Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Exp Med ; 184(1): 61-9, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8691150

ABSTRACT

PU.1 is a tissue-specific transcription factor that is expressed in cells of the hematopoietic lineage including macrophages, granulocytes, and B lymphocytes. Bone marrow-derived macrophages transfected with an antisense PU.1 expression construct or treated with antisense oligonucleotides showed a decrease in proliferation compared with controls. In contrast, bone marrow macrophages transfected with a sense PU.1 expression construct displayed enhanced macrophage colony-stimulating factor (M-CSF)-dependent proliferation. Interestingly, there was no effect of sense or antisense constructs of PU.1 on the proliferation of the M-CSF-independent cell line, suggesting that the response was M-CSF dependent. This was further supported by the finding that macrophages transfected with a sense or an antisense PU.1 construct showed, respectively, an increased or a reduced level of surface expression of receptors for M-CSF. The enhancement of proliferation seems to be selective for PU.1, since transfections with several other members of the ets family, including ets-2 and fli-1, had no effect. Various mutants of PU.1 were also tested for their ability to affect macrophage proliferation. A reduction in macrophage proliferation was found when cells were transfected with a construct in which the DNA-binding domain of PU.1 was expressed. The PEST (proline-, glutamic acid-, serine-, and threonine-rich region) sequence of the PU.1 protein, which is an important domain for protein-protein interactions in B cells, was found to have no influence on PU.1-enhanced macrophage proliferation when an expression construct containing PU.1 minus the PEST domain was transfected into bone marrow-derived macrophages. In vivo, PU.1 is phosphorylated on several serine residues. The transfection of plasmids containing PU.1 with mutations at each of five serines showed that only positions 41 and 45 are critical for enhanced macrophage proliferation. We conclude that PU.1 is necessary for the M-CSF-dependent proliferation of macrophages. One of the proliferation-relevant targets of this transcription factor could be the M-CSF receptor.


Subject(s)
Macrophage Activation , Macrophages/cytology , Proto-Oncogene Proteins/physiology , Trans-Activators , Animals , Base Sequence , Bone Marrow Cells , DNA, Antisense/chemistry , DNA-Binding Proteins/physiology , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Inbred DBA , Molecular Sequence Data
2.
Science ; 259(5101): 1622-5, 1993 Mar 12.
Article in English | MEDLINE | ID: mdl-8456286

ABSTRACT

PU.1 recruits the binding of a second B cell-restricted nuclear factor, NF-EM5, to a DNA site in the immunoglobulin kappa 3' enhancer. DNA binding by NF-EM5 requires a protein-protein interaction with PU.1 and specific DNA contacts. Dephosphorylated PU.1 bound to DNA but did not interact with NF-EM5. Analysis of serine-to-alanine mutations in PU.1 indicated that serine 148 (Ser148) is required for protein-protein interaction. PU.1 produced in bacteria did not interact with NF-EM5. Phosphorylation of bacterially produced PU.1 by purified casein kinase II modified it to a form that interacted with NF-EM5 and that recruited NF-EM5 to bind to DNA. Phosphopeptide analysis of bacterially produced PU.1 suggested that Ser148 is phosphorylated by casein kinase II. This site is also phosphorylated in vivo. Expression of wild-type PU.1 increased expression of a reporter construct containing the PU.1 and NF-EM5 binding sites nearly sixfold, whereas the Ser148 mutant form only weakly activated transcription. These results demonstrate that phosphorylation of PU.1 at Ser148 is necessary for interaction with NF-EM5 and suggest that this phosphorylation can regulate transcriptional activity.


Subject(s)
DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , B-Lymphocytes/immunology , Base Sequence , Cell Line , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , Enhancer Elements, Genetic , Immunoglobulin kappa-Chains/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligonucleotide Probes , Phosphorylation , Plasmacytoma , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Retroviridae Proteins, Oncogenic , Transfection , Tumor Cells, Cultured
3.
Mol Cell Biol ; 16(1): 390-7, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8524320

ABSTRACT

The B-lymphocyte- and macrophage-specific transcription factor PU.1 is a member of the ets family of proteins. To understand how PU.1 functions as a transcription factor, we initiated a series of experiments to define its activation domain. Using deletion analysis, we showed that the activation domain of PU.1 is located in the amino-terminal half of the protein. Within this region, we identified three acidic subdomains and one glutamine-rich subdomain. The deletion of any of these subdomains resulted in a significant loss in the ability of PU.1 to transactivate in cotransfection studies. Amino acid substitution analysis showed that the activation of transcription by PU.1 requires acidic residues between amino acids 7 and 74 and a group of glutamine residues between amino acids 75 and 84. These data show that PU.1 contains two types of known activation domains and that both are required for maximal transactivation.


Subject(s)
DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Acids/chemistry , Amino Acid Sequence , Binding Sites/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Glutamine/chemistry , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Mapping , Retroviridae Proteins, Oncogenic , Sequence Deletion , Transcription Factors/chemistry , Transcription Factors/genetics , Transfection
4.
Mol Cell Biol ; 12(1): 368-78, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1729611

ABSTRACT

PU.1 is a B-cell- and macrophage-specific transcription factor. By an electrophoretic mobility shift assay and dimethyl sulfate methylation interference assays, we show that PU.1 binds to DNA sequences within the immunoglobulin kappa 3' enhancer (kappa E3'). Binding of PU.1 to the kappa E3' enhancer assists the binding of a second tissue-restricted factor, NF-EM5, to an adjacent site. Binding of NF-EM5 to kappa E3' DNA sequences requires protein-protein interaction with PU.1 as well as specific protein-DNA interactions. This is the first known instance of PU.1 interacting with another cellular protein. NF-EM5 does not cofractionate with PU.1, suggesting that it is a distinct protein and is not a posttranslational modification of PU.1. UV-crosslinking studies and elution from sodium dodecyl sulfate-polyacrylamide gels indicate that NF-EM5 is a protein of approximately 46 kDa. Site-directed mutagenesis studies of the PU.1- and EM5-binding sites indicate that these sites play important roles in kappa E3' enhancer activity. By using a series of PU.1 deletion constructs, we have identified a region in PU.1 that is necessary for interaction with NF-EM5. This segment encompasses a 43-amino-acid region with PEST sequence homology, i.e., one that is rich in proline (P), glutamic acid (E), serine (S), and threonine (T).


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Immunoglobulin kappa-Chains/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Base Sequence , Binding Sites , Cell Line , DNA , Electrophoresis, Polyacrylamide Gel , Molecular Sequence Data , Mutagenesis, Site-Directed , Organ Specificity/genetics , Retroviridae Proteins, Oncogenic
5.
Mol Cell Biol ; 17(9): 4885-94, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9271368

ABSTRACT

The activities of ETS transcription factors are modulated by posttranscriptional modifications and cooperation with other proteins. Another factor which could alter the regulation of genes by ETS transcription factors is DNA methylation of their cognate binding sites. The optimal activity of the keratin 18 (K18) gene is dependent upon an ETS binding site within an enhancer region located in the first intron. The methylation of the ETS binding site was correlated with the repression of the K18 gene in normal human tissues and in K18 transgenic mouse tissues. Neither recombinant ETS2 nor endogenous spleen ETS binding activities bound the methylated site effectively. Increased expression of the K18 gene in spleens of transgenic mice by use of an alternative, cryptic promoter 700 bp upstream of the enhancer resulted in modestly decreased methylation of the K18 ETS site and increased RNA expression. Expression in transgenic mice of a mutant K18 gene, which was still capable of activation by ETS factors but was no longer a substrate for DNA methylation of the ETS site, was fivefold higher in spleen and heart. However, expression in other organs such as liver and intestine was similar to that of the wild-type gene. This result suggests that DNA methylation of the K18 ETS site may be functionally important in the tissue-specific repression of the K18 gene. Epigenetic modification of the binding sites for some ETS transcription factors may result in a refractory transcriptional response even in the presence of necessary trans-acting activities.


