Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Blood ; 127(26): 3369-81, 2016 06 30.
Article in English | MEDLINE | ID: mdl-27076172

ABSTRACT

Disrupting mutations of the RUNX1 gene are found in 10% of patients with myelodysplasia (MDS) and 30% of patients with acute myeloid leukemia (AML). Previous studies have revealed an increase in hematopoietic stem cells (HSCs) and multipotent progenitor (MPP) cells in conditional Runx1-knockout (KO) mice, but the molecular mechanism is unresolved. We investigated the myeloid progenitor (MP) compartment in KO mice, arguing that disruptions at the HSC/MPP level may be amplified in downstream cells. We demonstrate that the MP compartment is increased by more than fivefold in Runx1 KO mice, with a prominent skewing toward megakaryocyte (Meg) progenitors. Runx1-deficient granulocyte-macrophage progenitors are characterized by increased cloning capacity, impaired development into mature cells, and HSC and Meg transcription signatures. An HSC/MPP subpopulation expressing Meg markers was also increased in Runx1-deficient mice. Rescue experiments coupled with transcriptome analysis and Runx1 DNA-binding assays demonstrated that granulocytic/monocytic (G/M) commitment is marked by Runx1 suppression of genes encoding adherence and motility proteins (Tek, Jam3, Plxnc1, Pcdh7, and Selp) that support HSC-Meg interactions with the BM niche. In vitro assays confirmed that enforced Tek expression in HSCs/MPPs increases Meg output. Interestingly, besides this key repressor function of Runx1 to control lineage decisions and cell numbers in progenitors, our study also revealed a critical activating function in erythroblast differentiation, in addition to its known importance in Meg and G/M maturation. Thus both repressor and activator functions of Runx1 at multiple hematopoietic stages and lineages likely contribute to the tumor suppressor activity in MDS and AML.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Megakaryocytes/metabolism , Transcription, Genetic , Tumor Suppressor Proteins/metabolism , Animals , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Megakaryocytes/pathology , Mice , Mice, Knockout , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Tumor Suppressor Proteins/genetics
2.
Blood ; 114(12): 2476-88, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19584403

ABSTRACT

Acute myelogenous leukemia is driven by leukemic stem cells (LSCs) generated by mutations that confer (or maintain) self-renewal potential coupled to an aberrant differentiation program. Using retroviral mutagenesis, we identified genes that generate LSCs in collaboration with genetic disruption of the gene encoding interferon response factor 8 (Irf8), which induces a myeloproliferation in vivo. Among the targeted genes, we identified Mef2c, encoding a MCM1-agamous-deficiens-serum response factor transcription factor, and confirmed that overexpression induced a myelomonocytic leukemia in cooperation with Irf8 deficiency. Strikingly, several of the genes identified in our screen have been reported to be up-regulated in the mixed-lineage leukemia (MLL) subtype. High MEF2C expression levels were confirmed in acute myelogenous leukemia patient samples with MLL gene disruptions, prompting an investigation of the causal interplay. Using a conditional mouse strain, we demonstrated that Mef2c deficiency does not impair the establishment or maintenance of LSCs generated in vitro by MLL/ENL fusion proteins; however, its loss led to compromised homing and invasiveness of the tumor cells. Mef2c-dependent targets included several genes encoding matrix metalloproteinases and chemokine ligands and receptors, providing a mechanistic link to increased homing and motility. Thus, MEF2C up-regulation may be responsible for the aggressive nature of this leukemia subtype.


