Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 276(29): 26883-92, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11304527

ABSTRACT

The active elements of the beta-globin locus control region (LCR) are located within domains of unique chromatin structure. These nuclease hypersensitive sites (HSs) are characterized by high DNase I sensitivity, erythroid specificity, similar nucleosomal structure, and evolutionarily conserved clusters of cis-acting elements that are required for the formation and function of the core elements. To determine the requirements for HS core formation in the setting of nuclear chromatin, we constructed a series of artificial HS cores containing binding sites for GATA-1, NF-E2, and Sp1. In contrast to the results of previous in vitro experiments, we found that when constructs were stably integrated in mouse erythroleukemia cells the binding sites for NF-E2, GATA-1, or Sp1 alone or in any combination were unable to form core HS structures. We subsequently identified two new cis-acting elements from the LCR HS4 core that, when combined with the NF-E2, Sp1, and tandem inverted GATA elements, result in core structure formation. Both new cis-acting elements bind Sp1, and one binds erythroid Kruppel-like factor (EKLF). We conclude that in vivo beta-globin LCR HS core formation is more complex than previously thought and that several factors are required for this process to occur.


Subject(s)
Globins/genetics , Locus Control Region , Transcription Factors/metabolism , Base Sequence , Binding Sites , DNA, Complementary , Deoxyribonuclease I/metabolism , Globins/metabolism , Humans , Molecular Sequence Data , Mutagenesis
2.
Blood Cells Mol Dis ; 27(4): 767-80, 2001.
Article in English | MEDLINE | ID: mdl-11778661

ABSTRACT

In erythroid tissues the chromatin structure of the beta-globin gene locus is extensively remodeled. Changes include the formation of DNase I hypersensitive sites (HSs) over the promoters of actively expressed genes. To test the hypothesis that such "opening" of promoter chromatin structure is important for beta-globin gene expression, we placed a 101-bp erythroid-specific hypersensitive-site forming element (HSFE) from the core of LCR HS4 immediately upstream of a minimal beta-globin gene promoter. We then studied the effects of this element alone and in combination with other cis-acting elements on globin gene chromatin structure and gene expression in MEL cells and transgenic mice. Single or tandem HSFEs increased the size of the portion of the promoter accessible to DNase digestion, increased the proportion of promoters in an accessible conformation, and increased gene expression approximately 5-fold. These were equivalent to expression levels attained using a 2.8-kb microLCR construct. Inclusion of the LCR HS2 enhancer did not increase expression further. In transgenic mouse fetal liver cells the HSFE increased average expression 2.5-fold compared to the minimal promoter alone. These results indicate that a small cis-acting element is capable of remodeling local beta-globin promoter chromatin structure and producing expression similar to that seen with a microLCR construct.


Subject(s)
Chromatin/genetics , Gene Expression Regulation , Gene Rearrangement , Globins/genetics , Locus Control Region , Promoter Regions, Genetic , Animals , Chromatin/ultrastructure , DNA, Recombinant/drug effects , DNA, Recombinant/genetics , Deoxyribonuclease I/pharmacology , Enhancer Elements, Genetic , Globins/biosynthesis , Humans , Leukemia, Erythroblastic, Acute/pathology , Locus Control Region/genetics , Mice , Mice, Transgenic , RNA, Messenger/biosynthesis , Transfection , Tumor Cells, Cultured
3.
Gene Ther ; 7(8): 653-63, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10800088

ABSTRACT

One problem limiting the development of long-term gene replacement therapy is gene silencing. A variety of experiments have implicated DNA methylation and histone deacetylation in gene silencing and shown that the agents 5-azacytidine (5-Aza) and trichostatin A (TSA) are able to reverse these effects. To begin to investigate clinically relevant strategies to reverse silencing with these drugs, we transduced the MEL and FDCP-1 hematopoietic cell lines with Moloney murine leukemia virus (MMLV) and Harvey murine sarcoma virus (HMSV)-based retroviral vectors carrying the beta-galactosidase/neomycin resistance fusion gene (beta-geo). Fifty-one clones were isolated under G418 selection over 2 weeks and then allowed to grow without selection as beta-gal activity was monitored over time. More than 80% of these clones showed significant silencing over a period of 70-80 days. The clones were then exposed to a wide range of 5-Aza and TSA concentrations, both alone and in combination, in an effort to reverse silencing. Despite demonstration that the agents were able to decrease DNA methylation and increase histone acetylation, significant reversal of long-term silencing was not seen under any experimental condition. These results suggest that long-term retroviral silencing involves mechanisms in addition to DNA methylation and histone acetylation and that new pharmacologic strategies are needed to overcome the silencing process.


Subject(s)
Gene Silencing/drug effects , Genetic Therapy/methods , Genetic Vectors/genetics , Histone Deacetylase Inhibitors , Hydroxamic Acids/therapeutic use , Retroviridae/genetics , Acetylation , Acetyltransferases/antagonists & inhibitors , Azacitidine/therapeutic use , Cell Line , DNA Methylation/drug effects , Dose-Response Relationship, Drug , Drug Resistance , Enzyme Inhibitors/therapeutic use , Histones/metabolism , Humans , Treatment Failure , beta-Galactosidase/genetics
4.
Blood Cells Mol Dis ; 25(1): 47-60, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10349513

ABSTRACT

The chromatin structure of the human beta-globin gene locus assumes a transcriptionally-active conformation in erythroid cells. One feature of this chromatin reorganization is the formation of DNase 1 hypersensitive sites in the regions of active globin gene promoters. This reorganization requires the globin locus control region and is associated with normal expression of the beta-like globin genes. To determine whether it is possible to artificially enhance the opening of the chromatin structure of a minimal beta-globin promoter, we placed a 101bp, erythroid-specific DNase 1 hypersensitive site-forming element (HSFE) immediately upstream of the beta-globin promoter and gene. This element includes binding sites for NF-E2, AP-1, GATA-1 and Sp-1. Constructs were stably transfected into murine erythroleukemia cells and promoter chromatin structure and gene expression were analyzed. The HSFE induced an area of enhanced DNase 1 hypersensitivity extending from the transcriptional start site to -300bp of the artificial promoter and significantly increased the proportion of beta-globin promoters in an open chromatin configuration. This remodeling of promoter chromatin structure resulted in 3-fold increases in beta-globin gene transcription and induction, and inhibited long-term beta-globin gene silencing. These results indicate that a relatively small cis-acting element is able to enhance remodeling of promoter chromatin structure resulting in increased beta-globin gene expression.


Subject(s)
Chromatin/genetics , DNA/genetics , Globins/genetics , Promoter Regions, Genetic , Chromatin/ultrastructure , DNA-Binding Proteins/genetics , Erythrocytes , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Gene Expression Regulation , Humans , NF-E2 Transcription Factor , NF-E2 Transcription Factor, p45 Subunit , Transcription Factors/genetics , Transcription, Genetic
5.
Br J Cancer ; 79(2): 221-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9888461

ABSTRACT

In the present study, we investigated the mechanisms controlling constitutive transcription of collagenase-1 and its repression by all-trans-retinoic acid (RA) in the highly invasive metastatic and oestrogen-receptor-negative breast cancer cell line MDA231. A combination of in vivo and in vitro experiments that include DNAase I hypersensitivity assays, transient transfection of collagenase-1 promoter constructs, and electrophoretic mobility shift assays implicate several PEA3 sites, binding sites for Ets-related transcription factors, in the constitutive expression of the human collagenase-1 promoter. Transient transfection of promoter constructs linked to the luciferase reporter, along with gel retardation assays, revealed that repression of collagenase-1 transcription by RA is not dependent on the proximal AP-1 site, but, rather, requires sequences located in distal regions of the promoter. Transcriptional analyses and electrophoretic mobility shift assays suggest that the PEA3 site located at -3108 bp facilitates, at least in part, the transcriptional repression of the human collagenase-1 gene in MDA231 cells. We conclude that collagenase-1 repression in MDA231 cells occurs by a novel regulatory pathway that does not depend on the proximal AP-1 site at -73 bp, but does depend on distal regions in the collagenase-1 promoter.


Subject(s)
Collagenases/biosynthesis , Neoplasm Proteins/biosynthesis , Receptors, Retinoic Acid/drug effects , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Collagenases/genetics , Deoxyribonuclease I/pharmacology , Enzyme Repression/genetics , Female , Gene Expression/drug effects , Humans , Matrix Metalloproteinase 1 , Matrix Metalloproteinase Inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Transfection , Tumor Cells, Cultured/drug effects , Retinoic Acid Receptor gamma
6.
Blood ; 93(2): 703-12, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9885233

ABSTRACT

The function of the beta-globin locus control region (LCR) has been studied both in cell lines and in transgenic mice. We have previously shown that when a 248-kb beta-locus YAC was first microinjected into L-cells and then transferred into MEL cells by fusion, the YAC loci of the LxMEL hybrids displayed normal expression and developmental regulation.To test whether direct transfer of a beta-globin locus (beta-YAC) into MEL cells could be used for studies of the function of the LCR, a 155-kb beta-YAC that encompasses the entire beta-globin locus was used. This YAC was retrofitted with a PGK-neo selectable marker and with two I-PpoI sites at the vector arm-cloned insert junctions, allowing detection of the intact globin loci on a single I-PpoI fragment by pulsed field gel electrophoresis (PFGE). The Ppo-155 beta-YAC was used to directly lipofect MEL 585 cells. In 7 beta-YAC MEL clones with at least one intact copy of the YAC, the levels of total human globin mRNA (ie, epsilon + gamma + beta) per copy of integrated beta-YAC varied more than 97-fold between clones. These results indicated that globin gene expression was strongly influenced by the position of integration of the beta-YAC into the MEL cell genome and suggested that the LCR cannot function properly when the locus is directly transferred into an erythroid cell environment as naked beta-YAC DNA. To test whether passage of the beta-YAC through L-cells before transfer into MEL cells was the reason for the previously observed correct developmental regulation of human globin genes in the LxMEL hybrid cells, we transfected the YAC into L-cells by lipofection. Three clones carried the intact 144-kb I-PpoI fragment and transcribed the human globin genes with a fetal-like pattern. Subsequent transfer of the YAC of these L(beta-YAC) clones into MEL cells by fusion resulted in LxMEL hybrids that synthesized human globin mRNA. The variation in human beta-globin mRNA (ie, epsilon + gamma + beta) levels between hybrids was 2.5-fold, indicating that globin gene expression was independent of position of integration of the transgene, as expected for normal LCR function. The correct function of the LCR when the YAC is first transferred into the L-cell environment raises the possibility that normal activation of the LCR requires interaction with the transcriptional environment of an uncommitted, nonerythroid cell. We propose that the activation of the LCR may represent a multistep process initiated by the binding of ubiquitous transcription factors early during the differentiation of hematopoietic stem cells and completed with the binding of erythroid type of factors in the committed erythroid progenitors.


Subject(s)
Erythrocytes/metabolism , Gene Expression , Globins/genetics , Regulatory Sequences, Nucleic Acid , Transfection , Animals , Cell Fusion , Cell Line , Chromosomes, Artificial, Yeast , Erythroid Precursor Cells/metabolism , Humans , Hybrid Cells , L Cells/metabolism , Mice , Mice, Transgenic , Microinjections , RNA, Messenger/metabolism
7.
J Biol Chem ; 274(3): 1801-13, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9880563

ABSTRACT

Proliferation, differentiation, and apoptosis are tightly regulated during hematopoiesis, allowing amplification along specific lineages while preventing excessive proliferation of immature cells. The MCL1 member of the BCL2 family is up-regulated during the induction of monocytic differentiation (approximately 10-fold with 12-O-tetradecanoylphorbol 13-acetate (TPA)). MCL1 has effects similar to those of BCL2, up-regulation promoting viability, but differs from BCL2 in its rapid inducibility and its pattern of expression. Nuclear factors that regulate MCL1 transcription have now been identified, extending the previous demonstration of signal transduction through mitogen-activated protein kinase. A 162-base pair segment of the human MCL1 5'-flank was found to direct luciferase reporter activity, allowing approximately 10-fold induction with TPA that was suppressible upon inhibition of the extracellular signal-regulated kinase (ERK) pathway. Serum response factor (SRF), Elk-1, and Sp1 bound to cognate sites within this segment, SRF and Elk-1 acting coordinately to affect both basal activity and TPA inducibility, whereas Sp1 affected basal activity only. Thus, the mechanism of the TPA-induced increase in MCL1 expression seen in myelomonocytic cells at early stages of differentiation involves signal transduction through ERKs and transcriptional activation through SRF/Elk-1. This finding provides a parallel to early response genes (e.g. c-FOS and EGR1) that affect maturation commitment in these cells and therefore suggests a means through which enhancement of cell viability may be linked to the induction of differentiation.


Subject(s)
DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/cytology , Neoplasm Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Differentiation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Humans , Molecular Sequence Data , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Serum Response Factor , Tetradecanoylphorbol Acetate/pharmacology , Transcriptional Activation/drug effects , Tumor Cells, Cultured , Up-Regulation , ets-Domain Protein Elk-1
8.
Hematology ; 4(5): 437-55, 1999.
Article in English | MEDLINE | ID: mdl-27426849

ABSTRACT

The cloning of the human ß-globin genes more than 20 years ago led to predictions that ß-thalassemia and sickle cell disease would be among the first monogenic diseases to be successfully treated by gene replacement therapy. However, despite the worldwide enrollment of more than 3,000 patients in approved gene transfer protocols, none have involved therapy for these diseases. This has been due to several technical hurdles that need to be overcome before gene replacement therapy for ß-thalassemia and sickle cell disease can become practical. These problems include inefficient transduction of hematopoietic stem cells and an inability to achieve consistent, long-term, high-level expression of transferred ß-like globin genes with current gene transfer vectors. In this review we highlight the relationships between understanding the fundamental mechanisms of ß-globin gene locus function and basic vector biology and the development of strategies for ß-globin gene replacement therapy. Despite slow initial progress in this field, recent advances in a variety of critical areas provide hope that clinical trials may not be far away.

9.
Nucleic Acids Res ; 26(24): 5684-91, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9838000

ABSTRACT

Proper expression of the genes of the human beta-globin gene locus requires the associated locus control region (LCR). Structurally, the LCR is defined by the presence of four domains of erythroid-specific chromatin structure. These domains, which have been characterized as DNase I hypersensitive sites (HSs), comprise the active elements of the LCR. The major focus of this research is to define the cis -acting elements which are required for the formation of these domains of unique chromatin structure. Our previous investigations on the formation of LCR HS4 demonstrated that NF-E2 and tandem, inverted GATA binding sites are required for the formation of the native HS. Similarly arranged NF-E2 and tandem GATA sites are present within the core regions of the other human LCR HSs and are evolutionarily conserved. Using site-directed mutagenesis of human HSs 2 and 3 we have tested the hypothesis that these NF-E2 and GATA sites are common requirements for the formation of all LCR HSs. We find that mutation of these elements, and particularly the GATA elements, results in a decrease or complete loss of DNase I hypersensitivity. These data imply the presence of common structural elements within the core of each LCR HS which are required for erythroid-specific chromatin structure reorganization.


Subject(s)
Chromatin/genetics , DNA-Binding Proteins/genetics , Globins/genetics , Hemoglobins/genetics , Transcription Factors/genetics , Base Sequence , Chromatin/ultrastructure , Conserved Sequence , Erythrocytes/metabolism , Erythrocytes/ultrastructure , Erythroid-Specific DNA-Binding Factors , GATA2 Transcription Factor , Humans , Molecular Sequence Data , Mutation , NF-E2 Transcription Factor , NF-E2 Transcription Factor, p45 Subunit , Sequence Analysis, DNA
10.
Nucleic Acids Res ; 25(14): 2917-22, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9207043

ABSTRACT

HS-40 is the major regulatory element of the human alpha-globin locus, located 40 kb upstream of the zeta-globin gene. To test for potential interactions between HS-40 and the beta- or the gamma-globin gene promoters in stable transfection assays, the HS-40 core sequence was cloned upstream of either the beta promoter or the gamma promoter driving the neomycin phosphotransferase gene and enhancer activity was measured using a colony assay. In K562 or in MEL cells, enhancer activity of HS-40 was higher than that of the individual core sequences of the DNase I hypersensitive sites (HS) of the beta-globin locus control region (LCR), and approximately 60% of the enhancer activity of a 2.5 kb microLCR, which contains the core elements of DNase I hypersensitive sites 1-4. In contrast to the synergistic interaction between the DNase I hypersensitive sites of beta locus LCR, combination of HS-40 with these DNase I hypersensitive sites failed to display cooperativity in K562 cells and inhibited enhancer function in MEL cells. Inhibition of enhancer function was also observed when two copies of the HS-40 were arranged tandemly. We conclude that the core element of HS-40 (i) is a powerful enhancer of gamma- and beta-globin gene expression, (ii) in contrast to other classical enhancers, acts best as a single copy, (iii) does not cooperate with the regulatory elements of the beta-globin locus control region.


Subject(s)
Enhancer Elements, Genetic , Globins/genetics , Multigene Family , Animals , Binding Sites , Deoxyribonuclease I , Humans , Mice , Promoter Regions, Genetic , Repetitive Sequences, Nucleic Acid , Tumor Cells, Cultured
11.
EMBO J ; 14(1): 106-16, 1995 Jan 03.
Article in English | MEDLINE | ID: mdl-7828582

ABSTRACT

The beta-like globin genes require the upstream locus control region (LCR) for proper expression. The active elements of the LCR coincide with strong erythroid-specific DNase I-hypersensitive sites (HSs). We have used 5' HS4 as a model to study the formation of these HSs. Previously, we identified a 101 bp element that is required for the formation of this HS. This element binds six proteins in vitro. We now report a mutational analysis of the HS4 HS-forming element (HSFE). This analysis indicates that binding sites for the hematopoietic transcription factors NF-E2 and GATA-1 are required for the formation of the characteristic chromatin structure of the HS following stable transfection into murine erythroleukemia cells. Similarly arranged NF-E2 and GATA binding sites are present in the other HSs of the human LCR, as well as in the homologous mouse and goat sequences and the chicken beta-globin enhancer. A combination of DNase I and micrococcal nuclease sensitivity assays indicates that the characteristic erythroid-specific hypersensitivity of HS4 to DNase I is the result of tissue-specific alterations in both nucleosome positioning and tertiary DNA structure.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , Globins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/metabolism , Animals , Base Sequence , DNA Mutational Analysis , Deoxyribonuclease I/metabolism , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Gene Expression Regulation , Hematopoiesis/genetics , Humans , Leukemia, Erythroblastic, Acute , Mice , Models, Genetic , Molecular Sequence Data , NF-E2 Transcription Factor , NF-E2 Transcription Factor, p45 Subunit , Nucleosomes/metabolism , Protein Binding , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured
12.
Blood ; 84(4): 1298-308, 1994 Aug 15.
Article in English | MEDLINE | ID: mdl-8049444

ABSTRACT

A cis-acting DNA regulatory element 3' to the A gamma-globin gene contains eight distinct regions of DNA-protein interaction distributed over 750 bp of DNA. The sequences of two foot-printed regions (sites I and IV) are A-T rich and generate a highly retarded complex on gel shift analysis with nuclear extract from human erythroleukemia (K562) cells. We have purified a 98-kD protein that reproduces this gel shift. Tryptic cleavage and peptide sequence analysis demonstrated that the 98-kD protein is identical to a recently cloned protein, special A-T-rich binding protein 1 (SATB1), that binds selectively to nuclear matrix/scaffold-associated regions of DNA (MARs/SARs). We have shown by functional analysis that the 3' A gamma regulatory element associates with the nuclear matrix. SATB1 mRNA was identified in K562 cells, and reverse transcriptase-polymerase chain reaction (RT-PCR) demonstrated its transcript in several other hematopoietic lines. Antisera to SATB1 caused ablation of the gel shift complex generated by both the crude nuclear extract and the purified 98-kD protein with the site I oligonucleotide. Furthermore, oligonucleotides that bind SATB1 inhibited formation of the site I gel shift complex when added as excess unlabeled competitor. An immunoblot analysis of the site I gel shift complex documented the presence of SATB1. Binding of SATB1 to two sites within the 3' A gamma regulatory element and its MAR/SAR activity suggests that this element may influence gene expression through interaction with the nuclear matrix.


Subject(s)
DNA-Binding Proteins/metabolism , Globins/genetics , Matrix Attachment Region Binding Proteins , Nuclear Matrix/metabolism , Regulatory Sequences, Nucleic Acid , Amino Acid Sequence , Base Sequence , Binding Sites , Cell Line , DNA-Binding Proteins/isolation & purification , Electrophoresis, Polyacrylamide Gel , Globins/metabolism , Humans , Leukemia, Erythroblastic, Acute , Molecular Sequence Data , Mutagenesis , Oligodeoxyribonucleotides , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Tumor Cells, Cultured
13.
Proc Natl Acad Sci U S A ; 90(23): 11207-11, 1993 Dec 01.
Article in English | MEDLINE | ID: mdl-8248229

ABSTRACT

We demonstrate that transfer of a yeast artificial chromosome (YAC) containing 230 kb of the human beta-globin locus into mouse erythroleukemia cells by fusion results in correct developmental regulation of the human beta-like globin genes. Additionally, we show that early after hybrid formation, human embryonic epsilon- and fetal gamma-globin genes are coexpressed with the adult beta gene but that after 10-20 weeks in culture, globin gene expression switches to predominantly adult. Thus, in contrast to shorter gene constructs, the globin genes of the beta-globin locus YAC are regulated like the chromosomal globin genes. These results indicate that transfer of YACs into established cell lines can be used for the analysis of the developmental control of multigenic and developmentally regulated human loci.


Subject(s)
Globins/genetics , Animals , Chromosomes, Artificial, Yeast , Gene Expression Regulation , Gene Transfer Techniques , Humans , Leukemia, Erythroblastic, Acute/genetics , Mice , RNA, Messenger/genetics , Transcriptional Activation
15.
Proc Natl Acad Sci U S A ; 89(3): 1143-7, 1992 Feb 01.
Article in English | MEDLINE | ID: mdl-1736298

ABSTRACT

The human beta-like globin gene locus contains embryonic, fetal, and adult globin genes that are regulated in a developmentally timed, as well as a tissue-specific, manner. The locus control region (LCR), located 5' of the globin genes, is characterized by four erythroid-specific nuclease-hypersensitive sites within native chromatin. These sites contain the active elements of the LCR. The LCR establishes an active chromatin conformation across the globin locus and enhances globin gene expression in transfected erythroleukemia cells and transgenic mice. We have used 5' DNase I hypersensitive site (HS) 4 as a model to define the minimum elements necessary for site formation. We have identified a 101-base-pair fragment within 5' HS4 that is the active site-forming element. DNase I footprint and gel-mobility shift assays have identified binding sites for transcription factors AP-1/NF-E2, Sp-1, and GATA-1 within the HS-forming element. We conclude that HS formation, the characteristic feature of the LCR in nuclear chromatin, requires interaction between erythroid-specific and ubiquitous nuclear proteins.


Subject(s)
Deoxyribonuclease I/metabolism , Gene Expression Regulation , Globins/genetics , Regulatory Sequences, Nucleic Acid , Animals , Base Sequence , Binding Sites , Chromatin/ultrastructure , DNA Mutational Analysis , DNA-Binding Proteins/metabolism , Humans , In Vitro Techniques , Mice , Mice, Transgenic , Molecular Sequence Data , Nuclear Proteins/metabolism , Transcription Factors/metabolism
16.
J Biol Chem ; 266(18): 11965-74, 1991 Jun 25.
Article in English | MEDLINE | ID: mdl-2050690

ABSTRACT

The promoter of the human gamma-globin gene confers tissue specificity as well as developmental stage specificity to gamma gene expression. Earlier work in our laboratory suggested that a fragment of the gamma-globin promoter between -300 and -137 base pairs upstream of the transcription start site contributed to the developmental specificity of the promoter. In this paper, we have mapped potential regulatory elements within this upstream region of the gamma promoter by a combination of in vitro DNA-protein binding assays and functional determinations of promoter strength in transient expression studies. Four sites between -300 and -130 bind proteins present in nuclear extracts of erythroid and non-erythroid cell lines. Mutation of these binding sites by internal base substitution determined that three of the four influence overall promoter strength in transient assays. We have focused on two protein binding sites, -246 to -212 and -195 to -170, that have been reported to bind erythroid-specific factors. The erythroid binding protein NF-E1 and a ubiquitous octamer protein footprint the -195 to -170 site. While internal mutation of this site did not significantly alter promoter strength, a point mutation at position -175 that is associated with hereditary persistence of fetal hemoglobin increased the activity of a promoter construct 20-fold in erythroid cells. A detailed mutational analysis of this site suggests that NF-E1 binding is necessary but not sufficient for activation of the promoter by the -175 mutation, and we propose that a second protein or co-activator is required. The nucleotides between -246 and -212 appear to bind a complex of at least three proteins, at the core of which is a protein binding to a string of dA:dT residues. This complex also appears to form on the 3' A gamma-globin enhancer, and homologous sites have been identified within the locus activating region of the beta-globin cluster, suggesting that this element may mediate long range interactions with distant regulatory elements.


Subject(s)
Globins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Base Sequence , Binding Sites , DNA Fingerprinting , DNA, Neoplasm/genetics , HeLa Cells , Humans , Leukemia, Erythroblastic, Acute/pathology , Methylation , Molecular Sequence Data , Mutation , Regulatory Sequences, Nucleic Acid , Transcription, Genetic
17.
Nucleic Acids Res ; 18(20): 6011-7, 1990 Oct 25.
Article in English | MEDLINE | ID: mdl-2235483

ABSTRACT

An erythroid specific, inducible enhancer associated with hypersensitive site II (HS II) plays a central role in the function of the human beta globin dominant control region. The HS II enhancer consists of tandem AP-1 binding sites and has been shown to bind members of the ubiquitous jun and fos families of proteins. The same sites are now shown to bind the erythroid specific protein, NF-E2. Inducibility of the HS II enhancer depends on NF-E2 binding, even in the presence of another hypersensitive site. Further, increased activity of the enhancer in induced K562 cells correlates with the presence of NF-E2, which appears to be present in a modified form. NF-E2 is distinct from some enhancer binding proteins in K562 nuclear extracts, in that it does not contain Fos or Fra-1 protein. Thus, binding by NF-E2 may be the mechanism, whereby tandem AP-1 binding sites confer erythroid specificity on the HS II enhancer.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Globins/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Base Sequence , Cell Line , Erythroid-Specific DNA-Binding Factors , Globins/biosynthesis , Humans , Leukemia, Erythroblastic, Acute , Molecular Sequence Data , Mutagenesis, Site-Directed , NF-E2 Transcription Factor , NF-E2 Transcription Factor, p45 Subunit , Oligonucleotide Probes , Plasmids , Promoter Regions, Genetic , Transfection
18.
Anal Biochem ; 154(2): 424-30, 1986 May 01.
Article in English | MEDLINE | ID: mdl-3728961

ABSTRACT

The techniques of ion exchange and gel filtration have been combined in a single chromatographic column which allows the simultaneous isolation of hemoglobins glycosylated at their beta-amino termini from other hemoglobin species as well as from molecules differing in size from the hemoglobins. This method is unique because it makes possible isolation of preparative quantities of glycosylated hemoglobins within approximately 15 min. The method works most efficiently with a dry weight-to-weight ratio of Biorex 70 to Sephadex G25 of 1.4 to 1.0. The technique was applied to the determination of the apparent first-order rate constant for the deglycosylation of the labile form of hemoglobin AIc.


Subject(s)
Glycated Hemoglobin/isolation & purification , Chromatography, Gel/methods , Chromatography, Ion Exchange/methods , Humans , Pilot Projects
20.
J Biol Chem ; 260(21): 11611-8, 1985 Sep 25.
Article in English | MEDLINE | ID: mdl-3930480

ABSTRACT

HbA1c is the most prevalent of the minor human hemoglobins. It is formed by the nonenzymatic addition of glucose to the alpha-amino group of the beta chain by an initial condensation reaction and a subsequent intermolecular Amadori rearrangement. We have developed a method of analysis which utilizes high performance liquid chromatography to follow the formation of HbA1c and greatly simplifies the determination of the kinetic parameters associated with this reaction. This has allowed us to study the effects of several Hb ligands, including the hydrogen ion, on the kinetics of this glycosylation reaction. Both the initial condensation reaction and the subsequent rearrangement are shown to exhibit acid catalysis, but the rate of the condensation step is limited by the extent of protonation of the alpha-amino group. The variation in kinetic parameters as a function of hydrogen ion concentration has allowed us to determine the probable reaction mechanism of HbA1c formation by comparison to previously reported model systems of Schiff base formation and Amadori rearrangement. The formation of pre-HbA1c from deoxy-Hb shows an increased forward rate when compared to oxy-Hb. The presence of physiologic concentrations of CO2 causes a proportional decrease in both k1 and k-1. 2,3-Diphosphoglycerate causes a significant increase in the keq of the formation reaction. The effects of CO and the substitution of L-glucose for D-glucose are not significant.


Subject(s)
Glycated Hemoglobin/biosynthesis , 2,3-Diphosphoglycerate , Carbon Dioxide/metabolism , Diphosphoglyceric Acids/pharmacology , Humans , Hydrogen-Ion Concentration , Kinetics , Ligands
SELECTION OF CITATIONS
SEARCH DETAIL
...