Subject(s)
DNA Methylation , Introns , Keratins/genetics , Repressor Proteins , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Chromosome Mapping , DNA-Binding Proteins/metabolism , Humans , Keratins/metabolism , Mice , Mice, Transgenic , Molecular Sequence Data , Ribonucleases/metabolism , Transgenes
6.
Mol Cell Biol ; 13(12): 7393-8, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8246959

ABSTRACT

EWS/FLI-1 is a chimeric protein formed by a tumor-specific 11;22 translocation found in both Ewing's sarcoma and primitive neuroectodermal tumor of childhood. EWS/FLI-1 has been shown to be a potent transforming gene, suggesting that it plays an important role in the genesis of these human tumors. We now demonstrate that EWS/FLI-1 has the characteristics of an aberrant transcription factor. Subcellular fractionation experiments localized the EWS/FLI-1 protein to the nucleus of primitive neuroectodermal tumor cells. EWS/FLI-1 specifically bound in vitro an ets-2 consensus sequence similarly to normal FLI-1. When coupled to a GAL4 DNA-binding domain, the amino-terminal EWS/FLI-1 region was a much more potent transcriptional activator than the corresponding amino-terminal domain of FLI-1. Finally, EWS/FLI-1 efficiently transformed NIH 3T3 cells, but FLI-1 did not. These data suggest that EWS/FLI-1, functioning as a transcription factor, leads to a phenotype dramatically different from that of cells expressing FLI-1. EWS/FLI-1 could disrupt normal growth and differentiation either by more efficiently activating FLI-1 target genes or by inappropriately modulating genes normally not responsive to FLI-1.


Subject(s)
Oncogenes , Proto-Oncogene Proteins , Sarcoma, Ewing/genetics , Trans-Activators/genetics , 3T3 Cells , Animals , Base Sequence , Binding Sites , Cell Line , Cell Nucleus/metabolism , Cell Transformation, Neoplastic , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 22 , Cloning, Molecular , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Mice , Molecular Sequence Data , Proto-Oncogene Protein c-fli-1 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sarcoma, Ewing/metabolism , Trans-Activators/metabolism , Translocation, Genetic
7.
Biochim Biophys Acta ; 1172(1-2): 155-8, 1993 Feb 20.
Article in English | MEDLINE | ID: mdl-8439553

ABSTRACT

The v-ets oncogene family shares a conserved motif, termed the ETS-domain, that mediates sequence-specific DNA binding. This motif is unique among transcription factor families. Using partially degenerate oligonucleotides to highly conserved amino acids in this motif as primers for the polymerase chain reaction, a novel ETS-domain cDNA fragment was generated. This fragment was subsequently used to clone both mouse and human full length cDNAs for this gene. The amino acid sequence of the longest open reading frame showed that this gene was homologous to the mouse FLI-I gene, an ETS family gene activated by Friend erythroleukemia virus insertion. The gene is normally expressed only in hematopoietic cells. The gene was localized to chromosome 11q24, a region of aberrations in Ewing's sarcoma and neuroepithelioma. In the neuroepithelioma cell line TC-32 the FLI-1 transcript is present but has an aberrant structure, indicating that it may be rearranged in neuroepithelioma.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 11 , Neuroectodermal Tumors, Primitive, Peripheral/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , Base Sequence , Chromosome Banding , Chromosome Mapping , Cloning, Molecular , HeLa Cells , Humans , Mice , Molecular Sequence Data , Multigene Family , Oncogenes , Open Reading Frames , Protein-Tyrosine Kinases/genetics , Retroviridae Proteins, Oncogenic/genetics
8.
Curr Top Microbiol Immunol ; 211: 159-64, 1996.
Article in English | MEDLINE | ID: mdl-8585946

ABSTRACT

Transcriptional regulators control much of hematopoiesis. One such transcriptional regulator is the myeloid zinc finger gene MZF-1. MZF-1 has been localized to the telomere of chromosome 19q, where a large number of related zinc finger genes reside. It has been found to be essential in granulopoiesis. It is a bi-functional transcriptional regulator, repressing transcription in non-hematopoietic cells, and activating transcription in cells of hematopoietic origins. Its consensus DNA binding site has been isolated, and sites in several promoters of myeloid-specific genes, such as CD34, lactoferrin, and myeloperoxidase, have been defined. In co-transfection experiments MZF-1 has been found to regulate transcription from the CD34 promoter.


Subject(s)
DNA-Binding Proteins/physiology , Hematopoiesis/genetics , Transcription Factors/physiology , Transcription, Genetic , Zinc Fingers , Base Sequence , Chromosomes, Human, Pair 19 , DNA , DNA-Binding Proteins/genetics , Gene Expression Regulation , Humans , Kruppel-Like Transcription Factors , Molecular Sequence Data , Transcription Factors/genetics , Zinc Fingers/genetics
9.
DNA Cell Biol ; 18(12): 875-84, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10619599

ABSTRACT

Knockout studies have shown that PU.1 is required for the normal development of many blood cell lineages, yet overexpression of this transcription factor in erythroid cells can lead to erythroleukemia. Thus, how the tissue-specific expression of PU.1 is regulated is important to our understanding of hematopoiesis. In this study, we showed that B and macrophage cell lines expressing PU.1 contained DNase I-hypersensitive sites in intron 1 and were hypomethylated at three MspI sites flanking exon 1. Results from studies using several T-cell lines suggested that the pattern of methylation changed as these cells matured. A pre-T cell line that expresses PU.1 contained DNase I-hypersensitive sites in intron 1 and was also hypomethylated at both MspI sites. Other immature T-cell lines had methylated at least one of the MspI sites and displayed no hypersensitive sites. Mature T-cell lines had a methylation pattern more similar to that of fibroblasts. Treatment of an immature T-cell line with 5-azacytidine resulted in the expression of PU.1 transcripts. These data suggest that the tissue-specific expression of PU.1 is controlled by chromatin structure and DNA methylation and that this may be a mechanism used to shut off PU.1 expression in specific cell lineages during hematopoiesis.


Subject(s)
Chromatin/metabolism , DNA Methylation , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , B-Lymphocytes/metabolism , Cell Line , Chromatin/chemistry , CpG Islands , Deoxyribonuclease I/metabolism , Humans , Introns , Mice , Promoter Regions, Genetic , Proto-Oncogene Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Trans-Activators/biosynthesis
10.
Int J Hematol ; 59(4): 257-65, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8086619

ABSTRACT

V-ets was found as an oncogenic sequence in the E26 avian retrovirus, which produced leukemias in chickens. There are a large number of mammalian homologues of v-ets, all sharing a common sequence called the ETS domain. These mammalian homologues have been found to be transcriptional activators, and the ETS domain is the DNA binding portion of the gene. This family has been found to be important in regulating embryonic development and the response to growth stimuli. In addition, PU.1 and FLI-1 are dysregulated by Friend leukemia virus insertion in mice resulting in erythroleukemia. Significantly, the DNA binding portion of FLI-1 is the 3' part of an oncogenic fusion transcript (termed EWS-FLI) in human Ewing's sarcoma and neuroepithelioma.


Subject(s)
Neoplasms/genetics , Oncogenes , Retroviridae Proteins, Oncogenic/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Division , Humans , Molecular Sequence Data , Neoplasms/pathology , Neuroectodermal Tumors, Primitive, Peripheral/genetics , Retroviridae Proteins, Oncogenic/chemistry , Sarcoma, Ewing/genetics
11.
Leuk Lymphoma ; 15(5-6): 439-44, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7874001

ABSTRACT

The Forkhead gene (FKH) regulates morphogenesis in Drosophila. It is the prototype of a new family of transcriptional activators. We used the polymerase chain reaction (PCR) to analyze the expression pattern of this new transcriptional regulatory gene family in primitive hematopoeitic progenitors. Partially degenerate oligonucleotides to two conserved amino acid sequences of this family were used to prime a PCR amplification of cDNA synthesized from CD34+/HLA-DR- hematopoietic cells. Known and novel FKH genes were found to be expressed in these cells.


