CD34(+) cells of 10 of 10 JAK2-mutated patients but not in patients with wild-type JAK2. JAK2 was predominantly in the cytoplasmic fraction of differentiated granulocytic, megakaryocytic, or erythroid cells obtained from all patients. JAK2V617F up-regulates LMO2 in K562 and in JAK2V617F-positive CD34(+) cells. The selective JAK2 inhibitor AG490 normalizes the LMO2 levels in V617F-positive K562 and restores the cyto-plasmic localization of JAK2.


Subject(s)
Antigens, CD34/metabolism , Cell Nucleus/metabolism , Erythroid Cells/metabolism , Granulocytes/metabolism , Janus Kinase 2/genetics , Megakaryocytes/metabolism , Myeloproliferative Disorders/metabolism , Philadelphia Chromosome , Apoptosis , Blotting, Western , Bone Marrow/metabolism , Cells, Cultured , Cytoplasm/metabolism , Granulocytes/cytology , Humans , Janus Kinase 2/metabolism , K562 Cells , Megakaryocytes/cytology , Microscopy, Fluorescence , Mutation/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Phosphorylation , Prognosis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
6.
Proc Natl Acad Sci U S A ; 107(21): 9783-8, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20448201

ABSTRACT

By expressing EVI1 in murine bone marrow (BM), we previously described a myelodysplastic syndrome (MDS) model characterized by pancytopenia, dysmegakaryopoiesis, dyserythropoiesis, and BM failure. The mice invariably died 11-14 months after BM transplantation (BMT). Here, we show that a double point mutant EVI1-(1+6Mut), unable to bind Gata1, abrogates the onset of MDS in the mouse and re-establishes normal megakaryopoiesis, erythropoiesis, BM function, and peripheral blood profiles. These normal features were maintained in the reconstituted mice until the study was ended at 21 months after BMT. We also report that EVI1 deregulates several genes that control cell division and cell self-renewal. In striking contrast, these genes are normalized in the presence of the EVI1 mutant. Moreover, EVI1, but not the EVI1 mutant, seemingly deregulates these cellular processes by altering miRNA expression. In particular, the silencing of miRNA-124 by DNA methylation is associated with EVI1 expression, but not that of the EVI1 mutant, and appears to play a key role in the up-regulation of cell division in murine BM cells and in the hematopoietic cell line 32Dcl3. The results presented here demonstrate that EVI1 induces MDS in the mouse through two major pathways, both of which require the interaction of EVI1 with other factors: one, results from EVI1-Gata1 interaction, which deregulates erythropoiesis and leads to fatal anemia, whereas the other occurs by interaction of EVI1 with unidentified factors causing perturbation of the cell cycle and self-renewal, as a consequence of silencing miRNA-124 by EVI1 and, ultimately, ensues in BM failure.


Subject(s)
DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/cytology , MicroRNAs/genetics , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , RNA Interference , Transcription Factors/metabolism , Animals , Bone Marrow Transplantation , Cell Differentiation , Cell Proliferation , Cells, Cultured , CpG Islands , DNA Replication , DNA-Binding Proteins/genetics , Gene Expression Regulation , MDS1 and EVI1 Complex Locus Protein , Methylation , Mice , Molecular Sequence Data , Mutation , Myelodysplastic Syndromes/pathology , Promoter Regions, Genetic , Proto-Oncogenes/genetics , Transcription Factors/genetics
7.
Cancer Res ; 69(4): 1633-42, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19208846

ABSTRACT

EVI1 is an oncogene inappropriately expressed in the bone marrow (BM) of approximately 10% of myelodysplastic syndrome (MDS) patients. This disease is characterized by severe anemia and multilineage myeloid dysplasia that are thought to be a major cause of mortality in MDS patients. We earlier reported on a mouse model that constitutive expression of EVI1 in the BM led to fatal anemia and myeloid dysplasia, as observed in MDS patients, and we subsequently showed that EVI1 interaction with GATA1 blocks proper erythropoiesis. Whereas this interaction could provide the basis for the erythroid defects in EVI1-positive MDS, it does not explain the alteration of myeloid differentiation. Here, we have examined the expression of several genes activated during terminal myelopoiesis in BM cells and identified a group of them that are altered by EVI1. A common feature of these genes is their regulation by the transcription factor PU.1. We report here that EVI1 interacts with PU.1 and represses the PU.1-dependent activation of a myeloid promoter. EVI1 does not seem to inhibit PU.1 binding to DNA, but rather to block its association with the coactivator c-Jun. After mapping the PU.1-EVI1 interaction sites, we show that an EVI1 point mutant, unable to bind PU.1, restores the activation of PU.1-regulated genes and allows a normal differentiation of BM progenitors in vitro.


Subject(s)
DNA-Binding Proteins/physiology , Myelodysplastic Syndromes/genetics , Myelopoiesis/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogenes/physiology , Trans-Activators/genetics , Transcription Factors/physiology , 3T3 Cells , Anemia/genetics , Animals , Cell Differentiation , Cell Line , Chromatin/genetics , Colony-Forming Units Assay , DNA-Binding Proteins/genetics , Erythropoiesis/genetics , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , MDS1 and EVI1 Complex Locus Protein , Mice , Myelodysplastic Syndromes/pathology , Polymerase Chain Reaction , Proto-Oncogenes/genetics , Transcription Factors/genetics
8.
Cancer Res ; 69(1): 262-71, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19118011

ABSTRACT

Janus-activated kinase 2 (JAK2) mutations are common in myeloproliferative disorders; however, although they are detected in virtually all polycythemia vera patients, they are found in approximately 50% of essential thrombocythemia (ET) patients, suggesting that converging pathways/abnormalities underlie the onset of ET. Recently, the chromosomal translocation 3;21, leading to the fusion gene AML1/MDS1/EVI1 (AME), was observed in an ET patient. After we forced the expression of AME in the bone marrow (BM) of C57BL/6J mice, all the reconstituted mice died of a disease with symptoms similar to ET with a latency of 8 to 16 months. Peripheral blood smears consistently showed an elevated number of dysplastic platelets with anisocytosis, degranulation, and giant size. Although the AME-positive mice did not harbor Jak2 mutations, the BM of most of them had significantly higher levels of activated Stat3 than the controls. With combined biochemical and biological assays we found that AME binds to the Stat3 promoter leading to its up-regulation. Signal transducers and activators of transcription 3 (STAT3) analysis of a small group of ET patients shows that in about half of the patients, there is STAT3 hyperactivation independently of JAK2 mutations, suggesting that the hyperactivation of STAT3 by JAK2 mutations or promoter activation may be a critical step in development of ET.


Subject(s)
Janus Kinase 2/genetics , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/metabolism , Thrombocythemia, Essential/genetics , Aged , Animals , Blood Platelets/pathology , Bone Marrow Cells/pathology , Core Binding Factor Alpha 2 Subunit/biosynthesis , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Disease Models, Animal , Enzyme Activation , Humans , Janus Kinase 2/metabolism , K562 Cells , Megakaryocytes/pathology , Mice , Mice, Inbred C57BL , Middle Aged , Oncogene Proteins, Fusion/biosynthesis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Repressor Proteins/metabolism , STAT3 Transcription Factor/genetics , Thrombocythemia, Essential/blood , Thrombocythemia, Essential/enzymology , Thrombocythemia, Essential/metabolism , Up-Regulation
9.
J Cell Biochem ; 105(2): 344-52, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18655152

ABSTRACT

EVI1 is an oncoprotein inappropriately expressed in acute myeloid leukemia and myelodysplastic syndrome cells. In vitro studies indicate that diverse biological properties can be attributed to this protein. Its role in leukemogenesis is still unclear but it is thought that overall EVI1 can act mostly as a transcription repressor through its interaction with a subset of histone deacetylases. Studies with histone deacetylase inhibitors have however indicated that EVI1-mediated repression can be only partially rescued by deacetylase inhibitor drugs, suggesting that additional chromosomal modifications might occur to induce gene repression by EVI1. To investigate whether histone methylation contributes to the repressive potential of EVI1, we examined a potential association between EVI1, the histone methyltransferase (HMT) SUV39H1, and methyltransferase activity in vitro. We find that EVI1 directly interacts with SUV39H1 and that the proteins form an active complex with methyltransferase activity in vitro. Our data indicate that SUV39H1 enhances the transcription repressive potential of EVI1 in vivo. We suggest that EVI1 affects promoters' activity in two different pathways, by association with histone deacetylases and by recruiting chromatin-modifying enzymes to impose a heterochromatin-like structure establishing a lasting transcription repression.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation , Methyltransferases/metabolism , Proto-Oncogenes/physiology , Repressor Proteins/metabolism , Transcription Factors/physiology , Transcription, Genetic , Animals , Binding Sites , Cell Line , DNA-Binding Proteins/genetics , Histone Methyltransferases , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Leukemia/etiology , MDS1 and EVI1 Complex Locus Protein , Methylation , Mice , Protein Binding , Protein Methyltransferases , Proto-Oncogenes/genetics , Transcription Factors/genetics , Transcriptional Activation
11.
Blood Cells Mol Dis ; 40(2): 141-7, 2008.
Article in English | MEDLINE | ID: mdl-17913523

ABSTRACT

EVI1 was first identified as a preferential integration site of ecotropic retroviruses in the MDS1/EVI1 genomic locus leading to myeloid tumors in susceptible mice. Later studies showed that retroviral integration in the MDS1/EVI1 locus results in the emergence of a non-malignant dominant hematopoietic stem cell clone in non-susceptible mice strains, in non-human primates, and in patients, suggesting that a gene encoded by the locus could affect the self-renewal potential of a cell. The locus encodes two genes. One of them, EVI1, has long been associated with myeloid leukemia. To understand whether EVI1 has a role in self-renewal control, we forcibly expressed EVI1 in the bone marrow progenitors of two mice strains that differ in their proliferation and self-renewal potential. By comparing the response of the hematopoietic cells to EVI1, we conclude that EVI1 has a role in prolonging the self-renewal potential of the cells and that this ability of EVI1 is limited and modulated by inherent characteristics of the cells.


Subject(s)
DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/physiology , Hematopoietic System/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Gene Expression , Genetic Vectors , Hematopoietic Stem Cells/cytology , Hematopoietic System/cytology , MDS1 and EVI1 Complex Locus Protein , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Proto-Oncogenes , Retroviridae/genetics
12.
Cancer Res ; 67(12): 5658-66, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575132

ABSTRACT

Recurring chromosomal translocations observed in human leukemia often result in the expression of fusion proteins that are DNA-binding transcription factors. These altered proteins acquire new dimerization properties that result in the assembly of inappropriate multimeric transcription complexes that deregulate hematopoietic programs and induce leukemogenesis. Recently, we reported that the fusion protein AML1/MDS1/EVI1 (AME), a product of a t(3;21)(q26;q22) associated with chronic myelogenous leukemia and acute myelogenous leukemia, displays a complex pattern of self-interaction. Here, we show that the 8th zinc finger motif of MDS1/EVI1 is an oligomerization domain involved not only in interaction of AME with itself but also in interactions with the parental proteins, RUNX1 and MDS1/EVI1, from which AME is generated. Because the 8th zinc finger motif is also present in the oncoprotein EVI1, we have evaluated the effects of the interaction between RUNX1 and EVI1 in vitro and in vivo. We found that in vitro, this interaction alters the ability of RUNX1 to bind to DNA and to regulate a reporter gene, whereas in vivo, the expression of the isolated 8th zinc finger motif of EVI1 is sufficient to block the granulocyte colony-stimulating factor-induced differentiation of 32Dcl3 cells, leading to cell death. As EVI1 is not detected in normal bone marrow cells, these data suggest that its inappropriate expression could contribute to hematopoietic transformation in part by a new mechanism that involves EVI1 association with key hematopoietic regulators, leading to their functional impairment.


Subject(s)
Cell Transformation, Neoplastic , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Leukemia/genetics , Proto-Oncogenes/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Animals , Blotting, Western , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cloning, Molecular , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , Humans , MDS1 and EVI1 Complex Locus Protein , Mice , NIH 3T3 Cells , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Transcription Factors/metabolism , Transfection , Zinc Fingers/physiology
13.
J Biol Chem ; 282(18): 13542-51, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17355962

ABSTRACT

RUNX1 (AML1, CBFalpha2, PEBP2alphaB) is a transcription factor essential for the establishment of the hematopoietic stem cell. It is generally thought that RUNX1 exists as a monomer that regulates hematopoietic differentiation by interacting with tissue-specific factors and its DNA consensus through its N terminus. RUNX1 is frequently altered in human leukemia by gene fusions or point mutations. In general, these alterations do not affect the N terminus of the protein, and it is unclear how they consistently lead to hematopoietic transformation and leukemia. Here we report that RUNX1 homodimerizes through a mechanism involving C terminus-C terminus interaction. This RUNX1-RUNX1 interaction regulates the activity of the protein in reporter gene assays and modulates its ability to induce hematopoietic differentiation of hematopoietic cell lines. The promoters of genes regulated by RUNX1 often contain multiple RUNX1 binding sites. This arrangement suggests that RUNX1 could homodimerize to bring and hold together distant chromatin sites and factors and that if the dimerization region is removed by gene fusions or is altered by point mutations, as observed in leukemia, the ability of RUNX1 to regulate differentiation could be impaired.


Subject(s)
Cell Differentiation/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation , Animals , Binding Sites/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Dimerization , Gene Expression Regulation/genetics , HeLa Cells , Hematopoietic Stem Cells/metabolism , Humans , Leukemia/genetics , Leukemia/metabolism , Mice , NIH 3T3 Cells , Oncogene Proteins, Fusion , Point Mutation , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Response Elements
14.
Mol Cell Biol ; 26(20): 7658-66, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16954386

ABSTRACT

EVI1 is an aggressive nuclear oncoprotein deregulated by recurring chromosomal abnormalities in myelodysplastic syndrome (MDS). The expression of the corresponding gene is a very poor prognostic marker for MDS patients and is associated with severe defects of the erythroid lineage. We have recently shown that the constitutive expression of EVI1 in murine bone marrow results in a fatal disease with features characteristic of MDS, including anemia, dyserythropoiesis, and dysmegakaryopoiesis. These lineages are regulated by the DNA-binding transcription factor GATA1. EVI1 has two zinc finger domains containing seven motifs at the N terminus and three motifs at the C terminus. Supported by results of assays utilizing synthetic DNA promoters, it was earlier proposed that erythroid-lineage repression by EVI1 is based on the ability of this protein to compete with GATA1 for DNA-binding sites, resulting in repression of gene activation by GATA1. Here, however, we show that EVI1 is unable to bind to classic GATA1 sites. To understand the mechanism utilized by EVI1 to repress erythropoiesis, we used a combination of biochemical assays, mutation analyses, and in vitro bone marrow differentiation. The results indicate that EVI1 interacts directly with the GATA1 protein rather than the DNA sequence. We further show that this protein-protein interaction blocks efficient recognition or binding to DNA by GATA1. Point mutations that disrupt the geometry of two zinc fingers of EVI1 abolish the protein-protein interaction, leading to normal erythroid differentiation of normal murine bone marrow in vitro.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , DNA-Binding Proteins/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , GATA1 Transcription Factor/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , GATA1 Transcription Factor/genetics , Humans , Immunoprecipitation , Mice , Point Mutation/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Zinc Fingers
15.
Crit Rev Eukaryot Gene Expr ; 16(2): 183-92, 2006.
Article in English | MEDLINE | ID: mdl-16749897

ABSTRACT

The hematopoietic stem cell (HSC) has the potential to differentiate into mature cells with distinct phenotypes and functions. As suggested in recent reports, this plasticity can expand to include nonhematopoietic lineages, and, indeed, the HSC may repopulate liver and muscle tissues, as well. Considering the flexibility in HSC differentiation, these processes are regulated by a relatively small number of factors, some of which are expressed in all lineages, whereas others are activated only in a specific cell type. Combined evidence from many studies suggests that alternative subsets of these factors work in a combinatorial manner to regulate specific promoters for the induction of a specific lineage. RUNX1 and PU.1 have a fundamental role in HSC differentiation in that multifactor complexes are assembled around these proteins leading to tissue-specific and synergistic gene activation. Here we describe the relationship of RUNX1 with PU.1 as a facet of the combinatorial relationships that determine hematopoietic lineage commitment.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Hematopoiesis/physiology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/physiology , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Leukemia/genetics , Models, Biological , Mutation , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Transcriptional Activation , Translocation, Genetic
16.
Gene ; 368: 1-11, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16314052

ABSTRACT

The ecotropic viral integration site 1 (EVI1) gene was identified almost 20 years ago as the integration site of an ecotropic retrovirus leading to murine myeloid leukemia. Since its identification, EVI1 has slowly been recognized as one of the most aggressive oncogenes associated with human leukemia. Despite the effort of many investigators, still very little is known about this gene. The mechanism by which EVI1 operates in the transformation of hematopoietic cells is not known, but it is clear that EVI1 upregulates cell proliferation, impairs cell differentiation, and induces cell transformation. In this review, we summarize the biochemical properties of EVI1 and the effects of EVI1 in biological models.


Subject(s)
DNA-Binding Proteins , Hematologic Diseases/metabolism , Proto-Oncogenes , Transcription Factors , Amino Acid Sequence , Animals , DNA-Binding Proteins/history , DNA-Binding Proteins/physiology , Hematologic Diseases/pathology , History, 20th Century , History, 21st Century , Humans , MDS1 and EVI1 Complex Locus Protein , Mice , Molecular Sequence Data , Oncogene Proteins, Fusion , Phylogeny , Proto-Oncogenes/physiology , Sequence Homology, Amino Acid , Transcription Factors/history , Transcription Factors/physiology
17.
J Cell Physiol ; 207(3): 582-93, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16250015

ABSTRACT

Converging studies from many investigators indicate that RUNX1 has a critical role in the correct maintenance of essential cellular functions during embryonic development and after birth. The discovery that this gene is also frequently mutated in human leukemia has increased the interest in the role that RUNX1 plays in both normal and transforming pathways. Here, we provide an overview of the many roles of RUNX1 in hematopoietic self-renewal and differentiation and summarize the information that is currently available on the many mechanisms of RUNX1 deregulation in human leukemia.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation , Genome/genetics , Humans , Leukemia/genetics , Leukemia/metabolism , RNA Splicing/genetics
19.
Cancer Res ; 65(17): 7603-11, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140925

ABSTRACT

AML1/MDS1/EVI1 (AME) is a chimeric transcription factor produced by the (3;21)(q26;q22) translocation. This chromosomal translocation is associated with de novo and therapy-related acute myeloid leukemia and with the blast crisis of chronic myelogenous leukemia. AME is obtained by in-frame fusion of the AML1 and MDS1/EVI1 (ME) genes. The mechanisms by which AME induces a neoplastic transformation in bone marrow cells are unknown. AME interacts with the corepressors CtBP and HDAC1, and it was shown that AME is a repressor in contrast to the parent transcription factors AML1 and ME, which are transcription activators. Studies with murine bone marrow progenitors indicated that the introduction of a point mutation that destroys the CtBP-binding consensus impairs but does not abolish the disruption of cell differentiation and replication associated with AME expression, suggesting that additional events are required. Several chimeric proteins, such as AML1/ETO, BCR/ABL, and PML/RARa, are characterized by the presence of a self-interaction domain critical for transformation. We report that AME is also able to oligomerize and displays a complex pattern of self-interaction that involves at least three oligomerization regions, one of which is the distal zinc finger domain. Although the deletion of this short domain does not preclude the self-interaction of AME, it significantly reduces the differentiation defects caused in vitro by AME in primary murine bone marrow progenitors. The addition of a point mutation that inhibits CtBP binding completely abrogates the effects of AME on differentiation, suggesting that AME induces hematopoietic differentiation defects through at least two separate but cooperating pathways.


Subject(s)
Hematopoietic Stem Cells/cytology , Oncogene Proteins, Fusion/metabolism , Repressor Proteins/metabolism , Zinc Fingers/physiology , Alcohol Oxidoreductases , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Cell Transformation, Neoplastic/metabolism , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Humans , Mice , NIH 3T3 Cells , Oncogene Proteins, Fusion/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic , Protein Structure, Tertiary , Repressor Proteins/genetics , Transfection , Up-Regulation , Zinc Fingers/genetics
20.
Arch Biochem Biophys ; 441(2): 168-73, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16122695

ABSTRACT

Transcription repression in eukaryotes is mediated by a wide variety of transcription factors that usually recruit corepressors and form corepressor complexes at the specific promoter sites. One of these corepressors is the C-terminal-binding protein (CtBP) which was first identified as a protein that binds to the C-terminal region of the adenovirus E1A protein. CtBP has a strong role in both development and oncogenesis. Till date, the mechanism of transcription repression by CtBP is unknown. Here, we report that CtBP physically interacts in vivo with HAT enzymes from different families. The vast majority of the HAT enzymes have a potential consensus site for CtBP binding within the bromodomain but we show that additional site(s) exists for CBP. The interaction between CtBP and CBP is functionally important and leads to impairment of histone H3 acetylation by CBP at specific lysine residues (Lys9, Lys14, and Lys18) in a dose-dependent and NADH-dependent manner. Based on these results, we propose that CtBP1 mediates repression by blocking histone acetylation by HAT complexes.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Kidney/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , Alcohol Oxidoreductases , Amino Acid Sequence , Binding Sites , CREB-Binding Protein , Cell Line , Humans , Molecular Sequence Data , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...