Subject(s)
DNA-Binding Proteins/metabolism , Leukemia, Myelomonocytic, Acute/metabolism , Leukemia, Myelomonocytic, Acute/pathology , Myeloid-Lymphoid Leukemia Protein/metabolism , Myogenic Regulatory Factors/metabolism , Neoplastic Stem Cells/pathology , Transcription Factors/metabolism , Animals , Bone Marrow Transplantation , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Colony-Forming Units Assay , DNA-Binding Proteins/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Humans , Interferon Regulatory Factors/physiology , Leukemia Virus, Murine/physiology , Leukemia, Myelomonocytic, Acute/genetics , MEF2 Transcription Factors , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Transcription Factors/genetics , Transduction, Genetic
3.
Blood ; 111(9): 4532-41, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18326819

ABSTRACT

Mef2c is a MADS (MCM1-agamous-deficient serum response factor) transcription factor best known for its role in muscle and cardiovascular development. A causal role of up-regulated MEF2C expression in myelomonocytic acute myeloid leukemia (AML) has recently been demonstrated. Due to the pronounced monocytic component observed in Mef2c-induced AML, this study was designed to assess the importance of Mef2c in normal myeloid differentiation. Analysis of bone marrow (BM) cells manipulated to constitutively express Mef2c demonstrated increased monopoiesis at the expense of granulopoiesis, whereas BM isolated from Mef2c(Delta/-) mice showed reduced levels of monocytic differentiation in response to cytokines. Mechanistic studies showed that loss of Mef2c expression correlated with reduced levels of transcripts encoding c-Jun, but not PU.1, C/EBPalpha, or JunB transcription factors. Inhibiting Jun expression by short-interfering RNA impaired Mef2c-mediated inhibition of granulocyte development. Moreover, retroviral expression of c-Jun in BM cells promoted monocytic differentiation. The ability of Mef2c to modulate cell-fate decisions between monocyte and granulocyte differentiation, coupled with its functional sensitivity to extracellular stimuli, demonstrate an important role in immunity--and, consistent with findings of other myeloid transcription factors, a target of oncogenic lesions in AML.


Subject(s)
Myeloid Cells/cytology , Myogenic Regulatory Factors/physiology , Proto-Oncogene Proteins c-jun/physiology , Animals , Bone Marrow Cells , Cell Differentiation , Granulocytes/cytology , Hematopoiesis , MEF2 Transcription Factors , Mice , Mice, Mutant Strains , Monocytes/cytology , Transcription Factors/physiology
4.
Exp Hematol ; 34(9): 1192-201, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16939812

ABSTRACT

INTRODUCTION: The NUP98-TOP1 fusion gene is one of 18 distinct translocations identified in acute myeloid leukemia involving the N-terminal portion of the nucleoporin NUP98. We previously reported that expression of NUP98-TOP in murine bone marrow induces a lethal, transplantable leukemia. However, the long latency suggests the in vivo acquisition of additional mutations and/or time required for clonal outgrowth of rare transformed cells arising from the collaboration of NUP98-TOP1 and a cooperating event. The aim of this study was to test whether retroviral insertional mutagenesis contributes to disease onset and whether integration site analysis can identify collaborating genes. METHODS: The genomic sites of retroviral integration in NUP98-TOP1-induced leukemic mice were analyzed. This screen identified a proviral integration that disrupts expression of the Interferon consensus sequence binding protein (ICSBP) tumor suppressor gene. Intriguingly, an ICSBP deficiency induces a chronic myeloid leukemia-like disease in mice and its reduced expression has been observed in several human leukemias. To ascertain whether an ISCBP deficiency collaborates with NUP98-TOP1 in leukemogenesis, we expressed NUP98-TOP1 in ICSBP(-/-) bone marrow. RESULTS: The in vivo myeloproliferation induced by NUP98-TOP1 was markedly exaggerated with the ICSBP(-/-) deficiency. Moreover, NUP98-TOP1/ICSBP(-/-) mice had a reduced survival compared with NUP98-TOP1/ICSBP(+/+) mice. CONCLUSION: These results reveal the novel finding of collaboration between the ICSBP tumor suppressor gene and NUP98-TOP1 in leukemogenesis. Moreover they further illustrate the power of retroviral integration site analysis for identifying novel cooperating tumor suppressor genes.