Subject(s)
Hematopoietic Stem Cells/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Base Sequence , Forkhead Transcription Factors , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
13.
Cell Immunol ; 178(1): 53-61, 1997 May 25.
Article in English | MEDLINE | ID: mdl-9184698

ABSTRACT

With recent studies suggesting a key role for professional antigen presenting cells in the induction of major histocompatibility class I cellular immune responses, we initiated studies on the regulation of Tap-1 and Tap-2 gene expression in macrophages. Stimulation of the human macrophage cell line THP-1 with interferon-gamma (IFN-gamma) resulted in maximal induction of both Tap-1 and Tap-2 mRNA within 24 hr. Nuclear run-on analyses showed that the increased expression of Tap-1 and Tap-2 was controlled at the level of transcription. Half-life studies demonstrated that mRNAs for both genes became destabilized after stimulation of THP-1 cells with IFN-gamma for 24 hr, suggesting that a posttranscriptional mechanism down-regulates TAP gene expression following activation. Treatment of cells with both IFN-gamma and lipopolysaccharide (LPS) altered the kinetics and amount of Tap-1 mRNA and protein expression, compared to those with stimulation with IFN-gamma alone. These data suggest that LPS enhances the ability of macrophages stimulated with IFN-gamma to initiate a cellular immune response by altering the kinetics of TAP gene expression.


Subject(s)
ATP-Binding Cassette Transporters/biosynthesis , Gene Expression Regulation, Leukemic/drug effects , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP Binding Cassette Transporter, Subfamily B, Member 3 , ATP-Binding Cassette Transporters/genetics , Drug Synergism , Half-Life , Humans , Leukemia, Monocytic, Acute/pathology , Macrophages/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Transcription, Genetic , Tumor Cells, Cultured
14.
J Immunol ; 167(9): 5160-6, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11673528

ABSTRACT

Altering chromatin structure by blocking histone deacetylase activity with specific inhibitors such as trichostatin A can result in an up-regulation of gene expression. In this report, however, we show that expression of the ETS domain transcription factor PU.1 is down-regulated in cells following the addition of trichostatin A. The loss of PU.1 is seen at both the mRNA and protein levels in multiple cell lines and is reversible following removal of the drug. More importantly, we show that the loss of PU.1 results in a loss of PU.1 target gene expression, including CD11b, c-fms, Toll-like receptor 4, and scavenger receptor. Chromatin immunoprecipitation analysis of cells treated with trichostatin A showed a significant increase in the acetylation of histone H4, but not histone H3, across approximately 650 bp of the PU.1 promoter region. Our data suggest that the consequences of using drugs that inhibit histone deacetylase activity may be a loss of blood cell development and/or function due to a block in PU.1 gene expression.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Acetylation , Animals , Cell Line , Chromosome Mapping , Histones/metabolism , Hydroxamic Acids/pharmacology , Macrophage-1 Antigen/analysis , Mice , Promoter Regions, Genetic , RNA, Messenger/analysis
15.
J Immunol ; 165(6): 3190-7, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10975834

ABSTRACT

Proper regulation of the Tap-1 gene is critical for the initiation and continuation of a cellular immune response. Analysis of the Tap-1/low molecular mass polypeptide 2 bidirectional promoter showed that the IFN-gamma activation site element is critical for the rapid induction of the promoter by IFN-gamma following transfection into the human macrophage cell line THP-1. Furthermore, activation of STAT1 binding to this site was important for the synergistic response seen following the stimulation with both IFN-gamma and LPS. Mutation of an IFN-stimulated regulatory element that binds IFN regulatory factor 1 appeared to enhance the response to IFN-gamma and LPS. These data show that STAT1 is necessary for the activation of Tap-1 gene expression in APCs and initiation of cellular immune responses. Furthermore, our data suggest that bacterial products such as LPS may enhance cellular immune responses through augmenting the ability of STAT1 to regulate IFN-gamma-inducible genes.


Subject(s)
ATP-Binding Cassette Transporters/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation/immunology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Signal Transduction/genetics , Trans-Activators/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , Cell Line , DNA-Binding Proteins/metabolism , Drug Synergism , Extracellular Matrix Proteins/genetics , Humans , Interferon Regulatory Factor-1 , Interferon Regulatory Factors , Nerve Tissue Proteins/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic/immunology , Protein Binding/genetics , Protein Binding/immunology , Repressor Proteins/metabolism , STAT1 Transcription Factor , Signal Transduction/immunology , Trans-Activators/metabolism , Transfection/immunology
16.
Eur J Immunol ; 19(6): 1103-9, 1989 Jun.
Article in English | MEDLINE | ID: mdl-2502420

ABSTRACT

The purpose of this study was to obtain additional information on the mechanism by which interferon-gamma (IFN-gamma) is able to regulate gene expression in macrophages. The expression of the genes for class II histocompatibility I-A beta, tumor necrosis factor (TNF) and complement component C3 was assayed after treating bone marrow macrophages with IFN-gamma. Each gene displayed a characteristic pattern of regulation. First, the increase in the level of RNA for each gene followed different kinetics. The level of TNF RNA increased within 15 min after IFN-gamma treatment and reached a plateau after 4 h. In contrast, there was a lag of about 4 h before the level of I-A beta RNA began to rise and a plateau was not reached until 48 h after the IFN-gamma treatment began. C3 gene expression followed an intermediate time course between that for TNF and I-A beta. Second, the expression of I-A beta was inhibited when cells were treated with both IFN-gamma and cycloheximide, while the expression of TNF and C3 was not. Interestingly, the sensitivity to cycloheximide only lasted 30 min following the addition of IFN-gamma, after which cycloheximide had no effect on the expression of I-A beta. Third, lipopolysaccharide abolished the IFN-gamma-induced expression of I-A beta, but enhanced the expression of TNF. Based on these observations, we conclude that IFN-gamma must activate multiple pathways to regulate gene expression in macrophages.


Subject(s)
Complement C3/genetics , Histocompatibility Antigens Class II/genetics , Interferon-gamma/pharmacology , Macrophages/physiology , Tumor Necrosis Factor-alpha/genetics , Animals , Bone Marrow Cells , Cycloheximide/pharmacology , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred DBA , Recombinant Proteins , Time Factors , Transcription, Genetic/drug effects
17.
J Biol Chem ; 268(8): 5769-73, 1993 Mar 15.
Article in English | MEDLINE | ID: mdl-8449942

ABSTRACT

The recently cloned fli-1 gene is a member of the ets oncogene family that is preferentially expressed in hematopoietic cells. It is a target of dysregulation by Friend leukemia virus insertion and translocation in Ewing's sarcoma and neuroepithelioma. In this report, we have studied the function and regulation of both murine and human fli-1. Analysis of the human and mouse fli-1 proteins showed that fli-1 binds to specific DNA sequences highly related to m-ets-2 binding sites. Methylation protection experiments showed that fli-1 and m-ets-2 contacted the same nucleotides in two different binding sites. The fli-1 protein was shown to be a transcriptional activator in co-transfection studies. Stimulation of murine bone marrow macrophages by mediators of inflammation, such as lipopolysaccharide, phorbol 12-myristate 13-acetate, interleukin-1, and interferon-gamma resulted in the reduced expression of fli-1 mRNA. fli-1 was only expressed in a defined subset of human erythroleukemia cell lines.


Subject(s)
DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins , Retroviridae Proteins, Oncogenic/genetics , Trans-Activators/metabolism , Animals , Base Sequence , Binding Sites , Cell Line , DNA/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Humans , Macrophages/metabolism , Mice , Molecular Sequence Data , Proto-Oncogene Protein c-fli-1 , RNA, Messenger/metabolism , Trans-Activators/genetics , Transcription, Genetic , Transfection
18.
Genomics ; 20(2): 291-4, 1994 Mar 15.
Article in English | MEDLINE | ID: mdl-8020980

ABSTRACT

The v-ets oncogene family shares a conserved peptide motif called the ETS domain that mediates sequence-specific DNA binding. This motif is unique among transcription factor families. Using partially degenerate oligonucleotides from conserved regions of the ETS domain and the polymerase chain reaction, we isolated a new member of the v-ets family designated PE-1 from HL60 cells. PE-1 was expressed as an approximately 7.5-kb transcript in most cell lines tested. In the hairy cell leukemia line Eskol, there was an additional 1.8-kb transcript observed. PE-1 was the most common ETS domain gene found in CD34+HLA-DR- hematopoietic progenitors. PE-1 was localized to human chromosome 1q21-q23 using both in situ chromosomal hybridization and human-hamster hybrids.


Subject(s)
Chromosomes, Human, Pair 1 , DNA-Binding Proteins/genetics , Multigene Family , Oncogene Proteins/genetics , Oncogenes , Retroviridae Proteins, Oncogenic/genetics , Amino Acid Sequence , Blotting, Southern , Cell Line , Chromosome Mapping , DNA-Binding Proteins/chemistry , HeLa Cells , Humans , Molecular Sequence Data , Oncogene Proteins/chemistry , Polymerase Chain Reaction , Proto-Oncogene Proteins c-ets , Sequence Homology, Amino Acid , Tumor Cells, Cultured
19.
Blood ; 81(11): 2854-9, 1993 Jun 01.
Article in English | MEDLINE | ID: mdl-8499623

ABSTRACT

The forkhead gene (FKH) regulates morphogenesis in Drosophila. It is the prototype of a new family of transcriptional activators. Partially degenerate oligonucleotides to two conserved amino acid sequences of this family were used to prime a polymerase chain reaction (PCR) amplification of HEL cell cDNA. Two unique clones, designated H3 and H8, were isolated that contained homologies to FKH. A third novel clone, 5-3, was isolated by low stringency screening of a chronic myelogenous leukemia cDNA library using H8 as a probe. H3 and 5-3 are preferentially expressed in restricted hematopoietic lineages, while the expression of H8 was ubiquitous. Southern analysis showed that FKH 5-3 is conserved through yeast, which is rare among tissue-specific transcription factors. The H3 and 5-3 clones provide evidence that FKH family members are present in a tissue-restricted manner in humans.


Subject(s)
DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Base Sequence , Blotting, Southern , Cloning, Molecular , DNA-Binding Proteins/chemistry , Forkhead Transcription Factors , Gene Expression , Genes , Humans , Molecular Sequence Data , Morphogenesis , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides/chemistry , Polymerase Chain Reaction , RNA, Messenger/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/chemistry
20.
Cell ; 61(1): 113-24, 1990 Apr 06.
Article in English | MEDLINE | ID: mdl-2180582

ABSTRACT

We have isolated a cDNA clone, PU.1, that codes for a new tissue-specific DNA binding protein. Analysis of the binding site by methylation interference and DNAase 1 protection revealed that the PU.1 protein recognized a purine-rich sequence, 5'-GAGGAA-3' (PU box). The PU.1 protein was shown to be a transcriptional activator that is expressed in macrophages and B cells. cDNA constructions used to generate proteins lacking portions of either the amino- or carboxy-terminal ends of the PU.1 protein placed the DNA binding domain in the highly basic carboxy-terminal domain of the protein. The amino acid sequence in the binding domain of PU.1 has considerable identity with proteins belonging to the ets oncogene family.


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/genetics , Genes , Macrophages/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Nucleus/metabolism , Cloning, Molecular , DNA/genetics , DNA/isolation & purification , Gene Expression Regulation , Gene Library , Immunoblotting , Methylation , Mice , Molecular Sequence Data , Nucleic Acid Hybridization , Oligonucleotide Probes , Protein Biosynthesis , Protein Conformation , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-ets , Restriction Mapping , Retroviridae Proteins, Oncogenic , Sequence Homology, Nucleic Acid , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL