Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
BMC Cancer ; 19(1): 440, 2019 May 14.
Article in English | MEDLINE | ID: mdl-31088547

ABSTRACT

BACKGROUND: Palliative pemetrexed-based chemotherapy remains a standard of care treatment for the majority of patients with advanced non-squamous non-small-cell lung cancer (NSCLC). Currently, no predictive markers for pemetrexed treatment are available. METHODS: Resected tumour samples from pemetrexed-naïve NSCLC patients were collected. Gene expression profiling with respect to predicted sensitivity to pemetrexed classified predicted responders (60%) and non-responders (40%) based on differentially expressed genes encoding for pemetrexed target enzymes. Genes showing a strong correlation with these target genes were selected for measurement of corresponding protein expressions by immunohistochemical (IHC) staining. A semi-quantitative IHC scoring method was applied to construct a prediction model for response to pemetrexed. A retrospective cohort of patients with advanced NSCLC treated with first-line pemetrexed-based chemotherapy was used for external validation. RESULTS: From ninety-one patients resected tumour samples were collected. The majority of patients had early or locally advanced NSCLC (96.3%). Gene expression profiling revealed five markers, which mRNA levels strongly correlated to pemetrexed target genes mRNA levels: TPX2, CPA3, EZH2, MCM2 and TOP2A. Of 63 (69%) patients IHC staining scores of these markers were obtained, which significantly differed between predicted non-responders and responders (P < 0.05). The optimized prediction model included EZH2 (OR = 0.56, 95% CI 0.35-0.90) and TPX2 (OR = 0.55, 95% CI 0.30-1.01). The model had a sensitivity of 86.8%, specificity of 63.6% and showed a good ability to distinct between responders and non-responders (C-index 0.86). In the external study population (N = 23) the majority of patients had metastatic NSCLC (95.7%). Partial response (PR) was established in 26.1%. The sensitivity decreased drastically to 33.3%, with a specificity of 82.4% and a C-index of 0.73. CONCLUSIONS: Using external validation this prediction model with IHC staining of target enzyme correlated markers showed a good discrimination, but lacked sensitivity. The role of IHC markers as response predictors for pemetrexed in clinical practice remains questionable.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Gene Expression Profiling/methods , Lung Neoplasms/drug therapy , Pemetrexed/administration & dosage , Aged , Algorithms , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Middle Aged , Models, Theoretical , Pemetrexed/therapeutic use , ROC Curve , Retrospective Studies , Tissue Array Analysis/methods , Treatment Outcome
2.
Am J Hematol ; 82(5): 342-8, 2007 May.
Article in English | MEDLINE | ID: mdl-17133428

ABSTRACT

In this report we examine the (AC)n(AT)xTy motif residing -530 bp 5' upstream of the beta-globin gene in Chinese thalassaemic patients. This motif is a putative binding site for a repressor protein, termed beta protein 1 (BP1) (Berg et al., Nucleic Acids Res 1989;17:8833-8852). Variations in the (AC)n(AT)xTy repeats affect the binding affinity of BP1, thereby altering the expression of the beta-globin gene. Eight different configurations of this repeat motif are identified in our population of Chinese beta-thalassaemia patients. A (AC)3(AT)7T5 motif was identified among these thalassaemia patients and its influence in beta-globin gene expression was studied using stable transfection assay in murine erythroleukemia (MEL) cells. Our data demonstrated that the (AC)3(AT)7T5 motif has a moderately strong repressor effect on the expression of the cis-linked beta-globin gene. The high affinity of BP1 for this motif may result in the suppression of the transcription of the beta-globin gene (Berg et al., Am J Hematol 1991;36:42-47). We postulate that silencer elements in the beta-globin promoter play an important role in modifying the clinical presentation of the disease.


Subject(s)
5' Untranslated Regions/genetics , Globins/genetics , Promoter Regions, Genetic/genetics , Silencer Elements, Transcriptional/genetics , beta-Thalassemia/genetics , Animals , Binding Sites , Cell Differentiation , Cell Line, Tumor , Fetal Hemoglobin/analysis , Gene Expression Regulation , Globins/biosynthesis , Homeodomain Proteins/metabolism , Hong Kong , Humans , Leukemia, Erythroblastic, Acute/pathology , Mice , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Transfection , beta-Thalassemia/ethnology
3.
Genes Cells ; 6(8): 689-97, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11532028

ABSTRACT

BACKGROUND: Sp4 is a zinc finger transcription factor which is closely related to Sp1 and Sp3. All three proteins recognize the same DNA elements and can act as transcriptional activators through glutamine-rich activation domains. Unlike Sp1 and Sp3, which are ubiquitous proteins, Sp4 is highly abundant in the central nervous system, but also detectable in many other tissues. RESULTS: We have disrupted the mouse Sp4 gene by a targeted deletion of the exons encoding the N-terminal activation domains. Sp4 knockout mice show a complete absence of Sp4 expression. They develop until birth without obvious abnormalities. After birth, two-thirds die within 4 weeks. Surviving mice are growth retarded. Male Sp4null mice do not breed. The cause for the breeding defect remains obscure since they show complete spermatogenesis. In addition, pheromone receptor genes in the vomeronasal organ appear unaffected. Female Sp4null mice have a smaller thymus, spleen and uterus. In addition, they exhibit a pronounced delay in sexual maturation. CONCLUSIONS: The phenotype of the Sp4null mice differs significantly from those described for Sp1-/- and Sp3-/- mice. Thus, the structural similarities, the common recognition motif and the overlapping expression pattern of these three transcription factors do not reflect similar physiological functions.


Subject(s)
Transcription Factors/physiology , Alleles , Animals , Cell Line , Chromosome Mapping , Cloning, Molecular , DNA/metabolism , Embryo, Mammalian/metabolism , Embryonic and Fetal Development , Female , Gene Targeting , Growth/physiology , Growth Disorders/genetics , Homozygote , Male , Mice , Mice, Knockout , Mutation , Reproduction , Sexual Maturation/physiology , Sp4 Transcription Factor , Tissue Distribution , Transcription Factors/deficiency , Transcription Factors/genetics , Zinc Fingers
4.
Mech Dev ; 106(1-2): 77-83, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11472836

ABSTRACT

Sp3 is a ubiquitously expressed member of the Sp family of transcription factors. Recently, the mouse Sp3 gene has been disrupted by homologous recombination. Sp3 null mice die immediately after birth due to respiratory failure. In addition, these mice show a pronounced defect in late tooth formation. Here we show that Sp3 is also required for proper skeletal ossification. Both endochondral and intramembranous ossification are impaired in E18.5 Sp3-/- embryos. The delay in ossification is reflected by reduced expression of the osteoblast-specific marker gene osteocalcin. The transcription factor - core binding factor 1 (Cbfa1)--that is essential for bone formation, however, is expressed at normal levels. In vitro differentiation studies using Sp3-/- ES cells further support the conclusion that Sp3 is needed for correct bone formation. The capacity of Sp3-/- cells to undergo osteogenic differentiation in vitro is reduced and osteocalcin expression is significantly diminished. Our studies establish Sp3 as an essential transcription factor for late bone development.


Subject(s)
Bone and Bones/embryology , DNA-Binding Proteins/physiology , Neoplasm Proteins , Osteogenesis , Transcription Factors/physiology , Animals , Blotting, Northern , Cartilage/embryology , Cell Differentiation , Cell Line , Core Binding Factor Alpha 1 Subunit , Core Binding Factors , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Gene Expression , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mice , Mutation , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/genetics , Sp3 Transcription Factor , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Hum Mol Genet ; 10(4): 371-82, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11157800

ABSTRACT

We have cloned, sequenced and annotated segments of DNA spanning the mouse, chicken and pufferfish alpha globin gene clusters and compared them with the corresponding region in man. This has defined a small segment ( approximately 135-155 kb) of synteny and conserved gene order, which may contain all of the elements required to fully regulate alpha globin gene expression from its natural chromosomal environment. Comparing human and mouse sequences using previously described methods failed to identify the known regulatory elements. However, refining these methods by ranking identity scores of non-coding sequences, we found conserved sequences including the previously characterized alpha globin major regulatory element. In chicken and pufferfish, regions that may correspond to this element were found by analysing the distribution of transcription factor binding sites. Regions identified in this way act as strong enhancer elements in expression assays. In addition to delimiting the alpha globin chromosomal domain, this study has enabled us to develop a more sensitive and accurate routine for identifying regulatory elements in the human genome.


Subject(s)
Chromosomes/chemistry , Chromosomes/genetics , Globins/genetics , Multigene Family/genetics , Regulatory Sequences, Nucleic Acid , Animals , Base Sequence , Chickens , Conserved Sequence/genetics , CpG Islands/genetics , Evolution, Molecular , Fishes , Globins/chemistry , Humans , Mice , Molecular Sequence Data , Nucleic Acid Hybridization/methods , Protein Structure, Tertiary/genetics , Regulatory Sequences, Nucleic Acid/physiology
6.
EMBO J ; 19(21): 5856-63, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11060036

ABSTRACT

The CCAAT boxes of the beta-like globin genes interact with three proteins: NF-Y, GATA-1 and NFE-6. We demonstrate that NFE-6 contains C/EBPgamma, and address its role in globin gene regulation by erythroid overexpression of C/EBPgamma, and a dominant-negative form C/EBPgammaDeltaB, in mice. Elevated levels of C/EBPgamma, but not C/EBPgammaDeltaB, increase expression of the (fetal) gamma-globin relative to the (adult) beta-globin gene. Interestingly, fetal liver erythropoiesis is ablated when the C/EBPgamma and C/EBPgammaDeltaB levels are further increased in homozygous transgenics. We suggest that targeted expression of dominant-negative leucine zipper proteins is a generally applicable approach to ablate specific tissues in mice.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Erythropoiesis/genetics , Globins/genetics , Animals , Base Sequence , CCAAT-Binding Factor/genetics , DNA Primers/genetics , DNA-Binding Proteins/genetics , Erythroid-Specific DNA-Binding Factors , Fetal Blood/cytology , Fetal Blood/metabolism , GATA1 Transcription Factor , Gene Expression Regulation, Developmental , Leucine Zippers/genetics , Liver/embryology , Mice , Mice, Transgenic , Multigene Family , Transcription Factors/genetics
7.
EMBO J ; 19(18): 4986-96, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10990462

ABSTRACT

Locus control regions (LCRs) alleviate chromatin-mediated transcriptional repression. Incomplete LCRs partially lose this property when integrated in transcriptionally restrictive genomic regions such as centromeres. This frequently results in position effect variegation (PEV), i.e. the suppression of expression in a proportion of the cells. Here we show that this PEV is influenced by the heterochromatic protein SUV39H1 and by the Polycomb group proteins M33 and BMI-1. A concentration variation of these proteins modulates the proportion of cells expressing human globins in a locus-dependent manner. Similarly, the transcription factors Sp1 or erythroid Krüppel-like factor (EKLF) also influence PEV, characterized by a change in the number of expressing cells and the chromatin structure of the locus. However, in contrast to results obtained in a euchromatic locus, EKLF influences the expression of the gamma- more than the beta-globin genes, suggesting that the relief of silencing is caused by the binding of EKLF to the LCR and that genes at an LCR proximal position are more likely to be in an open chromatin state than genes at a distal position.


Subject(s)
Chromatin/metabolism , Globins/genetics , Suppression, Genetic , Transcription Factors/metabolism , Transcription, Genetic , Animals , Cell Line , Chromosome Mapping , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/metabolism , Gene Silencing , Globins/biosynthesis , Humans , In Situ Hybridization, Fluorescence , Kruppel-Like Transcription Factors , Liver/embryology , Liver/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Single-Strand Specific DNA and RNA Endonucleases/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/genetics , Transcriptional Activation
8.
Mol Reprod Dev ; 57(1): 4-15, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10954851

ABSTRACT

Previous studies have shown that the transcription of the fibroblast growth factor-4 (FGF-4) gene is regulated by a powerful enhancer located approximately three kilobases downstream of the transcription start site. Several conserved cis-regulatory elements in the promoter and the enhancer have been identified, including two Sp1 motifs located in the promoter and one Sp1 motif located in the enhancer. Each of these Sp1 motifs has been shown previously to bind the transcription factors Sp1 and Sp3 in vitro. The main objective of this study was to examine the potential interaction of the FGF-4 promoter and enhancer Sp1 motifs. Using site-directed mutagenesis, we demonstrate that disruption of these sites, individually or in combination, reduce the expression of FGF-4 promoter/reporter gene constructs in embryonal carcinoma cells. Importantly, we demonstrate that disruption of the enhancer Sp1 motif exerts a more pronounced effect on the expression of these constructs than disruption of the promoter Sp1 motifs. We also demonstrate that changing the spacing and the stereo-alignment of the enhancer Sp1 motif, relative to the other cis-regulatory elements of the enhancer, has little effect on the ability of the enhancer to stimulate transcription. Furthermore, embryonic stem cells that contain two disrupted Sp1 alleles were used to demonstrate that the transcription factor Sp1 is not necessary for expression of the endogenous FGF-4 gene. Finally, the significance of these findings relative to a looping model for the transcriptional activation of the FGF-4 gene is discussed.


Subject(s)
Fibroblast Growth Factors/genetics , Gene Expression Regulation , Proto-Oncogene Proteins/genetics , Sp1 Transcription Factor/metabolism , Amino Acid Motifs , Animals , Cell Line , Enhancer Elements, Genetic , Fibroblast Growth Factor 4 , High Mobility Group Proteins/metabolism , Mice , Promoter Regions, Genetic , Sp1 Transcription Factor/genetics , Transcription, Genetic
9.
Nature ; 406(6795): 519-24, 2000 Aug 03.
Article in English | MEDLINE | ID: mdl-10952313

ABSTRACT

GATA-1 is a tissue-specific transcription factor that is essential for the production of red blood cells. Here we show that overexpression of GATA-1 in erythroid cells inhibits their differentiation, leading to a lethal anaemia. Using chromosome-X-inactivation of a GATA-1 transgene and chimaeric animals, we show that this defect is intrinsic to erythroid cells, but nevertheless cell nonautonomous. Usually, cell nonautonomy is thought to reflect aberrant gene function in cells other than those that exhibit the phenotype. On the basis of our data, we propose an alternative mechanism in which a signal originating from wild-type erythroid cells restores normal differentiation to cells overexpressing GATA-1 in vivo. The existence of such a signalling mechanism indicates that previous interpretations of cell-nonautonomous defects may be erroneous in some cases and may in fact assign gene function to incorrect cell types.


Subject(s)
DNA-Binding Proteins/physiology , Erythropoiesis/physiology , Transcription Factors/physiology , Anemia/genetics , Animals , Animals, Genetically Modified , Apoptosis , Chimera , Crosses, Genetic , DNA-Binding Proteins/genetics , Dosage Compensation, Genetic , Erythroblasts/physiology , Erythroid Precursor Cells/physiology , Erythroid-Specific DNA-Binding Factors , Erythropoiesis/genetics , Female , GATA1 Transcription Factor , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Transcription Factors/genetics , X Chromosome
10.
EMBO J ; 19(4): 655-61, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10675334

ABSTRACT

Sp3 is a ubiquitously expressed transcription factor closely related to Sp1 (specificity protein 1). We have disrupted the mouse Sp3 gene by homologous recombination. Sp3-deficient embryos are growth retarded and invariably die at birth of respiratory failure. The cause for the observed breathing defect remains obscure since only minor morphological alterations were observed in the lung, and surfactant protein expression is indistinguishable from that in wild-type mice. Histological examinations of individual organs in Sp3(-/-) mice show a pronounced defect in late tooth formation. In Sp3 null mice, the dentin/enamel layer of the developing teeth is impaired due to the lack of ameloblast-specific gene products. Comparison of the Sp1 and Sp3 knockout phenotype shows that Sp1 and Sp3 have distinct functions in vivo, but also suggests a degree of functional redundancy.


Subject(s)
DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Odontogenesis/genetics , Odontogenesis/physiology , Transcription Factors/deficiency , Transcription Factors/genetics , Ameloblasts/metabolism , Animals , Animals, Newborn , Base Sequence , DNA Primers/genetics , DNA-Binding Proteins/physiology , Female , Gene Expression , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pregnancy , Respiratory Insufficiency/genetics , Respiratory Insufficiency/metabolism , Respiratory Insufficiency/pathology , Sp1 Transcription Factor/deficiency , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/physiology , Sp3 Transcription Factor , Tooth/growth & development , Tooth/metabolism , Tooth/pathology , Transcription Factors/physiology
11.
J Biol Chem ; 275(3): 1708-14, 2000 Jan 21.
Article in English | MEDLINE | ID: mdl-10636866

ABSTRACT

The monocyte chemoattractant protein-1 gene (MCP-1) is induced by the inflammatory cytokine tumor necrosis factor through the coordinate assembly of an NF-kappaB-dependent distal regulatory region and a proximal region that has been suggested to bind Sp1 as well as other factors. To provide a genetic correlation for Sp1 activity in this system, a cell line homozygous for a targeted truncation of the Sp1 gene was derived and examined. We found that the lack of Sp1 binding activity resulted in the inability of both the distal and proximal regions to assemble in vivo even though the binding of NF-kappaB to distal region DNA was unaffected in vitro. We also found that Sp1 and NF-kappaB were the minimal mammalian transcription factors required for efficient activity when transfected into Drosophila Schneider cells. Additionally, Sp3 was able to compensate for Sp1 in the Drosophila tissue cell system but not in the Sp1(-/-) cell line suggesting that Sp1 usage is site-specific and is likely to depend on the context of the binding site. Together, these data provide genetic and biochemical proof for Sp1 in regulating the MCP-1 gene.


Subject(s)
Chemokine CCL2/genetics , DNA-Binding Proteins/physiology , Promoter Regions, Genetic , Protein Kinases/physiology , Tumor Necrosis Factor-alpha/metabolism , 3T3 Cells , Animals , Base Sequence , Blotting, Northern , Cell Line , Chemokine CXCL10 , Chemokines, CXC/metabolism , DNA Footprinting , Drosophila/genetics , Gene Expression Regulation , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutagenesis , NF-kappa B/metabolism , Protein Binding , Recombinant Proteins/metabolism , Time Factors , Transfection
12.
Cell Mol Biol (Noisy-le-grand) ; 45(5): 709-16, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10512200

ABSTRACT

Oct-4 is expressed in embryonic stem cells and is one of the first transcription factors differentially regulated during mouse development. Recent ablation of Oct-4 function in vivo has demonstrated that its expression is essential for preventing embryonic cell differentiation. Several studies have indicated that activation of the Oct-4 TATA-less promoter is mediated by a GC-box representing a high affinity binding site for the transcription factor Sp1. In this study, we have analyzed Oct-4 expression and the activity of the Oct-4 promoter GC box in Sp1 deficient ES cells. We found that in these cells, Oct-4 RNA and protein levels were comparable to wt ES cells and the activity of the Oct-4 promoter GC-box was even increased. Furthermore, the pattern of Oct-4 down-regulation in Sp1 deficient ES cells was unchanged compared to wt cells. Analysis of GC-box binding proteins in extracts from Sp1-/- ES cells revealed that Sp3, another member of the Sp1 transcription factors family, efficiently bound to Sp1 site. These results show that in ES cells Sp1 is not essential for Oct-4 gene activity. Thus, Sp1 function in undifferentiated embryonic cells may be complemented by Sp3 and/or by other GC-box binding transcription factors.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Animals , Electrophoresis , Gene Expression Profiling , Mice , Octamer Transcription Factor-3 , Promoter Regions, Genetic , Sp1 Transcription Factor/genetics , Sp3 Transcription Factor , Stem Cells , Transcription Factors/genetics , Transfection
13.
Nucleic Acids Res ; 27(15): 2991-3000, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10454592

ABSTRACT

One of the most common regulatory elements is the GC box and the related GT/CACC box, which are widely distributed in promoters, enhancers and locus control regions of housekeeping as well as tissue-specific genes. For long it was generally thought that Sp1 is the major factor acting through these motifs. Recent discoveries have shown that Sp1 is only one of many transcription factors binding and acting through these elements. Sp1 simply represents the first identified and cloned protein of a family of transcription factors characterised by a highly conserved DNA-binding domain consisting of three zinc fingers. Currently this new family of transcription factors has at least 16 different mammalian members. Here, we will summarise and discuss recent advances that have been directed towards understanding the biological role of these proteins.


Subject(s)
Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Zinc Fingers , Amino Acid Sequence , Animals , Gene Expression Regulation , Humans , Molecular Sequence Data , Sequence Homology, Amino Acid , Sp1 Transcription Factor/chemistry , Sp1 Transcription Factor/classification , Terminology as Topic , Transcription Factors/chemistry , Transcription Factors/classification
14.
Biochem Biophys Res Commun ; 259(2): 364-9, 1999 Jun 07.
Article in English | MEDLINE | ID: mdl-10362515

ABSTRACT

Analysis of the human Bruton's agammaglobulinemia tyrosine kinase (Btk) gene promoter revealed that 280 bp upstream of the transcriptional start site is sufficient for a cell restricted expression pattern. Here, the interplay of the transcription factors Sp1, Sp3, and PU.1 binding to this promoter area was analysed. All three proteins are able to independently activate the promoter in Drosophila Schneider (SL2) cells lacking endogenous Sp- or PU.1-like activities. Furthermore, PU.1 is able to act synergistically with Sp1 as well as Sp3 to transactivate the promoter. This transactivation is mediated through adjacent binding sites rather than through the more distant Sp binding site, suggesting a possible direct interaction between PU.1 and Sp1/3. Expression of Btk was found in ES cells and levels of expression were the same as in ES cells with a targeted deletion of the Sp1 gene, suggesting that Sp3 acts as a positive regulator of Btk in vivo, in the absence of Sp1.


Subject(s)
DNA-Binding Proteins/genetics , Promoter Regions, Genetic , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Sp1 Transcription Factor/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Transcriptional Activation/genetics , Agammaglobulinaemia Tyrosine Kinase , Animals , Cells, Cultured , DNA Mutational Analysis , DNA-Binding Proteins/pharmacology , Drosophila , Gene Expression Regulation/genetics , Humans , Proto-Oncogene Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Sp1 Transcription Factor/pharmacology , Sp3 Transcription Factor , Trans-Activators/pharmacology , Transcription Factors/pharmacology , Transfection
15.
Eur J Hum Genet ; 7(3): 345-56, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10234511

ABSTRACT

We have studied 31 beta-thalassaemia intermedia, 30 beta-thalassaemia major patients and 50 normal individuals from Turkey, determining the relationship between the nucleotide variations in beta-globin gene cluster, the altered levels of foetal haemoglobin and the relative ratios of beta- and gamma mRNAs. We have found in beta-thalassaemia intermedia patients with high foetal haemoglobin expression that the three nucleotide variations in the 5' sequences of the gamma globin genes, A-->G at G gamma - 1396, the T-->C at A gamma - 228, and the GA-->AG at A gamma - 603/4, are linked to haplotype II in haplotypic homozygotes and the (AT)8N14(AT)7 motif in beta LCR. Conversely, the three single nucleotide substitutions in the 5' sequences of gamma globin genes, the G-->A at G gamma - 1225, the A-->G at A gamma + 25 and the C-->G at A gamma - 369, which have a strong linkage with haplotype I, V or VI in haplotypic homozygotes and the (AT)10N12(AT)12 and the (AT)9N12(AT)12 motifs in HS-2 of beta LCR are all associated with low foetal haemoglobin levels. The number of nucleotide changes in beta-globin gene cluster implied in our study are not the primary cause of the differences in haemoglobin F levels. They perhaps may contribute to the variations in the clinical severity observed among beta thalassaemia intermedia and major patients with other yet unknown gene conversions.


Subject(s)
Fetal Hemoglobin/genetics , Globins/genetics , Polymorphism, Genetic , Promoter Regions, Genetic , beta-Thalassemia/genetics , Adolescent , Adult , Child , Humans , Middle Aged
16.
Genes Dev ; 12(18): 2863-73, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9744863

ABSTRACT

The locus control region of the beta-globin cluster contains five DNase I hypersensitive sites (5'HS1-5) required for locus activation. 5'HS3 contains six G-rich motifs that are essential for its activity. Members of a protein family, characterized by three zinc fingers highly homologous to those found in transcription factor Sp1, interact with these motifs. Because point mutagenesis cannot distinguish between family members, it is not known which protein activates 5'HS3. We show that the function of such closely related proteins can be distinguished in vivo by matching point mutations in 5'HS3 with amino acid changes in the zinc fingers of Sp1 and EKLF. Testing their activity in transgenic mice shows that EKLF is a direct activator of 5'HS3.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA/genetics , DNA/metabolism , Globins/genetics , Mutation , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , CHO Cells , Cell Line , Cricetinae , Deoxyribonuclease I , Erythrocytes/metabolism , Gene Expression , Genes, Reporter , Humans , In Situ Hybridization , Kruppel-Like Transcription Factors , Locus Control Region , Mice , Mice, Transgenic , Molecular Sequence Data , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection , Zinc Fingers/genetics
17.
EMBO J ; 17(8): 2334-41, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9545245

ABSTRACT

Disruption of the gene for transcription factor EKLF (erythroid Krüppel-like factor) results in fatal anaemia caused by severely reduced expression of the adult beta-globin gene, while other erythroid-specific genes, including the embryonic epsilon- and fetal gamma-globin genes, are expressed normally. Thus, EKLF is thought to be a stage-specific factor acting through the CACC box in the beta-gene promoter, even though it is already present in embryonic red cells. Here, we show that a beta-globin gene linked directly to the locus control region (LCR) is expressed at embryonic stages, and that this is only modestly reduced in EKLF-/- embryos. Thus, embryonic beta-globin expression is not intrinsically dependent on EKLF. To investigate whether EKLF functions in the locus control region, we analysed the expression of LCR-driven lacZ reporters. This shows that EKLF is not required for reporter activation by the complete LCR. However, embryonic expression of reporters driven by 5'HS3 of the LCR requires EKLF. This suggests that EKLF interacts directly with the CACC motifs in 5'HS3 and demonstrates that EKLF is also a transcriptional activator in embryonic erythropoiesis. Finally, we show that overexpression of EKLF results in an earlier switch from gamma- to beta-globin expression. Adult mice with the EKLF transgene have reduced platelet counts, suggesting that EKLF levels affect the balance between the megakaryocytic and erythroid lineages. Interestingly, the EKLF transgene rescues the lethal phenotype of EKLF null mice, setting the stage for future studies aimed at the analysis of the EKLF protein and its role in beta-globin gene activation.


Subject(s)
DNA-Binding Proteins/metabolism , Erythroid Precursor Cells/metabolism , Globins/genetics , Transcription Factors/metabolism , Transcriptional Activation , Animals , DNA-Binding Proteins/genetics , Erythropoiesis , Gene Expression Regulation , Humans , Kruppel-Like Transcription Factors , Mice , Mice, Transgenic , Phenotype , Transcription Factors/genetics
18.
Cell ; 89(4): 619-28, 1997 May 16.
Article in English | MEDLINE | ID: mdl-9160753

ABSTRACT

Transcription factor Sp1 has been implicated in the expression of many genes. Moreover, it has been suggested that Sp1 is linked to the maintenance of methylation-free CpG islands, the cell cycle, and the formation of active chromatin structures. We have inactivated the mouse Sp1 gene. Sp1-/- embryos are retarded in development, show a broad range of abnormalities, and die around day 11 of gestation. In Sp1-/- embryos, the expression of many putative target genes, including cell cycle-regulated genes, is not affected, CpG islands remain methylation free, and active chromatin is formed at the globin loci. However, the expression of the methyl-CpG-binding protein MeCP2 is greatly reduced in Sp1-/- embryos. MeCP2 is thought to be required for the maintenance of differentiated cells. We suggest that Sp1 is an important regulator of this process.


Subject(s)
Embryonic and Fetal Development/physiology , Sp1 Transcription Factor/physiology , Animals , Base Sequence , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Division/genetics , Cell Division/physiology , Chimera , CpG Islands , DNA Methylation , DNA Primers/genetics , Embryonic and Fetal Development/genetics , Female , Gene Targeting , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Sp1 Transcription Factor/genetics
19.
Genes Funct ; 1(1): 11-24, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9680325

ABSTRACT

GATA-1 is a tissue-specific DNA-binding protein containing two zinc-finger-like domains. It is expressed predominantly in erythrocytes. Consensus binding sites for GATA-1 have been found in the regulatory elements of all erythroid-specific genes examined. GATA-1 protein is required for erythroid differentiation beyond the proerythroblast stage. In this paper, we demonstrate that the overexpression of GATA-1 in murine erythroleukaemia (MEL) cells alleviates DMSO-induced terminal erythroid differentiation. Hence, there is no induction of globin gene transcription and the cells do not arrest in the G1 phase of the cell cycle. Furthermore, we demonstrate that expression of GATA-1 in non-transformed erythroid precursors also affects their proliferative capacity and terminal differentiation, as assayed by adult globin gene transcription. To gain insight into the mechanism of this effect, we studied the levels and activities of regulators of cell-cycle progression during DMSO-induced differentiation. A decrease in cyclin D-dependent kinase activity was observed during the induction of both control and GATA-1-overexpressing MEL cells. However, cyclin E-dependent kinase activity decreased more than 20-fold in control but less than 2-fold in GATA-1-overexpressing MEL cells upon induction. Thus GATA-1 may exert its effects by regulating cyclin E-dependent kinase activity. We also show that GATA-1 binds to the retinoblastoma protein in vitro, but not to the related protein p107, which may indicate that GATA-1 interacts directly with specific members of the cell-cycle machinery in vivo. We conclude that GATA-1 regulates cell fate, in terms of differentiation or proliferation, by affecting the cell-cycle apparatus.


Subject(s)
Cell Cycle/genetics , DNA-Binding Proteins/physiology , Erythroblasts/cytology , Transcription Factors/physiology , Animals , Cell Differentiation , Cyclin-Dependent Kinases/metabolism , DNA-Binding Proteins/genetics , Dimethyl Sulfoxide , Erythroblasts/chemistry , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Gene Expression Regulation, Developmental , Globins/genetics , Humans , Leukemia, Experimental , Mice , Nuclear Proteins/metabolism , Phosphorylation , Protein Binding , Recombinant Fusion Proteins , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107 , Transcription Factors/genetics , Transcription, Genetic , Tumor Cells, Cultured
20.
Development ; 122(12): 3991-9, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9012519

ABSTRACT

The beta-globin locus control region (LCR) is contained on a 20 kb DNA fragment and is characterized by the presence of five DNaseI hypersensitive sites in erythroid cells, termed 5'HS1-5. A fully active 6.5 kb version of the LCR, called the muLCR, has been described. Expression of the beta-like globin genes is absolutely dependent on the presence of the LCR. The developmental expression pattern of the genes in the cluster is achieved through competition of the promoters for the activating function of the LCR. Transgenic mice experiments suggest that subtle changes in the transcription factor environment lead to the successive silencing of the embryonic epsilon-globin and fetal gamma-globin promoters, resulting in the almost exclusive transcription of the beta-globin gene in adult erythropoiesis. In this paper, we have asked the question whether the LCR and its individual hypersensitive sites 5'HS1-4 can activate a basic promoter in the absence of any other globin sequences. We have employed a minimal promoter derived from the mouse Hsp68 gene driving the bacterial beta-galactosidase (lacZ) gene. The results show that the muLCR and 5'HS3 direct erythroid-specific, embryonic expression of this construct, while 5'HS1, 5'HS2 and 5'HS4 are inactive at any stage of development. Expression of the muLCR and 5'HS3 transgenes is repressed during fetal stages of development. The transgenes are in an inactive chromatin conformation and the lacZ gene is not transcribed, as shown by in situ hybridization. These data are compatible with the hypothesis that the LCR requires the presence of an active promoter to adopt an open chromatin conformation and with models proposing progressive heterochromatization during embryogenesis. The results suggest that the presence of a beta-globin gene is required for LCR function as conditions become more stringent during development.


Subject(s)
Erythropoiesis/genetics , Gene Expression Regulation, Developmental , Globins/genetics , Regulatory Sequences, Nucleic Acid , Animals , Binding Sites , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , Humans , In Situ Hybridization , Lac Operon , Liver/embryology , Mice , Mice, Transgenic , Promoter Regions, Genetic , Recombinant Proteins/biosynthesis , Transcription, Genetic , beta-Galactosidase/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...