Subject(s)
Genes, Tumor Suppressor , Interferon Regulatory Factors/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Oncogene Proteins, Fusion/genetics , Retroviridae , Virus Integration , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Transformation, Viral/genetics , DNA Mutational Analysis/methods , Gene Expression Regulation, Leukemic/genetics , Humans , Interferon Regulatory Factors/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Knockout , Mutagenesis, Insertional/genetics , Mutagenesis, Insertional/methods , Oncogene Proteins, Fusion/metabolism
5.
Oncogene ; 24(51): 7579-91, 2005 Nov 17.
Article in English | MEDLINE | ID: mdl-16044150

ABSTRACT

The t(12;21) translocation, generating the TEL/AML1 fusion protein, is the most common genetic lesion in childhood cancer. Using a bone marrow transplantation model, we demonstrate that TEL/AML1 expression impinges on normal hematopoietic differentiation, leading to the in vivo accumulation and persistence of an early progenitor compartment with a Sca1(+)/Kit(hi)/CD11b(+) phenotype and an increased self-renewal capacity, as documented by replating assays in vitro. Differentiation of these cells is not blocked, but the frequency of mature blood cells arising from TEL/AML1-transduced progenitors is low. Impaired differentiation is prominently observed in the pro-B-cell compartment, resulting in an proportional increase in early progenitors in vivo, consistent with the t(12;21) ALL phenotype. Despite the accumulation of both multipotent and B-cell progenitors in vivo, no leukemia induction was observed during an observation period of over 1 year. These results are consistent with findings in twins with concordant ALL, showing that TEL/AML1 generates a preleukemic clone in utero that persists for several years in a clinically covert fashion. Furthermore, our studies showed that the pointed domain of TEL/AML1, which recruits transcriptional repressors and directs oligomerization with either TEL/AML1 or wild-type TEL, was essential for the observed differentiation impairment and could not be replaced with another oligomerization domain.


Subject(s)
Cell Transformation, Neoplastic/genetics , Core Binding Factor Alpha 2 Subunit/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , Preleukemia/genetics , Animals , B-Lymphocytes , Bone Marrow Transplantation , Cell Differentiation , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 21 , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/physiology , Hematopoietic Stem Cells , Humans , Mice , Mice, Inbred C57BL , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Phenotype , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Preleukemia/physiopathology , Translocation, Genetic
6.
Leuk Lymphoma ; 46(2): 265-72, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15621811

ABSTRACT

AML1-ETO is generated by the t(8;21) translocation found in approximately 12% of acute myelogenous leukemia. Studies to delineate the mechanism by which AML1-ETO induces leukemia have primarily relied on transformed human cell lines or murine model systems. The goal of this study was to determine the effect of AML1-ETO expression on primary human hematopoietic cells in vitro and in a xenograft model. We used a FMEV retroviral vector for the transfer of AML1/ETO into human CD34 + cells. The repopulation, self-renewal, and differentiation potential of infected cells were assessed in serum-free liquid culture, colony assays, and in transplanted NOD-SCID mice. High transcription levels were confirmed by real-time PCR. AML1-ETO expressing cells were expandable for up to 12 weeks and retained an immature morphology. The capacity for prolonged survival, however, did not abrogate maturation, as AML1-ETO cells gave rise to normal colonies in a CFU-assay. AML1/ETO-expressing cells also contributed to myeloid (CD15, CD33), B-lymphoid (CD20), NK-cell (CD56) and erythroid (GPA) lineages in xenografted NOD/SCID mice. Although able to engraft all major lineages, AML1/ETO transplanted cells were primarily found in less differentiated fractions as measured by cell surface markers CD34 and CD38. In spite of a good engraftment and prolonged observation period none of the NOD/SCID-mice developed an acute myelogenous leukemia. Our findings demonstrate that AML1/ETO promotes the maintenance of early human hematopoietic progenitors, but does not abrogate their physiologic differentiation. Furthermore, the leukemogenic potential of AML1/ETO expressed in human progenitors is low, despite transcription levels equivalent to those found in AMLs.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/cytology , Oncogene Proteins, Fusion/physiology , Transcription Factors/physiology , Animals , Cell Lineage , Cell Proliferation , Cells, Cultured , Core Binding Factor Alpha 2 Subunit , Hematopoietic Stem Cell Transplantation , Humans , Leukemia/etiology , Mice , Mice, Inbred NOD , Mice, SCID , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein , Transcription Factors/genetics , Transfection , Transplantation, Heterologous
7.
Eur J Biochem ; 271(20): 4123-31, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15479241

ABSTRACT

Two chloroplast phosphoglycerate kinase isoforms from the photosynthetic flagellate Euglena gracilis were purified to homogeneity, partially sequenced, and subsequently cDNAs encoding phosphoglycerate kinase isoenzymes from both the chloroplast and cytosol of E. gracilis were cloned and sequenced. Chloroplast phosphoglycerate kinase, a monomeric enzyme, was encoded as a polyprotein precursor of at least four mature subunits that were separated by conserved tetrapeptides. In a Neighbor-Net analysis of sequence similarity with homologues from numerous prokaryotes and eukaryotes, cytosolic phosphoglycerate kinase of E. gracilis showed the highest similarity to cytosolic and glycosomal homologues from the Kinetoplastida. The chloroplast isoenzyme of E. gracilis did not show a close relationship to sequences from other photosynthetic organisms but was most closely related to cytosolic homologues from animals and fungi.


Subject(s)
Chloroplasts/enzymology , Euglena gracilis/enzymology , Phosphoglycerate Kinase/genetics , Symbiosis/genetics , Amino Acid Sequence , Animals , Blotting, Northern , Cloning, Molecular , Cytosol/enzymology , DNA, Complementary/genetics , Electrophoresis, Polyacrylamide Gel , Eukaryota/genetics , Isoenzymes , Molecular Sequence Data , Phosphoglycerate Kinase/isolation & purification , Phylogeny , Protein Biosynthesis/genetics , Protein Precursors/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Sequence Analysis, Protein/methods , Sequence Homology, Amino Acid
8.
Blood ; 103(7): 2744-52, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14656889

ABSTRACT

The CCAAT/enhancer binding protein alpha (C/EBPalpha) is an essential transcription factor for granulocytic differentiation. C/EBPalpha mutations are found in approximately 8% of acute myeloid leukemia (AML) patients. Most of these mutations occur in the N-terminal coding region, resulting in a frame shift and the enhanced translation of a dominant-negative 30-kDa protein, which may be responsible for the differentiation block observed in AML. To test this hypothesis, we introduced a cDNA encoding an N-terminal mutated C/EBPalpha (mut10) into primary hematopoietic progenitors using a retroviral vector. Expression of mut10 in human CD34+ cord blood cells dramatically inhibited differentiation of both myeloid and erythroid lineages. Immunohistochemical analysis demonstrated coexpression of both myeloid and erythroid markers in the immature transformed cells. Surprisingly, mut10 did not block myelocytic differentiation in murine progenitors but did alter their differentiation kinetics and clonogenicity. Experiments were performed to confirm that the differential effect of mut10 on murine and human progenitors was not due to species-specific differences in C/EBPalpha protein sequences, expression levels, or inefficient targeting of relevant cells. Taken together, our results underline the intrinsic differences between hematopoietic controls in mouse and human and support the hypothesis that mutations in CEBPA are critical events in the disruption of myeloid differentiation in AMLs.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/pharmacology , Erythroid Precursor Cells/drug effects , Leukemia, Myeloid, Acute/genetics , Mutation , Myeloid Progenitor Cells/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Cloning, Molecular , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/pathology , Female , Fetal Blood/cytology , Genes, Dominant , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Infant, Newborn , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/pathology , Recombinant Proteins/pharmacology , Species Specificity
9.
Exp Hematol ; 31(12): 1206-14, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14662326

ABSTRACT

OBJECTIVE: Current protocols of retroviral gene transfer into murine hematopoietic stem cells (HSC) result in variable gene transfer efficiency and involve various procedures that are not clinically applicable. We developed and evaluated a reliable transduction protocol that is more related to clinical methods. MATERIALS AND METHODS: HSC were enriched from steady-state bone marrow by magnetic cell sorting (lineage depletion) and cultured in defined serum-free medium containing an improved growth factor cocktail (Flt3-ligand, stem cell factor, interleukin-3, interleukin-11). Cell-free ecotropic retroviral vector particles, generated by transient transfection of human 293T-based packaging cells, were preloaded at defined titers on CH296-coated tissue culture plates, thus largely avoiding serum contamination. These conditions were evaluated in 17 experiments involving 29 transduction cultures and 185 recipient mice. RESULTS: After two rounds of infection, the gene marking rates in cultured mononuclear cells and stem/progenitor cells (Lin(-)c-Kit(+)) were 15 to 85% (53.7%+/-21.7%, n=23) and 30 to 95% (69.8%+/-20.4%, n=17), respectively. Even after one round of infection, gene transfer was efficient (31.2%+/-15.1%, n=12). Using identical conditions, gene transfer rates were highly reproducible. Average transgene expression in reconstituted animals correlated well with pretransplant data. Using a moderate multiplicity of infection, the majority of transduced cells carried less than three transgene copies. In addition, coinfection was possible to establish two different vectors in single cells. CONCLUSION: The protocol described here achieves efficient retroviral transduction of murine bone marrow repopulating cells with a defined gene dosage, largely avoiding procedures that decrease stem cell output and repopulating capacity. This protocol may help to improve the predictive value of preclinical efficiency/toxicity studies for gene therapeutic interventions and basic research.


Subject(s)
Genetic Vectors , Hematopoietic Stem Cells/metabolism , Transduction, Genetic/methods , Animals , Bone Marrow Cells , Gene Dosage , Gene Transfer Techniques/standards , Immunomagnetic Separation , Mice , Mice, Inbred Strains , Retroviridae/genetics , Transduction, Genetic/standards , Transgenes/genetics
10.
J Exp Med ; 196(9): 1227-40, 2002 Nov 04.
Article in English | MEDLINE | ID: mdl-12417632

ABSTRACT

The translocation (8;21), generating the AML1-ETO fusion protein, is one of the most frequent chromosomal abnormalities associated with acute myelogenous leukemia (AML). To elucidate its role in oncogenesis, bone marrow (BM) cells were infected with a retroviral vector carrying AML1-ETO and transplanted into mice. In contrast to previous transgenic mouse models, we show that AML1-ETO directly stimulates granulopoiesis, suppresses erythropoiesis, and impairs the maturation of myeloid, B, and T lymphoid cells in vivo. To determine the significance of earlier findings that expression of the tumor suppressor ICSBP is often downregulated in AML myeloblasts, AML1-ETO was introduced into BM cells derived from mice lacking the interferon regulatory factor ICSBP. Our findings demonstrate that AML1-ETO synergizes with an ICSBP deficiency to induce myeloblastic transformation in the BM, reminiscent of AML.


Subject(s)
Hematopoiesis/physiology , Oncogene Proteins, Fusion/physiology , Repressor Proteins/physiology , Transcription Factors/physiology , Animals , B-Lymphocytes/cytology , Bone Marrow Transplantation , Cell Differentiation , Cell Lineage , Core Binding Factor Alpha 2 Subunit , Erythropoiesis , Gene Expression , Genetic Vectors , Interferon Regulatory Factors , Lymphopoiesis/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein , Repressor Proteins/genetics , Retroviridae , Transcription Factors/genetics , Transduction, Genetic
11.
Nucleic Acids Res ; 30(12): e59, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12060697

ABSTRACT

Site-specific recombination in genetically modified cells can be achieved by the activity of Cre recombinase from bacteriophage P1. Commonly an expression vector encoding Cre is introduced into cells; however, this can lead to undesired side-effects. Therefore, we tested whether cell-permeable Cre fusion proteins can be directly used for lox-specific recombination in a cell line tailored to shift from red to green fluorescence after loxP-specific recombination. Comparison of purified recombinant Cre proteins with and without a heterologous 'protein transduction domain' surprisingly showed that the unmodified Cre recombinase already possesses an intrinsic ability to cross the membrane border. Addition of purified recombinant Cre enyzme to primary bone marrow cells isolated from transgenic C/EBPalpha(fl/fl) mice also led to excision of the 'floxed' C/EBPalpha gene, thus demonstrating its potential for in vivo applications. We conclude that Cre enyzme itself or its intrinsic membrane-permeating moiety are attractive tools for direct manipulation of mammalian cells.


Subject(s)
Gene Targeting/methods , Integrases/metabolism , Recombination, Genetic , Viral Proteins/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Line , Cell Membrane/enzymology , Cells, Cultured , Genes, Reporter , Integrases/genetics , Mice , Mice, Transgenic , Protein Transport , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Viral Proteins/genetics
12.
EMBO J ; 21(3): 211-20, 2002 Feb 01.
Article in English | MEDLINE | ID: mdl-11823414

ABSTRACT

Mice lacking transcription factor interferon consensus sequence binding protein (ICSBP) develop a syndrome similar to human chronic myeloid leukemia and are immunodeficient. In order to define the molecular mechanisms responsible for the cellular defects of ICSBP(-/-) mice, we used bone marrow-derived macrophages (BMM) to identify genes deregulated in the absence of ICSBP. Here, we report that disabled-2 (Dab2), a signal phosphoprotein, is transcriptionally up-regulated and accumulates in the cytoskeleton/membrane fraction of ICSBP(-/-) BMM. Moreover, our results revealed Dab2 as a novel IFN-gamma-response gene. Both ICSBP and the Ets-transcription factor PU.1 bind to the Dab2 promoter, whereby ICSBP represses PU.1-induced Dab2 promoter transactivation in vitro. Notably, repression of Dab2 expression by ICSBP is also found in myeloid progenitors. Overexpression of Dab2 leads to accelerated cell adhesion and spreading, accompanied by enhanced actin fiber formation. Furthermore, cell adhesion induces transient Dab2 phosphorylation and its translocation to the cytoskeletal/membrane fraction. Our results identify a novel role of Dab2 as an inducer of cell adhesion and spreading, and strongly suggest that the up-regulation of Dab2 contributes to the hematopoietic defect seen in ICSBP(-/-) mice.


Subject(s)
Adaptor Proteins, Vesicular Transport , Macrophages/physiology , Proteins/genetics , Repressor Proteins/genetics , Transcriptional Activation , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Cell Adhesion/genetics , Genes, Tumor Suppressor , Hematopoiesis/genetics , Humans , Interferon Regulatory Factors , K562 Cells , Macrophages/cytology , Mice , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Tumor Suppressor Proteins , Up-Regulation
13.
J Virol ; 76(1): 303-12, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11739695

ABSTRACT

The objective of this work was to identify, in the context of chromosomally integrated DNA, the contribution of defined transcription factor binding motifs to the function of a complex retrovirus enhancer in hematopoietic cells in vivo. Repopulating murine hematopoietic cells were transduced with equal gene dosages of replication-incompetent retrovirus vectors encoding enhanced green fluorescent protein. Enhancer sequences were derived from mouse spleen focus-forming virus. Destruction of GC-rich sites representing overlapping targets for SP1 or EGR1 uniformly attenuated gene expression (approximately 25 to 70% of wild-type levels) in all hematopoietic lineages, as shown by multicolor flow cytometry of peripheral blood and bone marrow cells at various time points posttransplantation. In contrast, a point mutation within a dual ETS/GATA motif that abolished transactivation by ETS factors but not by GATA-1 slightly increased activity in erythroid cells and significantly attenuated enhancer function in T lymphocytes. This study shows that controlled gene transfer in transplantable hematopoietic cells allows a functional analysis of distinct cis elements within a complex retrovirus enhancer, as required for the characterization and engineering of various cellular and viral regulatory sequences in basic research and gene therapy.


Subject(s)
Enhancer Elements, Genetic , Hematopoietic Stem Cells/virology , Retroviridae/genetics , Terminal Repeat Sequences , Animals , Base Sequence , DNA-Binding Proteins/physiology , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation , Sp1 Transcription Factor/physiology , Spleen Focus-Forming Viruses/genetics , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...