Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 40(14): 6461-76, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22495934

ABSTRACT

The transcription factor Sox2 is essential for neural stem cells (NSC) maintenance in the hippocampus and in vitro. The transcription factor Emx2 is also critical for hippocampal development and NSC self-renewal. Searching for 'modifier' genes affecting the Sox2 deficiency phenotype in mouse, we observed that loss of one Emx2 allele substantially increased the telencephalic ß-geo (LacZ) expression of a transgene driven by the 5' or 3' Sox2 enhancer. Reciprocally, Emx2 overexpression in NSC cultures inhibited the activity of the same transgene. In vivo, loss of one Emx2 allele increased Sox2 levels in the medial telencephalic wall, including the hippocampal primordium. In hypomorphic Sox2 mutants, retaining a single 'weak' Sox2 allele, Emx2 deficiency substantially rescued hippocampal radial glia stem cells and neurogenesis, indicating that Emx2 functionally interacts with Sox2 at the stem cell level. Electrophoresis mobility shift assays and transfection indicated that Emx2 represses the activities of both Sox2 enhancers. Emx2 bound to overlapping Emx2/POU-binding sites, preventing binding of the POU transcriptional activator Brn2. Additionally, Emx2 directly interacted with Brn2 without binding to DNA. These data imply that Emx2 may perform part of its functions by negatively modulating Sox2 in specific brain areas, thus controlling important aspects of NSC function in development.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Homeodomain Proteins/metabolism , SOXB1 Transcription Factors/genetics , Telencephalon/metabolism , Transcription Factors/metabolism , Alleles , Animals , Binding Sites , Cell Line, Tumor , Cells, Cultured , Genes, Reporter , Hippocampus/metabolism , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , POU Domain Factors/antagonists & inhibitors , POU Domain Factors/metabolism , Transcription Factors/genetics
2.
J Biol Chem ; 276(45): 41700-9, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11544252

ABSTRACT

The nuclear receptor COUP-TFII was recently shown to bind to the promoter of the epsilon- and gamma-globin genes and was identified as the nuclear factor NF-E3. Transgenic experiments and genetic evidence from humans affected with hereditary persistence of fetal hemoglobin suggest that NF-E3 may be a repressor of adult epsilon and gamma expression. We show that, on the epsilon-promoter, recombinant COUP-TFII binds to two sites, the more downstream of which overlaps with an NF-Y binding CCAAT box. Binding occurs efficiently to either the 5' or the 3' COUP-TFII site but not to both sites simultaneously. However, adding recombinant NF-Y induces the formation of a stable COUP-TFII.NF-Y-promoter complex at concentrations of COUP-TFII that would not give significant binding in the absence of NF-Y. Mutations of the promoter indicate that COUP-TFII cooperates with NF-Y when bound to the 5' site, whereas binding at the 3' site is mutually exclusive. Likewise, in the gamma-promoter, COUP-TFII binds to a site overlapping the distal member of a duplicated CCAAT box, competing with NF-Y binding. Transfections in K562 cells show that both the mutation of the 5' COUP-TFII or of the NF-Y site on the epsilon-promoter decrease the activity of a luciferase reporter; the mutation of the 3' COUP-TFII site has little effect. These results, together with transgenic experiments suggesting a repressive activity of COUP-TFII on the epsilon-promoter and the observation that, on the 3' site, COUP-TFII and NF-Y binding is mutually exclusive, suggest that COUP-TFII may exert different effects on epsilon transcription depending on whether it binds to the 5' or to the 3' site. At the 5' site, COUP-TFII might cooperate with NF-Y, forming a stable complex, and stimulate transcription; at the 3' site, COUP-TFII might compete for binding with NF-Y and, directly or indirectly, decrease gene activity.


Subject(s)
CCAAT-Binding Factor/metabolism , DNA-Binding Proteins/metabolism , Globins/genetics , Promoter Regions, Genetic , Receptors, Steroid , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Binding, Competitive , COUP Transcription Factor II , COUP Transcription Factors , Humans , Mice , Mice, Transgenic , Molecular Sequence Data
3.
Development ; 127(11): 2367-82, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10804179

ABSTRACT

Sox2 is one of the earliest known transcription factors expressed in the developing neural tube. Although it is expressed throughout the early neuroepithelium, we show that its later expression must depend on the activity of more than one regionally restricted enhancer element. Thus, by using transgenic assays and by homologous recombination-mediated deletion, we identify a region upstream of Sox2 (-5.7 to -3.3 kb) which can not only drive expression of a (beta)-geo transgene to the developing dorsal telencephalon, but which is required to do so in the context of the endogenous gene. The critical enhancer can be further delimited to an 800 bp fragment of DNA surrounding a nuclease hypersensitive site within this region, as this is sufficient to confer telencephalic expression to a 3.3 kb fragment including the Sox2 promoter, which is otherwise inactive in the CNS. Expression of the 5.7 kb Sox2(beta)-geo transgene localizes to the neural plate and later to the telencephalic ventricular zone. We show, by in vitro clonogenic assays, that transgene-expressing (and thus G418-resistant) ventricular zone cells include cells displaying functional properties of stem cells, i.e. self-renewal and multipotentiality. We further show that the majority of telencephalic stem cells express the transgene, and this expression is largely maintained over two months in culture (more than 40 cell divisions) in the absence of G418 selective pressure. In contrast, stem cells grown in parallel from the spinal cord never express the transgene, and die in G418. Expression of endogenous telencephalic genes was similarly observed in long-term cultures derived from the dorsal telencephalon, but not in spinal cord-derived cultures. Thus, neural stem cells of the midgestation embryo are endowed with region-specific gene expression (at least with respect to some networks of transcription factors, such as that driving telencephalic expression of the Sox2 transgene), which can be inherited through multiple divisions outside the embryonic environment.


Subject(s)
DNA-Binding Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Stem Cells/metabolism , Telencephalon/cytology , Animals , Brain/cytology , Cell Line , Central Nervous System/cytology , DNA-Binding Proteins/genetics , Deoxyribonuclease I/metabolism , Female , Gene Expression , HMGB Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Neurons/cytology , Nuclear Proteins/genetics , Regulatory Sequences, Nucleic Acid , SOXB1 Transcription Factors , Spinal Cord/cytology , Stem Cells/cytology , Telencephalon/embryology , Telencephalon/metabolism , Transcription Factors , Transgenes , beta-Galactosidase/genetics
4.
J Biol Chem ; 275(14): 10567-76, 2000 Apr 07.
Article in English | MEDLINE | ID: mdl-10744751

ABSTRACT

The erythroid-enriched transcription factor NF-E2 is composed of two subunits, p45 and p18, the former of which is mainly expressed in the hematopoietic system. We have isolated and characterized the mouse p45 NF-E2 gene; we show here that, similar to the human gene, the mouse gene has two alternative promoters, which are differentially active during development and in different hematopoietic cells. Transcripts from the distal promoter are present in both erythroid and myeloid cells; however, transcripts from an alternative proximal 1b promoter, lying in the first intron, are abundant in erythroid cells, but barely detectable in myeloid cells. During development, both transcripts are detectable in yolk sac, fetal liver, and bone marrow. Transfection experiments show that proximal promoter 1b has a strong activity in erythroid cells, which is completely dependent on the integrity of a palindromic GATA-1 binding site. In contrast, the distal promoter 1a is not active in this assay. When the promoter 1b is placed 3' to the promoter 1a and reporter gene, in an arrangement that resembles the natural one, it acts as an enhancer to stimulate the activity of the upstream promoter la.


Subject(s)
Alternative Splicing , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Erythroid-Specific DNA-Binding Factors , Exons , Fetus , GATA1 Transcription Factor , Humans , Introns , Macromolecular Substances , MafK Transcription Factor , Mice , Molecular Sequence Data , NF-E2 Transcription Factor , NF-E2 Transcription Factor, p45 Subunit , Nuclear Proteins/genetics , Sequence Alignment , Sequence Homology, Nucleic Acid
5.
FEBS Lett ; 468(1): 59-64, 2000 Feb 18.
Article in English | MEDLINE | ID: mdl-10683441

ABSTRACT

We applied cDNA selection methods to a genomic clone (YAC 761B5) from chromosome 21 located in the so-called 'Down critical region' in 21q22.3. Starting from human fetal heart and brain mRNAs we obtained and sequenced several cDNA clones. One of these clones (Down region aspartic protease (DRAP), named also BACE2 according to the gene nomenclature) revealed a striking nucleotide and amino acid sequence identity with several motifs present in members of the aspartic protease family. In particular the amino acid sequences comprising the two catalytic sites found in all mammalian aspartic proteases are perfectly conserved. Interestingly, the predicted protein shows a typical membrane spanning region; this is at variance with most other known aspartic proteases, which are soluble molecules. We present preliminary evidence, on the basis of in vitro translation studies and cell transfection, that this gene encodes a glycosylated protein which localizes mainly intracellularly but to some extent also to the plasma membrane. Furthermore DRAP/BACE2 shares a high homology with a newly described beta-secretase enzyme (BACE-1) which is a transmembrane aspartic protease. The implications of this finding for Down syndrome are discussed.


Subject(s)
Aspartic Acid Endopeptidases/genetics , Chromosomes, Human, Pair 21/genetics , Down Syndrome/genetics , Glycoproteins/genetics , Membrane Proteins/genetics , Amino Acid Sequence , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases/biosynthesis , Base Sequence , Cell-Free System/metabolism , Chromosome Mapping , Chromosomes, Artificial, Yeast/genetics , Endopeptidases , Glycosylation , HeLa Cells , Humans , Membrane Proteins/biosynthesis , Mice , Molecular Sequence Data , Multigene Family/genetics , Sequence Analysis, DNA , Sequence Homology, Amino Acid
6.
Blood ; 93(10): 3276-85, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10233879

ABSTRACT

Targeted expression to specific tissues or cell lineages is a necessary feature of a gene therapy vector for many clinical applications, such as correction of hemoglobinopathies or thalassemias by transplantation of genetically modified hematopoietic stem cells. We developed retroviral vectors in which the constitutive viral enhancer in the U3 region of the 3' LTR is replaced by an autoregulatory enhancer of the erythroid-specific GATA-1 transcription factor gene. The replaced enhancer is propagated to the 5' LTR upon integration into the target cell genome. The modified vectors were used to transduce human hematopoietic cell lines, cord blood-derived CD34(+) stem/progenitor cells, and murine bone marrow repopulating stem cells. The expression of appropriate reporter genes (triangle upLNGFR, EGFP) was analyzed in the differentiated progeny of transduced stem cells in vitro, in liquid culture as well as in clonogenic assay, and in vivo, after bone marrow transplantation in lethally irradiated mice. The GATA-1 autoregulatory enhancer effectively restricts the expression of the LTR-driven proviral transcription unit to the erythroblastic progeny of both human progenitors and mouse-repopulating stem cells. Packaging of viral particles, integration into the target genome, and stability of the integrated provirus are not affected by the LTR modification. Enhancer replacement is therefore an effective strategy to target expression of a retroviral transgene to a specific progeny of transduced hematopoietic stem cells.


Subject(s)
DNA-Binding Proteins/genetics , Erythroblasts/cytology , Hematopoietic Stem Cells/physiology , Transcription Factors/genetics , Transcription, Genetic , Animals , Cell Line , DNA-Binding Proteins/biosynthesis , Enhancer Elements, Genetic , Erythroblasts/physiology , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Genes, Reporter , Genetic Therapy/methods , Genetic Vectors , Green Fluorescent Proteins , Hematopoietic Stem Cells/cytology , Humans , K562 Cells , Leukemia, Erythroblastic, Acute , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/genetics , Regulatory Sequences, Nucleic Acid , Retroviridae , Transcription Factors/biosynthesis , Transfection , Tumor Cells, Cultured , U937 Cells
7.
J Mol Biol ; 285(4): 1441-55, 1999 Jan 29.
Article in English | MEDLINE | ID: mdl-9917388

ABSTRACT

NF-Y (CBF) is a CCAAT-binding trimer that activates 25 % of eukaryotic promoters. It contains putative histone fold motifs (HFMs) and distorts DNA. By using electrophoretic mobility shift assays with the twin CCAAT boxes of the human gamma-globin promoter and several combinations of subunit mutants, we dissected some of the structural features of CCAAT-box binding. NF-YA and NF-YC Q-rich domains significantly influence bending angles quantitatively, but not qualitatively, since they do not modify DNA orientation. They are both required for co-operative interactions among NF-Y molecules: for this, a precise alignement of two CCAAT boxes, 32 bp, three turns of the helix, is essential. Unlike the wild-type (wt) protein, steric hindrance does not impede simultaneous binding of the mutant composed of the short homology domains to CCAAT boxes closer than 22 bp: the addition of 11 amino acid residues to NF-YB and 13 to NF-YC flanking the HFM, restores wt behaviour. These stretches are predicted to form H2B-like alphaC and H2A-like alphaN fourth helices. A further support to this hypothesis comes from off-rates analysis of mutant combinations: the half-life of NF-Y, which is dependent on the type of NF-YB used, is extremely shortened, when the putative alphaC is present, nearly as much as in the wt NF-YB. These data (i) provide further evidence that NF-YB-NF-YC belong to the H2B-H2A subclasses, (ii) uncover new features of Q-rich domains, and (iii) define rules for NF-Y synergy that are potentially important for the regulation of many eukaryotic promoters.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites/genetics , CCAAT-Enhancer-Binding Proteins , DNA/genetics , DNA-Binding Proteins/genetics , Globins/genetics , Histones/chemistry , Humans , In Vitro Techniques , Kinetics , Molecular Sequence Data , Mutation , Promoter Regions, Genetic , Protein Conformation , Protein Folding , Protein Structure, Secondary
8.
J Biol Chem ; 273(27): 16880-9, 1998 Jul 03.
Article in English | MEDLINE | ID: mdl-9642249

ABSTRACT

The CCAAT-binding activator NF-Y is formed by three evolutionary conserved subunits, two of which contain putative histone-like domains. We investigated NF-Y binding to all CCAAT boxes of globin promoters in direct binding, competition, and supershift electrophoretic mobility shift assay; we found that the alpha, zeta, and proximal gamma CCAAT boxes of human and the prosimian Galago bind avidly, and distal gamma CCAAT boxes have intermediate affinity, whereas the epsilon and beta sequences bind NF-Y very poorly. We developed an efficient in vitro transcription system from erythroid K562 cells and established that both the distal and the proximal CCAAT boxes are important for optimal gamma-globin promoter activity. Surprisingly, NF-Y binding to a mutated distal CCAAT box (a C to T at position -114) is remarkably increased upon occupancy of the high affinity proximal element, located 27 base pairs away. Shortening the distance between the two CCAAT boxes progressively prevents simultaneous CCAAT binding, indicating that NF-Y interacts in a mutually exclusive way with CCAAT boxes closer than 24 base pairs apart. A combination of circular permutation and phasing analysis proved that (i) NF-Y-induced angles of the two gamma-globin CCAAT boxes have similar amplitudes; (ii) occupancy of the two CCAAT boxes leads to compensatory distortions; (iii) the two NF-Y bends are spatially oriented with combined twisting angles of about 100 degrees. Interestingly, such distortions are reminiscent of core histone-DNA interactions. We conclude that NF-Y binding imposes a high level of functionally important coordinate organization to the gamma-globin promoter.


Subject(s)
DNA-Binding Proteins/metabolism , Globins/genetics , Transcription Factors/metabolism , Base Sequence , CCAAT-Enhancer-Binding Proteins , DNA, Recombinant , Molecular Sequence Data , Protein Binding
9.
Genomics ; 47(2): 238-45, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9479496

ABSTRACT

We report the cloning of the human homologue of the rat metalloprotease N-arginine dibasic convertase (NRD convertase). This endopeptidase is responsible for the processing, at the Arg-Lys dibasic site on the N-terminal side of the arginine residue, of propeptides and proproteins. Comparisons of the human and rat full-length cDNAs show similarity and identity of 94 and 91%, respectively. In humans NRD convertase is predominantly expressed in heart, skeletal muscle, and testis. We have also studied the expression of this gene in mouse at various developmental stages and found that the neural tissue is the almost exclusive site of expression in early development (between E 10.5 and E 16.5). To gain information about the possibility that defects in this gene are linked to inherited neuromuscular disorders, we determined the chromosomal location of the human NRD convertase gene by FISH analysis, showing that the gene resides at 1p32.2.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , Adult , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Central Nervous System/metabolism , Cloning, Molecular , Conserved Sequence , Embryo, Mammalian , Humans , In Situ Hybridization, Fluorescence , Male , Metalloendopeptidases/isolation & purification , Mice , Molecular Sequence Data , Organ Specificity/genetics , RNA, Messenger/metabolism
10.
DNA Seq ; 9(3): 163-9, 1998.
Article in English | MEDLINE | ID: mdl-10520746

ABSTRACT

We defined a sub-family of zinc finger proteins by computer analyses and comparisons of five new finger domains against protein databases. This subclass of the cysteine-cysteine/histidine-histidine motif shows additional well conserved amino acid patterns and belongs to the human kox and gli-Kruppel gene family, sharing also the same stretches of regulatory zinc finger-containing proteins of mouse and Xenopus. We particularly describe ZF6 cDNA which contains the most interesting sequence, encoding a putative multi-domain regulatory protein.


Subject(s)
Zinc Fingers/genetics , Amino Acid Sequence , Animals , Base Sequence , Conserved Sequence , DNA, Complementary/genetics , Databases, Factual , Humans , Mice , Molecular Sequence Data , Multigene Family , Phylogeny , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Xenopus laevis
11.
Br J Haematol ; 98(3): 549-59, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9332307

ABSTRACT

A human megakaryocyte cell line (B1647) has been established from bone marrow cells obtained from a patient with acute myelogenous leukaemia (FAB M2). The cells were CD34-, CD33+, HLA-DR+, CD38+, and expressed the immunophenotypic markers of the megakaryocyte lineage (CD41 and von Willebrand factor). Moreover the cells expressed the c-mpl (thrombopoietin receptor) mRNA and protein. On the other hand, the B1647 cells also possessed erythroid lineage characteristics: the vast majority of cells were glycophorin positive, and about 10% of unstimulated cells stained with an anti-globin gamma chain MoAb. In addition, S1 protection analysis demonstrated expression of beta-globin mRNA, and Epo receptor (Epo-R) protein was detected by cytofluorimetric assay. Several growth factors, when tested alone or in combination, failed to influence the B1647 cell growth. A significant increase of cell proliferation was observed only after the addition, in serum-free culture, of recombinant human megakaryocyte growth development factor (MGDF), a recombinant c-mpl ligand encompassing the receptor-binding domain and identical to thrombopoietin (TPO), at concentrations ranging from 0.01 to 1 ng/ml. Interestingly, MGDF failed to induce megakaryocytic differentiation of the B1647 cells, but significantly increased the synthesis of the globin gamma-chain. B1647 cells could be a useful model for studying the biological effect of TPO on common megakaryocyte and erythroid progenitors.


Subject(s)
Erythroid Precursor Cells/metabolism , Erythropoietin/pharmacology , Globins/biosynthesis , Megakaryocytes/metabolism , Receptors, Erythropoietin/metabolism , Thrombopoietin/pharmacology , Cell Division , Cell Line , Cytokines/pharmacology , Erythroid Precursor Cells/cytology , Humans , Megakaryocytes/cytology , Phenotype , RNA, Messenger/metabolism , Serotonin/pharmacology
12.
Oncogene ; 14(25): 3093-8, 1997 Jun 26.
Article in English | MEDLINE | ID: mdl-9223673

ABSTRACT

Pheochromocytomas are adrenal medullary tumors which arise from the transformation of neural crest-derived cells. In the course of studies of mice transgenic for an SV40 T-gene ectopically expressed in the adrenal medulla, we observed the occurrence of large, mainly bilateral tumors in a high proportion of transgenic animals. From these tumors we established immortalized cell lines which grow in vitro at 32 degrees C (the permissive temperature for the tsA58 T-protein encoded by the transgene), but not at 38 C. These cells demonstrate characteristics of both neuronal (160 kd neurofilament) and endocrine (chromogranins) cells. The expression of Mash-1 and ret supports their initial characterization as early bipotential neuro-endocrine progenitors.


Subject(s)
Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/pathology , Antigens, Polyomavirus Transforming/genetics , Mice, Transgenic/genetics , Adrenal Gland Neoplasms/mortality , Adrenal Medulla/pathology , Animals , Blotting, Northern , Cell Division/drug effects , Chromogranin A , Chromogranins/metabolism , Cytokines/pharmacology , DNA-Binding Proteins/genetics , Erythroid-Specific DNA-Binding Factors , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Neoplastic , Mice , Mutation , Nerve Growth Factors/pharmacology , Neural Crest/cytology , Neural Crest/pathology , Transcription Factors/genetics , Tumor Cells, Cultured
13.
Hum Mol Genet ; 6(2): 259-65, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9063746

ABSTRACT

The CATCH 22 acronym outlines the main clinical features of 22q11.2 deletions (cardiac defects, abnormal facies, thymic hypoplasia, cleft palate and hypocalcemia), usually found in DiGeorge (DGS) and velo-cardio-facial (VCFS) syndromes. Hemizygosity of this region may also be the cause of over 100 different clinical signs. The CATCH 22 locus maps within a 1.5 Mb region, which encompasses several genes. However, no single defect in 22q11.2 hemizygous patients can be ascribed to any gene so far isolated from the critical region of deletion. We have identified a gene in the CATCH 22 critical region, whose functional features and tissue-specific expression suggest a distinct role in embryogenesis. This gene, UFD1L, encodes the human homolog of the yeast ubiquitin fusion degradation 1 protein (UFD1p), involved in the degradation of ubiquitin fusion proteins. Cloning and characterization of the murine homolog (Ufd1l) showed it to be expressed during embryogenesis in the eyes and in the linear ear primordia. These data suggest that the proteolytic pathway that recognizes ubiquitin fusion proteins for degradation is conserved in vertebrates and that the UFD1L gene hemizygosity is the cause of some of the CATCH 22-associated developmental defects.


Subject(s)
Chromosome Aberrations/genetics , Chromosomes, Human, Pair 22 , Gene Deletion , Proteins/genetics , Ubiquitins/metabolism , Adaptor Proteins, Vesicular Transport , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Chromosome Aberrations/embryology , Chromosome Disorders , Chromosome Mapping , DNA, Complementary , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , Proteins/metabolism , Sequence Homology, Amino Acid , Syndrome
14.
Biochem Biophys Res Commun ; 231(2): 299-304, 1997 Feb 13.
Article in English | MEDLINE | ID: mdl-9070267

ABSTRACT

GATA-1 is a transcription factor expressed both in the hematopoietic system and in the Sertoli cells of the testis, and is essential for correct erythropoiesis. Hematopoietic and Sertoli cells transcribe GATA-1 from two different promoters: the proximal (erythroid) is active in hematopoietic cells; the distal (testis) is active in Sertoli cells. We investigated by RT-PCR the possibility that GATA-1 might be transcribed from the testis promoter also in hematopoietic cells. Testis promoter-derived transcripts are present at low levels in vivo at all stages of hematopoietic development. Purified multipotent progenitors, fractionated into populations expressing low or high levels of GATA-1, do not contain any "testis" transcripts. However, when grown in vitro, they rapidly express GATA-1 from the testis promoter in the presence of Erythropoietin (Epo) but not in that of other growth factors. This result reflects an Epo-dependent differentiation event, rather than a direct effect of Epo. Indeed, immortalized progenitor cell lines which respond to both Epo and SCF, continue to express testis-derived transcripts when switched from Epo to SCF.


Subject(s)
Alternative Splicing , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , Testis/metabolism , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Erythroid-Specific DNA-Binding Factors , Erythropoietin/pharmacology , GATA1 Transcription Factor , Hematopoietic Stem Cells/cytology , Male , Mice , Stem Cell Factor/pharmacology , Testis/cytology , Testis/growth & development
15.
Genes Funct ; 1(4): 245-58, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9678901

ABSTRACT

The GATA-1 gene encodes a transcription factor expressed in early multipotent haemopoietic progenitors, in more mature cells of the erythroid, megakaryocytic and other lineages, but not in late myeloid precursors; its function is essential for the normal development of the erythroid and megakaryocytic system. To define regulatory elements of the mouse GATA-1 gene, we mapped DNaseI-hypersensitive sites in nuclei of erythroid and haemopoietic progenitor cells. Five sites were detected. The two upstream sites, site 1 and site 2, represent a new and a previously defined erythroid enhancer respectively. The site 1 enhancer activity depends both on a GATA-binding site (also footprinted in vivo) and on several sites capable of binding relatively ubiquitous factors. A DNA fragment encompassing site 1, placed upstream of a GATA-1 minimal promoter, is able to drive expression of a simian virus 40 (SV40) T-antigen in the yolk sac, but not bone-marrow cells, obtained from mice transgenic for this construct, allowing in vitro establishment of immortalized yolk-sac cells. A similar construct including site 2, instead of site 1, and previously shown to be able to immortalize adult marrow cells is not significantly active in yolk-sac cells. Sites 4 and 5, located in the first large intron, have no enhancer activity; they include a long array of potential Ets-binding sites. MnlI restriction sites, overlapping some of the Ets sites, are highly accessible, in intact nuclei, to MnlI. Although these sites are present in all GATA-1-expressing cells studied, they are the only strong sites detectable in FDCP-mix multipotent progenitor cells, most of which do not yet express GATA-1. The data indicate that appropriate GATA-1 regulation may require the co-operation of different regulatory elements acting at different stages of development and cell differentiation.


Subject(s)
DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , Animals , Base Sequence , Bone Marrow Cells , Cell Differentiation , Cell Line , DNA Footprinting , Deoxyribonuclease I/metabolism , Erythroid Precursor Cells/physiology , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Hematopoietic Stem Cells/physiology , Mice , Mice, Transgenic , Molecular Sequence Data , Promoter Regions, Genetic , Restriction Mapping , Transfection , Yolk Sac/cytology
16.
Blood ; 87(7): 2885-90, 1996 Apr 01.
Article in English | MEDLINE | ID: mdl-8639908

ABSTRACT

Anthracycline antitumor drugs such as aclacinomycin (ACM) and doxorubicin (DOX) used in subtoxic concentrations induce erythroid differentiation of the erythroleukemic cell line K562. To elucidate the possible role of erythroid genes of the erythropoietin receptor (EpoR) and the transcription factor GATA-1 in this effect, the regulatory regions of the above genes and human epsilon- and gamma-globin and porphobilinogen deaminase (PBGD) genes were fused to the firefly luciferase gene. The resulting reporter constructs were tested in a transfection assay of the erythroleukemic cell line K562 stimulated to differentiate by treatment with the anthracycline drugs ACM and DOX or hemin (HEM). The results showed activation of the tested promoters after cell treatment with ACM, but not with DOX or HEM. In contrast to the mouse EpoR gene promoter, the activity of the human EpoR gene promoter (-659/-60) in the reporter construct was not modified by addition of the first intron sequence. In ACM-treated K562 cells, EpoR gene promoter activity completely correlated with EpoR and GATA-1 mRNA levels and the degree of erythroid maturation. In addition, ACM strongly activated the erythroid gene promoters that contain GATA binding sites. Nevertheless, less activation was also observed for the GATA-1 gene promoter (-312/-31) lacking any known GATA binding sites. Insertion of the GATA-1 gene enhancer with two canonic GATA binding sites, stimulated the ACM activation effect for EpoR and GATA-1 promoter-containing constructs. Mutation of the enhancer GATA binding sites abolished this effect. All the regulatory regions tested (except gamma-globin promoter) were completely inactive in nonerythroid COS7 cells. These data indicate that (1) two structurally different anthracycline analogues, DOX and ACM, differ in their differentiation mechanisms, and (2) ACM switches on the erythroid program of K562 cells, at least in part because of interaction with a factor(s) that recognizes the GATA binding sites in the promoter region of erythroid genes leading to their activation.


Subject(s)
Aclarubicin/analogs & derivatives , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Leukemia, Erythroblastic, Acute/pathology , Promoter Regions, Genetic/genetics , Aclarubicin/pharmacology , Base Sequence , Cell Differentiation/drug effects , Erythropoiesis/drug effects , Humans , Leukemia, Erythroblastic, Acute/genetics , Molecular Sequence Data , Tumor Cells, Cultured
17.
EMBO J ; 15(1): 143-9, 1996 Jan 02.
Article in English | MEDLINE | ID: mdl-8598197

ABSTRACT

Hereditary persistence of fetal haemoglobin (HPFH) is a clinically important condition in which a change in the developmental specificity of the gamma-globin genes results in varying levels of expression of fetal haemoglobin in the adult. The condition is benign and can significantly alleviate the symptoms of thalassaemia or sickle cell anaemia when co-inherited with these disorders. We have examined structure-function relationships in the -117 HPFH gamma promoter by analysing the effect of mutating specific promoter elements on the functioning of the wild-type and HPFH promoters. We find that CCAAT box mutants dramatically affect expression from the HPFH promoter in adult blood but have little effect on embryonic/fetal expression from the wild-type promoter. Our results suggest that there are substantial differences in the structure of the wild-type gamma promoter expressed early in development and the adult HPFH promoter. Together with previous results, this suggests that gamma silencing is a complex multifactorial phenomenon rather than being the result of a simple repressor binding to the promoter. We present a model for gamma-globin gene silencing that has significant implications for attempts to reactivate the gamma promoters in human adults by pharmacological means.


Subject(s)
CCAAT-Enhancer-Binding Proteins , Fetal Hemoglobin/genetics , Globins/genetics , Promoter Regions, Genetic , Animals , Base Sequence , DNA-Binding Proteins/metabolism , Erythroid-Specific DNA-Binding Factors , Gene Expression Regulation , Humans , Liver/embryology , Mice , Mice, Transgenic , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Binding , Transcription Factors/metabolism , Yolk Sac/metabolism
18.
Acta Haematol ; 95(3-4): 229-35, 1996.
Article in English | MEDLINE | ID: mdl-8677748

ABSTRACT

To understand the molecular mechanisms of erythroid differentiation, we analyzed by semiquantitative RT-PCR the expression of the transcription factor GATA1, the erythropoietin receptor (EpoR), and erythroid (beta-globin) differentiation markers in purified hematopoietic stem cells (HSCs) after in-vitro-induced differentiation. Whether GATA1 transcription was from the proximal (with respect to the AUG, also known as erythroid) or the distal (also known as testis) promoter was analyzed as well. Low-density marrow cells which bind to wheat germ agglutinin, but not to the antibody 15.1.1, and which either do or do not retain the dye rhodamine-123 (Rho-bright and Rho-dull, respectively), were purified. Rho-dull, but not Rho-bright cells permanently reconstitute lymphomyelopoiesis in W/Wv and severe-combined-immunodeficiency mice and, therefore, contain HSCs. Both Rho-dull and Rho-bright cells give rise to progenitor and differentiated cells (peak values at days 15 and 5, respectively) in liquid culture. Multilineage, erythroid-restricted or myeloid-restricted differentiation is observed when the cultures are stimulated with stem cell factor (SCF) + interleukin (IL)-3, SCF + IL-3 + Epo, or SCF + IL-3 + granulocyte-colony-stimulating factor, respectively. Rho-dull cells have barely detectable reconstitution potential at day 5 of culture. None of the genes examined were expressed in purified Rho-bright or Rho-dull cells. The only exception was GATA1 which was expressed at maximal levels in Rho-bright cells at the onset of culture. Rho-dull cells did not express GATA1 before day 3 of culture (maximal expression at days 10-15). Activation of GATA1 and EpoR was observed in all growth of mRNA for the two genes expressed by the cells. In contrast, beta-globin mRNA was detected only in the presence of Epo. The transcription of GATA1 was exclusively from the proximal promoter in the absence of Epo but both proximal and distal transcripts were observed in its presence. Maximum transcription from the distal promoter (approximately equal to 0.2% of total GATA1 mRNA) coincided with maximal globin mRNA levels (day 5 or day 15 for Rho-bright and Rho-dull cells, respectively). These results indicate that GATA1 is activated at the transition point between HSCs and pluripotent progenitor cells and erythroid-specific GATA1 regulation involves activation of the distal GATA1 promoter.


Subject(s)
DNA-Binding Proteins/genetics , Erythroid Precursor Cells/metabolism , Erythropoiesis , Growth Substances/pharmacology , Hematopoietic Stem Cells/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Actins/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Erythroid Precursor Cells/cytology , Erythroid-Specific DNA-Binding Factors , Erythropoietin , GATA1 Transcription Factor , Gene Expression , Globins/genetics , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/cytology , Interleukin-3/pharmacology , Mice , Mice, Inbred Strains , Polymerase Chain Reaction , Receptors, Erythropoietin/genetics , Stem Cell Factor/pharmacology
19.
J Biol Chem ; 270(37): 21934-41, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7545172

ABSTRACT

Naturally occurring nondeletional mutations affecting the distal CCAAT box of the human gamma-globin gene promoter result in hereditary persistence of fetal hemoglobin in adult life. Although the distal CCAAT box is the target of several factors, including CP1/NFY, CDP, GATA-1 and NFE3, only NFE3 binding activity is consistently sensitive to well characterized mutations in this region such as G-117-->A, C-114-->T, and delta 13 hereditary persistence of fetal hemoglobin. We extensively characterized the binding specificities of NFE3 and demonstrated that NFE3 has unique properties with respect to other CCAAT box-binding proteins. Affinity-purified NFE3 from erythroid K562 cells binds the distal but not the proximal human gamma-globin CCAAT box, the single CCAAT box of the human epsilon-globin promoter, and the proximal CCAAT box of the evolutionarily related Galago crassicaudatus gamma-globin gene. Within the epsilon-globin CCAAT box, NFE3 represents the major and almost exclusive binding activity. Disruption of such a binding site essentially inactivates the epsilon-globin promoter, suggesting that NFE3 plays an important role in the embryonic expression of this gene.


Subject(s)
CCAAT-Enhancer-Binding Proteins , DNA-Binding Proteins/metabolism , Fungal Proteins/metabolism , Globins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Saccharomyces cerevisiae Proteins , Transcription Factors/metabolism , Adult , Base Sequence , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Binding Sites , Fetal Hemoglobin/genetics , Helix-Loop-Helix Motifs , Humans , Methylation , Molecular Sequence Data , Mutagenesis, Site-Directed
20.
Blood ; 86(1): 323-8, 1995 Jul 01.
Article in English | MEDLINE | ID: mdl-7795240

ABSTRACT

Juvenile chronic myelocytic leukemia (JCML) is a rare disorder of early childhood. Characteristic of JCML are the progressive appearance of high levels of fetal hemoglobin (HbF), reflecting a true reversion to a fetal type of erythropoiesis, and the presence of colony-forming cells able to grow in vitro spontaneously in the absence of growth factors. To better understand the relationship between the erythroid abnormalities and the leukemic process, we analyzed the expression pattern of specific genes related to erythroid differentiation--GATA-1, EPOR, alpha-globin, beta-globin, and gamma-globin genes--in JCML peripheral blood (PB) cells and in vitro-derived colonies. Northern blot analysis of PB cells from five JCML patients indicated levels of GATA-1 transcripts much higher than those usually found in other types of leukemic cells, and S1 nuclease protection assay detected significantly increased expression of gamma-globin mRNA. Reverse transcription-polymerase chain reaction (RT-PCR) analysis of single granulocyte-macrophage colony-forming unit (CFU-GM) colonies, obtained in vitro in the absence of added growth factors from four JCML patients, detected GATA-1, EPOR, and globin (alpha and gamma) transcripts in most of the colonies tested, in contrast with control CFU-GM from normal bone marrow, which were positive only for GATA-1. Single JCML colonies were tested for the presence of two different transcripts; whereas alpha- and gamma-globin genes appeared mostly coexpressed, beta-globin mRNA was detected only in a minority of the gamma-globin-positive colonies, indicating that the leukemic pattern of hemoglobin synthesis is mainly fetal. In addition, the leukemic cells occurring during blast crisis of one of our patients displayed the typical features of a stem cell leukemia (CD34+, CD19-, CD2-, myeloperoxidase-). In this sorted CD34+ population, we detected the presence of a marker chromosome, der(12)t(3;12), previously identified in bone marrow cells at diagnosis and an expression pattern superimposable to that of the JCML colonies, consistently displaying a high gamma-globin:beta-globin mRNA ratio. The expression of erythroid markers within populations of leukemic cells, both in vivo and in vitro, supports the hypothesis that abnormal JCML erythroid cells may originate from the same mutated progenitor that sustains the growth of the leukemic cells.


Subject(s)
DNA-Binding Proteins/genetics , Erythropoietin/genetics , Gene Expression Regulation, Leukemic , Globins/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Neoplasm Proteins/genetics , Transcription Factors/genetics , Cell Differentiation , Child , DNA-Binding Proteins/biosynthesis , Erythroid-Specific DNA-Binding Factors , Erythropoietin/biosynthesis , GATA1 Transcription Factor , Globins/biosynthesis , Humans , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/metabolism , Neoplasm Proteins/biosynthesis , Transcription Factors/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...