Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Syst Evol Microbiol ; 70(1): 83-88, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31596191

ABSTRACT

Auritidibacter ignavus is a Gram-stain-positive bacillus derived from otorrhea. Four strains derived from ear discharges in Canada and Switzerland, with features consistent with but distinguishable from Auritidibacter ignavus IMMIB L-1656T (accession number FN554542) by 16S rRNA gene sequencing (97.5 % similarity), were thought to represent a novel species of the genus Auritidibacter. Auritidibacter ignavus DSM 45359T (=IMMIB L-1656T) was acquired to compare with Canadian and Swiss strains by whole-genome sequencing (WGS). Unexpectedly, those isolates were observed to be consistent with A. ignavus DSM 45359T by WGS (ANIb scores >98 %), MALDI-TOF (Bruker), cellular fatty acid analysis and biochemically (some differences were observed). A nearly full 16S rRNA gene sequence could not be readily prepared from A. ignavus DSM 45359T, even after multiple attempts. A 16S rRNA gene chimeric consensus sequence created from the genome assembly of A. ignavus DSM 45359T had only 97.5 % similarity to that of A. ignavus IMMIB L-1656T, implying that 16S rRNA sequence accession number FN554542 could not be replicated. We concluded that our isolates of members of the genus Auritidibacter were consistent with A. ignavus DSM 45359T, did not represent a novel species, and that the sequence corresponding to FN554542 was not reproducible. By WGS, A. ignavus DSM 45359T had genome of 2.53×106 bp with a DNA G+C content of 59.34%, while genomes of Canadian and Swiss isolates ranged from 2.47 to 2.59×106 bp with DNA G+C contents of 59.3-59.52 %. A. ignavus NML 100628 (=NCTC 14178=LMG 30897) did not demonstrate a rodcoccus cycle. Emendation of Auritidibacter ignavus was proposed based on these results.


Subject(s)
Micrococcaceae/classification , Phylogeny , Aged , Bacterial Typing Techniques , Base Composition , Canada , DNA, Bacterial/genetics , Ear/microbiology , Fatty Acids/chemistry , Female , Humans , Male , Middle Aged , Nucleic Acid Hybridization , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Switzerland
2.
J Cell Biol ; 107(3): 865-76, 1988 Sep.
Article in English | MEDLINE | ID: mdl-3047152

ABSTRACT

The soluble cytoplasmic protein pyruvate kinase (PK) has been expressed at the cell surface in a membrane-anchored form (APK). The hybrid protein contains the NH2-terminal signal/anchor domain of a class II integral membrane protein (hemagglutinin/neuraminidase, of the paramyxovirus SV5) fused to the PK NH2 terminus. APK contains a cryptic site that is used for N-linked glycosylation but elimination of this site by site-specific mutagenesis does not prevent cell surface localization. Truncated forms of the APK molecule, with up to 80% of the PK region of APK removed, can also be expressed at the cell surface. These data suggest that neither the complete PK molecule nor its glycosylation are necessary for intracellular transport of PK to the cell surface, and it is possible that specific signals may not be needed in the ectodomain of this hybrid protein to specify cell surface localization.


Subject(s)
Cell Membrane/enzymology , Cytoplasm/enzymology , Pyruvate Kinase/metabolism , Animals , Fluorescent Antibody Technique , Gene Expression Regulation , Glycosylation , HN Protein , Plasmids , Precipitin Tests , Pyruvate Kinase/biosynthesis , Pyruvate Kinase/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
3.
Oncogene ; 26(1): 91-101, 2007 Jan 04.
Article in English | MEDLINE | ID: mdl-16799637

ABSTRACT

The t(8;21) chromosomal translocation that generates the fusion oncoprotein RUNX1-ETO predominates in leukemia patients of the French-American-British (FAB) class M2 subtype. The oncoprotein has the capacity to promote expansion of hematopoietic stem/progenitor cells and induces leukemia in association with other genetic alterations. Here, we show that RUNX1-ETO undergoes degradation in response to treatment with histone deacetylase inhibitors, one of which, depsipeptide (DEP), is currently undergoing phase II clinical testing in a variety of malignancies. These compounds induce turnover of RUNX1-ETO without affecting the stability of RUNX1-ETO partner proteins. In addition, RUNX1-ETO physically interacts with heat shock protein 90 (HSP90). DEP treatment interrupts the association of RUNX1-ETO with HSP90 and induces proteasomal degradation of RUNX1-ETO. DEP and the HSP90 antagonist 17-allylamino-geldanamycin (17-AAG) both triggered RUNX1-ETO degradation, but without any additive or cooperative effects. These findings may stimulate the development of more rational and effective approaches for treating t(8;21) patients using histone deacetylase inhibitors or HSP90 inhibitors.


Subject(s)
Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Proto-Oncogene Proteins/genetics , Transcription Factors/genetics , Translocation, Genetic , Cell Line, Tumor , Humans , Hydrolysis , Immunoprecipitation , RUNX1 Translocation Partner 1 Protein
4.
Oncogene ; 25(42): 5777-86, 2006 Sep 21.
Article in English | MEDLINE | ID: mdl-16652147

ABSTRACT

RUNX1 (AML1) is a gene that is frequently disrupted by chromosomal translocations in acute leukemia. Like its Drosophila homolog Runt, RUNX1 both activates and represses transcription. Both Runt and RUNX1 are required for gene silencing during development and a central domain of RUNX1, termed repression domain 2 (RD2), was defined as being required for transcriptional repression and for the silencing of CD4 during T-cell maturation in thymic organ cultures. Although transcriptional co-repressors are known to contact other repression domains in RUNX1, the factors that bind to RD2 had not been defined. Therefore, we tested whether RD2 contacts histone-modifying enzymes that may mediate both repression and gene silencing. We found that RD2 contacts SUV39H1, a histone methyltransferase, via two motifs and that endogenous Suv39h1 associates with a Runx1-regulated repression element in murine erythroleukemia cells. In addition, one of these SUV39H1-binding motifs is also sufficient for binding to histone deacetylases 1 and 3, and both of these domains are required for full RUNX1-mediated transcriptional repression. The association between RUNX1, histone deacetylases and SUV39H1 provides a molecular mechanism for repression and possibly gene silencing mediated by RUNX1.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Histone Deacetylases/metabolism , Methyltransferases/metabolism , Repressor Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Humans , Jurkat Cells , Transfection
5.
Mol Cell Biol ; 13(6): 3384-91, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8497257

ABSTRACT

Studies of naturally occurring mutations of the RB1 tumor suppressor gene have indicated that the E1A/T antigen-binding domain is important for pRb function. Mutations engineered within the C-terminal 135 amino acids of pRb also abrogate its growth-suppressive function during the G1 interval of the cell cycle. Both the pRb E1A/T antigen-binding domain and the C-terminal domain are required for interaction with the E2F transcription factor. A series of mutated pRb proteins has been used to define the C-terminal sequences which determine E2F binding, adenovirus E2 promoter inhibition, and negative growth control. Deletion of the C terminus to residue 870 allowed full pRb function, while further deletion to residue 841 inactivated pRb in each assay. Amino acid sequences immediately C-terminal to the E1A/T antigen-binding domain were absolutely required for pRb activity. Mutations which prevented pRb from interacting with E2F also eliminated pRb-mediated E2 promoter repression and inactivated the ability of pRb to suppress cell growth.


Subject(s)
Adenovirus E2 Proteins/genetics , Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Genes, Retinoblastoma , Promoter Regions, Genetic , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Adenovirus E1A Proteins/genetics , Binding Sites , Cell Cycle/physiology , Cell Division , E2F Transcription Factors , Female , G1 Phase , Humans , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transfection , Tumor Cells, Cultured , Uterine Cervical Neoplasms
6.
Mol Cell Biol ; 15(4): 1974-82, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7891692

ABSTRACT

The AML-1/CBF beta transcription factor complex is targeted by both the t(8;21) and the inv(16) chromosomal alterations, which are frequently observed in acute myelogenous leukemia. AML-1 is a site-specific DNA-binding protein that recognizes the enhancer core motif TGTGGT. The t(8;21) translocation fuses the first 177 amino acids of AML-1 to MTG8 (also known as ETO), generating a chimeric protein that retains the DNA-binding domain of AML-1. Analysis of endogenous AML-1 DNA-binding complexes suggested the presence of at least two AML-1 isoforms. Accordingly, we screened a human B-cell cDNA library and isolated a larger, potentially alternatively spliced, form of AML1, termed AML1B. AML-1B is a protein of 53 kDa that binds to a consensus AML-1-binding site and complexes with CBF beta. Subcellular fractionation experiments demonstrated that both AML-1 and AML-1/ETO are efficiently extracted from the nucleus under ionic conditions but that AML-1B is localized to a salt-resistant nuclear compartment. Analysis of the transcriptional activities of AML-1, AML-1B, and AML-1/ETO demonstrated that only AML-1B activates transcription from the T-cell receptor beta enhancer. Mixing experiments indicated that AML-1/ETO can efficiently block AML-1B-dependent transcriptional activation, suggesting that the t(8;21) translocation creates a dominant interfering protein.


Subject(s)
DNA-Binding Proteins/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Proto-Oncogene Proteins , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Activation , Alternative Splicing , Amino Acid Sequence , Base Sequence , Cell Compartmentation , Cell Nucleus/chemistry , Cloning, Molecular , Consensus Sequence , Core Binding Factor Alpha 2 Subunit , Core Binding Factors , DNA, Complementary/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Leukemia, Myeloid, Acute/genetics , Molecular Sequence Data , Neoplasm Proteins/isolation & purification , Neoplasm Proteins/metabolism , Protein Binding , RUNX1 Translocation Partner 1 Protein , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism
7.
Mol Cell Biol ; 10(5): 1989-2001, 1990 May.
Article in English | MEDLINE | ID: mdl-2183015

ABSTRACT

The role of N-linked glycosylation in protein maturation and transport has been studied by using the simian virus 5 hemagglutinin-neuraminidase (HN) protein, a model class II integral membrane glycoprotein. The sites of N-linked glycosylation on HN were identified by eliminating each of the potential sites for N-linked glycosylation by oligonucleotide-directed mutagenesis on a cDNA clone. Expression of the mutant HN proteins in eucaryotic cells indicated that four sites are used in the HN glycoprotein for the addition of N-linked oligosaccharide chains. These functional glycosylation sites were systematically eliminated in various combinations from HN to form a panel of mutants in which the roles of individual carbohydrate chains and groups of carbohydrate chains could be analyzed. Alterations in the normal glycosylation pattern resulted in the impairment of HN protein folding and assembly which, in turn, affected the intracellular transport of HN. The severity of the consequences on HN maturation depended on both the number of deleted carbohydrate sites and their position in the HN molecule. Analysis of the reactivity pattern of HN conformation-specific monoclonal antibodies with the mutant HN proteins indicated that one specific carbohydrate chain plays a major role in promoting the correct folding of HN. Another carbohydrate chain, which is not essential for the initial folding of HN was found to play a role in preventing the aggregation of HN oligomers. The HN molecules which were misfolded, owing to their altered glycosylation pattern, were retained in the endoplasmic reticulum. Double-label immunofluorescence experiments indicate that misfolded HN and folded HN are segregated in the same cell. Misfolded HN forms disulfide-linked aggregates and is stably associated with the resident endoplasmic reticulum protein, GRP78-BiP, whereas wild-type HN forms a specific and transient complex with GRP78-BiP during its folding process.


Subject(s)
HN Protein/ultrastructure , HSP70 Heat-Shock Proteins , Membrane Glycoproteins/ultrastructure , Antibodies, Monoclonal , Biological Transport , Cell Compartmentation , DNA Mutational Analysis , Endoplasmic Reticulum/metabolism , Fluorescent Antibody Technique , Glycosylation , HN Protein/genetics , HN Protein/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Paramyxoviridae/genetics , Paramyxoviridae/metabolism , Protein Binding , Protein Conformation , Protein Processing, Post-Translational , Structure-Activity Relationship , Temperature , Tunicamycin/pharmacology
8.
Mol Cell Biol ; 18(6): 3604-11, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9584201

ABSTRACT

Chromosomal translocations in acute leukemia that affect the AML-1/CBFbeta transcription factor complex create dominant inhibitory proteins. However, the mechanisms by which these proteins act remain obscure. Here we demonstrate that the multidrug resistance 1 (MDR-1) promoter is a target for AML/ETO transcriptional repression. This repression is of basal, not activated, expression from the MDR-1 promoter and thus represents a new mechanism for AML/ETO function. We have defined two domains in AML/ETO that are required for repression of basal transcription from the MDR-1 promoter: a hydrophobic heptad repeat (HHR) motif and a conserved zinc finger (ZnF) domain termed the MYND domain. The HHR mediates formation of AML/ETO homodimers and AML/ETO-ETO heterodimers. Single serine substitutions at conserved cysteine residues within the predicted ZnFs also abrogate transcriptional repression. Finally, we observe that AML/ETO can also inhibit Ets-1 activation of the MDR-1 promoter, indicating that AML/ETO can disrupt both basal and Ets-1-dependent transcription. The fortuitous inhibition of MDR-1 expression in t(8;21)-containing leukemias may contribute to the favorable response of these patients to chemotherapeutic drugs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Oncogene Proteins, Fusion , Promoter Regions, Genetic , Repressor Proteins/metabolism , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/metabolism , Dimerization , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Point Mutation , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , RUNX1 Translocation Partner 1 Protein , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism , Translocation, Genetic , Tumor Cells, Cultured , Zinc Fingers/genetics
9.
Mol Cell Biol ; 13(10): 6336-45, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8413232

ABSTRACT

The AML1 gene on chromosome 21 is disrupted in the (8;21)(q22;q22) translocation associated with acute myelogenous leukemia and encodes a protein with a central 118-amino-acid domain with 69% homology to the Drosophila pair-rule gene, runt. We demonstrate that AML-1 is a DNA-binding protein which specifically interacts with a sequence belonging to the group of enhancer core motifs, TGT/cGGT. Electrophoretic mobility shift analysis of cell extracts identified two AML-1-containing protein-DNA complexes whose electrophoretic mobilities were slower than those of complexes formed with AML-1 produced in vitro. Mixing of in vitro-produced AML-1 with cell extracts prior to gel mobility shift analysis resulted in the formation of higher-order complexes. Deletion mutagenesis of AML-1 revealed that the runt homology domain mediates both sequence-specific DNA binding and protein-protein interactions. The hybrid product, AML-1/ETO, which results from the (8;21) translocation and retains the runt homology domain, both recognizes the AML-1 consensus sequence and interacts with other cellular proteins.


Subject(s)
Chromosomes, Human, Pair 21 , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins , Transcription Factors , Transcription, Genetic , Translocation, Genetic , Animals , Base Sequence , Cell Line , Core Binding Factor Alpha 2 Subunit , DNA/metabolism , Drosophila , Drosophila Proteins , Enhancer Elements, Genetic , Humans , Leukemia, Myeloid, Acute/genetics , Molecular Sequence Data , Neoplasm Proteins/genetics , Nuclear Proteins , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured
10.
Mol Cell Biol ; 9(7): 3109-13, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2528683

ABSTRACT

The phenotype of a differentiated cell results from the expression of a unique set of genes in that cell. The differentiation of F9 teratocarcinoma cells in response to retinoic acid and cyclic AMP is an excellent example of this process, as the appearance of several gene products during the course of the differentiation process has been documented. In principle, the activation of gene expression could be due to the appearance of positive-acting factors, the loss of negative-acting factors, or a combination of both. Since F9 cells have been shown to express a cellular E1A analog whereas differentiated F9 cells do not, and it is known that the viral E1A gene exerts a negative effect on transcription of both viral and cellular genes, we determined whether the cellular genes activated during F9 cell differentiation are subject to E1A negative control. We found that infection of differentiated F9 cells with wild-type adenovirus resulted in a decline in the levels of collagen type IV mRNA and plasminogen activator mRNA, both of which are induced by differentiation. At least for the collagen gene, this phenomenon appears to involve a transcriptional repression.


Subject(s)
Adenoviridae/genetics , Cell Differentiation , Collagen/biosynthesis , Gene Expression Regulation , Oncogene Proteins, Viral/physiology , Tissue Plasminogen Activator/genetics , Adenovirus Early Proteins , Animals , Collagen/genetics , Genetic Complementation Test , Oncogene Proteins, Viral/genetics , Phenotype , Tissue Plasminogen Activator/biosynthesis , Transcription, Genetic , Tumor Cells, Cultured
11.
Mol Cell Biol ; 17(3): 1049-56, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9032231

ABSTRACT

Mutations in the retinoblastoma (pRb) tumor suppressor pathway including its cyclin-cdk regulatory kinases, or cdk inhibitors, are a hallmark of most cancers and allow unrestrained E2F-1 transcription factor activity, which leads to unregulated G1-to-S-phase cell cycle progression. Moderate levels of E2F-1 overexpression are tolerated in interleukin 3 (IL-3)-dependent 32D.3 myeloid progenitor cells, yet this induces apoptosis when these cells are deprived of IL-3. However, when E2F activity is augmented by coexpression of its heterodimeric partner, DP-1, the effects of survival factors are abrogated. To determine whether enforced E2F-1 expression selectively sensitizes cells to cytotoxic agents, we examined the effects of chemotherapeutic agents and radiation used in cancer therapy. E2F-1 overexpression in the myeloid cells preferentially sensitized cells to apoptosis when they were treated with the topoisomerase II inhibitor etoposide. Although E2F-1 alone induces moderate levels of p53 and treatment with drugs markedly increased p53, the deleterious effects of etoposide in E2F-1-overexpressing cells were independent of p53 accumulation. Coexpression of Bcl-2 and E2F-1 in 32D.3 cells protected them from etoposide-mediated apoptosis. However, Bcl-2 also prevented apoptosis of these cells upon exposure to 5-fluorouracil and doxorubicin, which were also cytotoxic for control cells. Pretreating E2F-1-expressing cells with ICRF-193, a second topoisomerase II inhibitor that does not damage DNA, protected the cells from etoposide-induced apoptosis. However, ICRF-193 cooperated with DNA-damaging agents to induce apoptosis. Therefore, topoisomerase II inhibition and DNA damage can cooperate to selectively induce p53-independent apoptosis in cells that have unregulated E2F-1 activity resulting from mutations in the pRb pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Carrier Proteins , Cell Cycle Proteins , DNA Damage , DNA-Binding Proteins , Topoisomerase II Inhibitors , Transcription Factors/physiology , Bone Marrow Cells , Cell Cycle , Cell Survival/drug effects , Cell Survival/radiation effects , Diketopiperazines , Doxorubicin/pharmacology , E2F Transcription Factors , E2F1 Transcription Factor , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Fluorouracil/pharmacology , Gamma Rays , Gene Expression , Humans , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/genetics , Tumor Suppressor Protein p53/physiology
12.
Mol Cell Biol ; 18(7): 3915-25, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9632776

ABSTRACT

Control elements of many genes are regulated by multiple activators working in concert to confer the maximal level of expression, but the mechanism of such synergy is not completely understood. The promoter of the human macrophage colony-stimulating factor (M-CSF) receptor presents an excellent model with which we can study synergistic, tissue-specific activation for two reasons. First, myeloid-specific expression of the M-CSF receptor is regulated transcriptionally by three factors which are crucial for normal hematopoiesis: PU.1, AML1, and C/EBPalpha. Second, these proteins interact in such a way as to demonstrate at least two examples of synergistic activation. We have shown that AML1 and C/EBPalpha activate the M-CSF receptor promoter in a synergistic manner. As we report here, AML1 also synergizes, and interacts physically, with PU. 1. Detailed analysis of the physical and functional interaction of AML1 with PU.1 and C/EBPalpha has revealed that the proteins contact one another through their DNA-binding domains and that AML1 exhibits cooperative DNA binding with C/EBPalpha but not with PU.1. This difference in DNA-binding abilities may explain, in part, the differences observed in synergistic activation. Furthermore, the activation domains of all three factors are required for synergistic activation, and the region of AML1 required for synergy with PU.1 is distinct from that required for synergy with C/EBPalpha. These observations present the possibility that synergistic activation is mediated by secondary proteins contacted through the activation domains of AML1, C/EBPalpha, and PU.1.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins , COS Cells , Cell Line , Chlorocebus aethiops , Core Binding Factor Alpha 2 Subunit , DNA/metabolism , HeLa Cells , Humans , Mutagenesis , Phosphorylation , Promoter Regions, Genetic , Transcriptional Activation
13.
Mol Cell Biol ; 14(8): 5558-68, 1994 Aug.
Article in English | MEDLINE | ID: mdl-8035830

ABSTRACT

The myeloperoxidase (MPO) and neutrophil elastase genes are expressed specifically in immature myeloid cells. The integrity of a polyomavirus enhancer core sequence, 5'-AACCACA-3', is critical to the activity of the murine MPO proximal enhancer. This element binds two species, myeloid nuclear factors 1 alpha and 1 beta (MyNF1 alpha and -beta), present in 32D cl3 myeloid cell nuclear extracts. The levels of the MyNF1s increase during early 32D cl3 cell granulocytic differentiation. Both MyNF1 alpha and -beta supershift with an antiserum raised by using a peptide derived from the N terminus of polyomavirus enhancer-binding protein 2/core-binding factor (PEBP2/CBF) alpha subunit. The specific peptide inhibits these supershifts. In vitro-translated PEBP2/CBF DNA-binding domain binds the murine MPO PEBP2/CBF site. An alternate PEBP2/CBF consensus site, 5'-GACCGCA-3', but not a simian virus 40 enhancer core sequence, 5'-TTCCACA-3', binds the MyNF1s in vitro and activates a minimal murine MPO-thymidine kinase promoter in vivo. The murine neutrophil elastase gene 100-bp 5'-flanking sequences contain several functional elements, including potential binding sites for PU.1, C/EBP, c-Myb, and PEBP2/CBF. The functional element 5'-GGCCACA-3' located at positions -66 to 72 differs from the PEBP2/CBF consensus (5'-PuACCPuCA-3') only by an A-to-G transition at position 2. This DNA element binds MyNF1 alpha and -beta weakly. The N terminis of two PEBP2/CBF alpha subunit family members, PEBP2 alpha A and PEBP2 alpha B (murine AML1), are nearly identical, and 32D c13 cl3 cells contain both corresponding mRNAs. Since t(8;21), t(3;21), and inv(16), associated with myeloid leukemias, disrupt subunits of PEBP2/CBF, we speculate that the resulting oncoproteins, AML1-ETO, AML1-EAP, AML1-Evi1, and CBF beta-MYH11, inhibit early myeloid differentiation.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Enzymologic , Leukocytes/enzymology , Neoplasm Proteins , Pancreatic Elastase/genetics , Peroxidase/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , Animals , Base Sequence , Binding Sites , Cloning, Molecular , Core Binding Factor Alpha 1 Subunit , Core Binding Factor alpha Subunits , Core Binding Factor beta Subunit , Core Binding Factors , In Vitro Techniques , Leukocyte Elastase , Mice , Molecular Sequence Data , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides/chemistry , RNA, Messenger/genetics , Transcription Factor AP-2
14.
Mol Cell Biol ; 15(12): 6864-74, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8524253

ABSTRACT

The E2F DNA binding activity consists of a heterodimer between E2F and DP family proteins, and these interactions are required for association of E2F proteins with pRb and the pRb-related proteins p107 and p130, which modulate E2F transcriptional activities. E2F-1 expression is sufficient to release fibroblasts from G0 and induce entry into S phase, yet it also initiates apoptosis. To investigate the mechanisms of E2F-induced apoptosis, we utilized interleukin-3 (IL-3)-dependent 32D.3 myeloid cells, a model of hematopoietic progenitor programmed cell death. In the absence of IL-3, E2F-1 alone was sufficient to induce apoptosis, and p53 levels were diminished. DP-1 alone was not sufficient to induce cell cycle progression or alter rates of death following IL-3 withdrawal. However, overexpression of both E2F-1 and DP-1 led to the rapid death of cells even in the presence of survival factors. In the presence of IL-3, levels of endogenous wild-type p53 increased in response to E2F-1, and coexpression of DP-1 further augmented p53 levels. These results provide evidence that E2F is a functional link between the tumor suppressors p53 and pRb. However, induction of p53 alone was not sufficient to trigger apoptosis, suggesting that the ability of E2F to override survival factors involves additional effectors.


Subject(s)
Apoptosis/physiology , Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Transcription Factors/metabolism , Tumor Suppressor Protein p53/biosynthesis , Animals , Cell Line , DNA Damage , Dexamethasone/pharmacology , E2F Transcription Factors , E2F1 Transcription Factor , Fibroblasts , Gene Expression/drug effects , Humans , Interleukin-3/pharmacology , Kinetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Retinoblastoma-Binding Protein 1 , Time Factors , Transcription Factor DP1 , Transcription Factors/biosynthesis , Transcription Factors/isolation & purification , Transfection
15.
Mol Cell Biol ; 18(1): 322-33, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9418879

ABSTRACT

AML-1B is a hematopoietic transcription factor that is functionally inactivated by multiple chromosomal translocations in human acute myeloblastic and B-cell lymphocytic leukemias. The t(8;21)(q22;q22) translocation replaces the C terminus, including the transactivation domain of AML-1B, with ETO, a nuclear protein of unknown function. We previously showed that AML-1-ETO is a dominant inhibitor of AML-1B-dependent transcriptional activation. Here we demonstrate that AML-1-ETO also inhibits C/EBP-alpha-dependent activation of the myeloid cell-specific, rat defensin NP-3 promoter. AML-1B bound the core enhancer motifs present in the NP-3 promoter and activated transcription approximately sixfold. Similarly, C/EBP-alpha bound NP-3 promoter sequences and activated transcription approximately sixfold. Coexpression of C/EBP-alpha with AML-1B or its family members, AML-2 and murine AML-3, synergistically activated the NP-3 promoter up to 60-fold. The t(8;21) product, AML-1-ETO, repressed AML-1B-dependent activation of NP-3 and completely inhibited C/EBP-alpha-dependent activity as well as the synergistic activation. In contrast, the inv(16) product, which indirectly targets AML family members by fusing their heterodimeric DNA binding partner, CBF-beta, to the myosin heavy chain, inhibited AML-1B but not C/EBP-alpha activation or the synergistic activation. AML-1-ETO and C/EBP-alpha were coimmunoprecipitated and thus physically interact in vivo. Deletion mutants demonstrated that the C terminus of ETO was required for AML-1-ETO-mediated repression of the synergistic activation but not for association with C/EBP-alpha. Finally, overexpression of AML-1-ETO in myeloid progenitor cells prevented granulocyte colony-stimulating factor-induced differentiation. Thus, AML-1-ETO may contribute to leukemogenesis by specifically inhibiting C/EBP-alpha- and AML-1B-dependent activation of myeloid promoters and blocking differentiation.


Subject(s)
Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , DNA-Binding Proteins/genetics , Granulocytes/metabolism , Nuclear Proteins/genetics , Oncogene Proteins, Fusion , Transcription Factors/genetics , Transcription, Genetic , Translocation, Genetic , Animals , CCAAT-Enhancer-Binding Proteins , COS Cells , Cell Differentiation/genetics , Core Binding Factor Alpha 2 Subunit , Gene Expression Regulation, Neoplastic , Granulocytes/cytology , Hematopoiesis/genetics , Humans , Leukemia/genetics , Promoter Regions, Genetic/genetics , RUNX1 Translocation Partner 1 Protein , Rats , Recombinant Fusion Proteins/genetics
16.
Mol Cell Biol ; 19(10): 6566-74, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10490596

ABSTRACT

t(12;21) is the most frequent translocation found in pediatric B-cell acute lymphoblastic leukemias. This translocation fuses a putative repressor domain from the TEL DNA-binding protein to nearly all of the AML-1B transcription factor. Here, we demonstrate that fusion of the TEL pointed domain to the GAL4 DNA-binding domain resulted in sequence-specific transcriptional repression, indicating that the pointed domain is a portable repression motif. The TEL pointed domain functioned equally well when the GAL4 DNA-binding sites were moved 600 bp from the promoter, suggesting an active mechanism of repression. This lead us to demonstrate that wild-type TEL and the t(12;21) fusion protein bind the mSin3A corepressor. In the fusion protein, both TEL and AML-1B contribute mSin3 interaction domains. Deletion mutagenesis indicated that both the TEL and AML-1B mSin3-binding domains contribute to repression by the fusion protein. While both TEL and AML-1B associate with mSin3A, TEL/AML-1B appears to bind this corepressor much more stably than either wild-type protein, suggesting a mode of action for the t(12;21) fusion protein.


Subject(s)
DNA-Binding Proteins/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins , Repressor Proteins/genetics , Transcription Factors/genetics , Translocation, Genetic , Burkitt Lymphoma/genetics , Child , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 21 , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/metabolism , Humans , Models, Genetic , Neoplasm Proteins , Oncogene Proteins, Fusion/metabolism , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-ets , RUNX1 Translocation Partner 1 Protein , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex , Transcription Factors/metabolism , ETS Translocation Variant 6 Protein
17.
Mol Cell Biol ; 20(6): 2075-86, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10688654

ABSTRACT

The ETO protein was originally identified by its fusion to the AML-1 transcription factor in translocation (8;21) associated with the M2 form of acute myeloid leukemia (AML). The resulting AML-1-ETO fusion is an aberrant transcriptional regulator due to the ability of ETO, which does not bind DNA itself, to recruit the transcriptional corepressors N-CoR, SMRT, and Sin3A and histone deacetylases. The promyelocytic leukemia zinc finger (PLZF) protein is a sequence-specific DNA-binding transcriptional factor fused to retinoic acid receptor alpha in acute promyelocytic leukemia associated with the (11;17)(q23;q21) translocation. PLZF also mediates transcriptional repression through the actions of corepressors and histone deacetylases. We found that ETO is one of the corepressors recruited by PLZF. The PLZF and ETO proteins associate in vivo and in vitro, and ETO can potentiate transcriptional repression by PLZF. The N-terminal portion of ETO forms complexes with PLZF, while the C-terminal region, which was shown to bind to N-CoR and SMRT, is required for the ability of ETO to augment transcriptional repression by PLZF. The second repression domain (RD2) of PLZF, not the POZ/BTB domain, is necessary to bind to ETO. Corepression by ETO was completely abrogated by histone deacetylase inhibitors. This identifies ETO as a cofactor for a sequence-specific transcription factor and indicates that, like other corepressors, it functions through the action of histone deactylase.


Subject(s)
Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid/genetics , Proto-Oncogene Proteins , Transcription Factors/genetics , Translocation, Genetic , Acute Disease , Animals , COS Cells , Humans , Kruppel-Like Transcription Factors , Promyelocytic Leukemia Zinc Finger Protein , RUNX1 Translocation Partner 1 Protein , Transfection , Zinc Fingers
18.
Mol Cell Biol ; 16(4): 1349-55, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8657108

ABSTRACT

The t(12;21) translocation is present in up to 30% of childhood B-cell acute lymphoblastic and fuses a potential dimerization motif from the ets-related factor TEL to the N terminus of AML1. The t(12;21) translocation encodes a 93-kDa fusion protein that localizes to a high-salt- and detergent-resistant nuclear compartment. This protein binds the enhancer core motif, TGTGGT, and interacts with the AML-1-binding protein, core-binding factor beta. Although TEL/AML-1B retains the C-terminal domain of AML-1B that is required for transactivation of the T-cell receptor beta enhancer, it fails to activate transcription but rather inhibits the basal activity of this enhancer. TEL/AML-1B efficiently interferes with AML-1B dependent transactivation of the T-cell receptor beta enhancer, and coexpression of wild-type TEL does not reverse this inhibition. The N-terminal TEL helix-loop-helix domain is essential for TEL/AML-1B-mediated repression. Thus, the t(12;21) fusion protein dominantly interferes with AML-1B-dependent transcription, suggesting that the inhibition of expression of AML-1 genes is critical for B-cell leukemogenesis.


Subject(s)
Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 21 , DNA-Binding Proteins/genetics , Leukemia/genetics , Repressor Proteins , Transcription Factors/genetics , Transcription, Genetic , Translocation, Genetic , Base Sequence , Enhancer Elements, Genetic/genetics , Helix-Loop-Helix Motifs , Humans , Molecular Sequence Data , Proto-Oncogene Proteins c-ets , Recombinant Fusion Proteins/genetics , Sequence Deletion , ETS Translocation Variant 6 Protein
19.
Mol Cell Biol ; 16(3): 1231-40, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8622667

ABSTRACT

Transcription factors play a key role in the development and differentiation of specific lineages from multipotential progenitors. Identification of these regulators and determining the mechanism of how they activate their target genes are important for understanding normal development of monocytes and macrophages and the pathogenesis of a common form of adult acute leukemia, in which the differentiation of monocytic cells is blocked. Our previous work has shown that the monocyte-specific expression of the macrophage colony-stimulating factor (M-CSF) receptor is regulated by three transcription factors interacting with critical regions of the M-CSF receptor promoter, including PU.1 and AML1.PU.1 is essential for myeloid cell development, while the AML1 gene is involved in several common leukemia-related chromosome translocations, although its role in hematopoiesis has not been fully identified. Along with AML1, a third factor, Mono A, interacts with a small region of the promoter which can function as a monocyte-specific enhancer when multimerized and linked to a heterologous basal promoter. Here, we demonstrate by electrophoretic mobility shift assays with monocytic nuclear extracts, COS-7 cell-transfected factors, and specific antibodies that the monocyte-enriched factor Mono A is CCAAT enhancer-binding protein (C/EBP). C/EBP has been shown previously to be an important transcription factor involved in hepatocyte and adipocyte differentiation; in hematopoietic cells, C/EBP is specifically expressed in myeloid cells. In vitro binding analysis reveals a physical interaction between C/EBP and AML1. Further transfection studies show that C/EBP and AML1 in concert with the AML1 heterodimer partner CBF beta synergistically activate M-CSF receptor by more then 60 fold. These results demonstrate that C/EBP and AML1 are important factors for regulating a critical hematopoietic growth factor receptor, the M-CSF receptor, suggesting a mechanism of how the AML1 fusion protein could contribute to acute myeloid leukemia. Furthermore, they demonstrate physical and functional interactions between AML1 and C/EBP transcription factor family members.


Subject(s)
DNA-Binding Proteins/metabolism , Monocytes/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins , Cell Line , Core Binding Factor Alpha 2 Subunit , Humans , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Protein Binding , Receptor, Macrophage Colony-Stimulating Factor/genetics , Signal Transduction
20.
Mol Cell Biol ; 21(19): 6470-83, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11533236

ABSTRACT

t(8;21) and t(16;21) create two fusion proteins, AML-1-ETO and AML-1-MTG16, respectively, which fuse the AML-1 DNA binding domain to putative transcriptional corepressors, ETO and MTG16. Here, we show that distinct domains of ETO contact the mSin3A and N-CoR corepressors and define two binding sites within ETO for each of these corepressors. In addition, of eight histone deacetylases (HDACs) tested, only the class I HDACs HDAC-1, HDAC-2, and HDAC-3 bind ETO. However, these HDACs bind ETO through different domains. We also show that the murine homologue of MTG16, ETO-2, is also a transcriptional corepressor that works through a similar but distinct mechanism. Like ETO, ETO-2 interacts with N-CoR, but ETO-2 fails to bind mSin3A. Furthermore, ETO-2 binds HDAC-1, HDAC-2, and HDAC-3 but also interacts with HDAC-6 and HDAC-8. In addition, we show that expression of AML-1-ETO causes disruption of the cell cycle in the G(1) phase. Disruption of the cell cycle required the ability of AML-1-ETO to repress transcription because a mutant of AML-1-ETO, Delta469, which removes the majority of the corepressor binding sites, had no phenotype. Moreover, treatment of AML-1-ETO-expressing cells with trichostatin A, an HDAC inhibitor, restored cell cycle control. Thus, AML-1-ETO makes distinct contacts with multiple HDACs and an HDAC inhibitor biologically inactivates this fusion protein.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Histone Deacetylases/metabolism , Leukemia, Myelomonocytic, Acute/genetics , Oncogene Proteins, Fusion/physiology , Proto-Oncogene Proteins , Repressor Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcription Factors/physiology , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/physiology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Mice , Models, Genetic , Molecular Sequence Data , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Nuclear Receptor Co-Repressor 1 , Oncogene Proteins, Fusion/antagonists & inhibitors , Protein Structure, Tertiary , RUNX1 Translocation Partner 1 Protein , Sequence Homology, Amino Acid , Sin3 Histone Deacetylase and Corepressor Complex , Transcription Factors/antagonists & inhibitors , Transcription, Genetic , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL