Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
Add more filters

Publication year range
1.
Nat Genet ; 18(4): 331-7, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9537414

ABSTRACT

Several proteins that contribute to epigenetic mechanisms of gene regulation contain a characteristic motif of unknown function called the SET (Suvar3-9, Enhancer-of-zeste, Trithorax) domain. We have demonstrated that SET domains mediate highly conserved interactions with a specific family of proteins that display similarity with dual-specificity phosphatases (dsPTPases). These include myotubularin, the gene of which is mutated in a subset of patients with X-linked myotubular myopathy, and Sbf1, a newly isolated homologue of myotubularin. In contrast with myotubularin, Sbf1 lacks a functional catalytic domain which dephosphorylates phospho-tyrosine and serine-containing peptides in vitro. Competitive interference of endogenous SET domain-dsPTPase interactions by forced expression of Sbf1 induced oncogenic transformation of NIH 3T3 fibroblasts and impaired the in vitro differentiation of C2 myoblast cells. We conclude that myotubularin-type phosphatases link SET-domain containing components of the epigenetic regulatory machinery with signalling pathways involved in growth and differentiation.


Subject(s)
Carrier Proteins/physiology , Intracellular Signaling Peptides and Proteins , Protein Tyrosine Phosphatases/physiology , Proteins/metabolism , 3T3 Cells/cytology , 3T3 Cells/metabolism , Amino Acid Sequence , Animals , Binding Sites , Carrier Proteins/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Division/physiology , Cell Transformation, Neoplastic/genetics , Chromosomal Proteins, Non-Histone , Conserved Sequence/genetics , Conserved Sequence/physiology , DNA-Binding Proteins , Histone Chaperones , Humans , Mice , Molecular Sequence Data , Myocardium/cytology , Myocardium/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases, Non-Receptor , Proteins/chemistry , Proteins/isolation & purification , Sequence Homology, Amino Acid , Transcription Factors
2.
J Exp Med ; 164(1): 315-20, 1986 Jul 01.
Article in English | MEDLINE | ID: mdl-3014037

ABSTRACT

Our results indicate that there are two major breakpoint cluster regions in chromosome 18 DNA for t(14;18) translocations in follicular lymphomas. The absence of a pFL-1 homologous transcript in a cell line containing a pFL-2-detectable translocation suggests that there may be two different pathogenetic consequences of t(14;18) translocations. One possibility is that, despite the distances between them (greater than 20 kb), breakpoints in the two cluster regions in some way affect transcription of the same gene product, which has not yet been identified. Alternatively, two separate transcriptional units may be involved. The availability of DNA probes for each of the two t(14;18) breakpoint cluster regions will allow further studies regarding the biologic significance of these two genetically distinct classes of t(14;18) translocations.


Subject(s)
Chromosomes, Human, 13-15 , Chromosomes, Human, 16-18 , Lymphoma/genetics , Translocation, Genetic , Cell Line , Cloning, Molecular , DNA/analysis , DNA Restriction Enzymes , Humans , Karyotyping , Nucleic Acid Hybridization
3.
J Exp Med ; 161(4): 850-63, 1985 Apr 01.
Article in English | MEDLINE | ID: mdl-2984307

ABSTRACT

Configurations of Ig gene DNA were examined in multiple biopsy specimens from seven cases of human B cell lymphoma that showed histologic differences among the specimens within each case. Analysis by Southern blot hybridizations with DNA probes for each of the three Ig loci revealed that the configurations of DNA within these loci were identical among the specimens in two of the cases. This result indicated the monoclonality of these lymphomas, despite differences in histology between biopsy specimens. In contrast, no common nongermline configurations of Ig gene DNA were detected among multiple biopsies in each of three other cases. Therefore, different histologies correlated with separate clones of proliferating B cells in these cases. In the last two cases, the configurations of light chain gene DNA were the same among biopsies in each case, consistent with a monoclonal origin in both lymphomas. However, differences were detected in the configuration of the heavy chain gene DNA. Analysis with a series of DNA probes of the mu heavy chain region indicated that the differences in the DNA configurations of the heavy chain genes from the biopsies probably arose from postrearrangement deletions of either the switch or constant regions of the mu gene. These studies indicate that, contrary to the conventional belief, individual tumors that contain different histologic types of lymphoma within the same patient frequently arise from separate clones of neoplastic cells. Furthermore, the heavy chain genes of monoclonal tumors may show postrearrangement deletions, often resulting from instability of DNA sequences within or around the mu switch region.


Subject(s)
B-Lymphocytes/pathology , Genes , Immunoglobulin Idiotypes/genetics , Lymphoma/immunology , B-Lymphocytes/classification , Clone Cells/classification , Clone Cells/pathology , Cloning, Molecular , DNA Restriction Enzymes , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Idiotypes/immunology , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/immunology , Lymphoma/classification , Lymphoma/genetics , Lymphoma/pathology
4.
J Exp Med ; 167(2): 682-7, 1988 Feb 01.
Article in English | MEDLINE | ID: mdl-3162254

ABSTRACT

DNA spanning a t(7;19) chromosomal translocation breakpoint was isolated from the human T cell line SUP-T7 established from an acute lymphoblastic leukemia. Nucleotide sequence analysis showed that the point of crossover on chromosome 7 occurred immediately adjacent to joining segment J beta 1.1 within the TCR-beta gene, suggesting that this translocation resulted from an error in TCR gene rearrangement. On chromosome 19, the translocation occurred within a previously uncharacterized transcriptional unit for which we propose the name lyl-1. An approximately 1.5-kb RNA is transcribed from this gene in a wide variety of hematolymphoid cell lines. The t(7;19) results in truncation of the lyl-1 gene and production of abnormal-sized RNAs, suggesting a role for lyl-1 in the pathogenesis of this leukemia.


Subject(s)
Leukemia, Lymphoid/genetics , Receptors, Antigen, T-Cell/genetics , Translocation, Genetic , Base Sequence , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 7 , Cloning, Molecular , Humans , Molecular Sequence Data , Transcription, Genetic
5.
J Exp Med ; 167(2): 582-97, 1988 Feb 01.
Article in English | MEDLINE | ID: mdl-3126254

ABSTRACT

To investigate the possible relatedness of the subpopulations that make up so-called biclonal lymphomas, we examined five bigenotypic and biphenotypic follicular lymphomas using DNA probes specific for the t(14;18) chromosomal translocation, which is a characteristic feature of these neoplasms. On Southern blot analysis, both subpopulations from four of five lymphomas contained comigrating t(14;18) DNA rearrangements, confirming the single cell origins for these neoplasms. No comigrating t(14;18) DNA rearrangements were observed in the fifth lymphoma, but nucleotide sequence analysis of cloned, breakpoint DNA showed identical t(14;18) crossovers in the two subpopulations. The migration differences of both the Ig and chromosome 18 DNA rearrangements were shown to result from somatically acquired mutations of the Ig genes from the fifth lymphoma. These studies indicate that Ig gene rearrangements and idiotope expression are not consistently stable clonal markers since they are subject to variability as a result of somatic mutation. Although translocated chromosome 18 DNA rearrangements are more reliable, they may also vary among cells of some tumors since somatic mutation can affect, as well, DNA of translocated alleles in follicular lymphomas.


Subject(s)
B-Lymphocytes/classification , Genotype , Lymphocyte Activation , Lymphoma/genetics , Phenotype , B-Lymphocytes/pathology , Base Sequence , Clone Cells/classification , Clone Cells/pathology , Cloning, Molecular , Genes, Immunoglobulin , Humans , Immunoglobulin Constant Regions/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Joining Region/genetics , Lymphoma/immunology , Lymphoma/pathology , Molecular Sequence Data , Recombination, Genetic
6.
Science ; 228(4700): 722-4, 1985 May 10.
Article in English | MEDLINE | ID: mdl-2986287

ABSTRACT

Cotton-top tamarins were inoculated with sufficient Epstein-Barr virus to induce multiple tumors in each animal within 14 to 21 days. The tumors consisted of large-cell lymphomas that contained multiple copies of the Epstein-Barr virus genome and generated Epstein-Barr virus-carrying cell lines showing no detectable consistent chromosomal abnormality. Hybridization of tumor DNA with immunoglobulin gene probes revealed that each lymphoma was oligo- or monoclonal in origin and that individual tumors from the same animal arose from different B-cell clones. Thus the virus induced multiple transformation events in tamarins in vivo to cause malignant tumors resembling the Epstein-Barr virus-associated lymphomas of patients with organ transplants.


Subject(s)
B-Lymphocytes/microbiology , Burkitt Lymphoma/microbiology , Animals , Burkitt Lymphoma/genetics , Cell Line , DNA, Neoplasm/genetics , Heart Transplantation , Herpesvirus 4, Human , Humans , Neoplasms, Experimental/genetics , Neoplasms, Experimental/microbiology , Nucleic Acid Hybridization , Saguinus
7.
Science ; 246(4928): 379-82, 1989 Oct 20.
Article in English | MEDLINE | ID: mdl-2799390

ABSTRACT

The gene (E2A) that codes for proteins with the properties of immunoglobulin enhancer binding factors E12/E47 was mapped to chromosome region 19p13.2-p13.3, a site associated with nonrandom translocations in acute lymphoblastic leukemias. The majority of t(1;19)(q23;p13)-carrying leukemias and cell lines studied contained rearrangements of E2A as determined by DNA blot analyses. The rearrangements altered the E2A transcriptional unit, resulting in the synthesis of a transcript larger than the normal-sized E2A mRNAs in one of the cell lines with this translocation. These observations indicate that the gene for a transcription factor is located at the breakpoint of a consistently recurring chromosomal translocation in many acute leukemias and suggest a direct role for alteration of such factors in the pathogenesis of some malignancies.


Subject(s)
Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 1 , DNA-Binding Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/genetics , Translocation, Genetic/physiology , Child , Chromosome Mapping , Humans , Tumor Cells, Cultured
8.
Science ; 250(4980): 559-62, 1990 Oct 26.
Article in English | MEDLINE | ID: mdl-2237408

ABSTRACT

Chronic myelogeneous leukemia (CML) is genetically characterized by fusion of the bcr and abl genes on chromosomes 22 and 9, respectively. In most cases, the fusion involves a reciprocal translocation t(9;22)(q34;q11), which produces the cytogenetically distinctive Philadelphia chromosome (Ph1). Fusion can be detected by Southern (DNA) analysis or by in vitro amplification of the messenger RNA from the fusion gene with polymerase chain reaction (PCR). These techniques are sensitive but cannot be applied to single cells. Two-color fluorescence in situ hybridization (FISH) was used with probes from portions of the bcr and abl genes to detect the bcr-abl fusion in individual blood and bone marrow cells from six patients. The fusion event was detected in all samples analyzed, of which three were cytogenetically Ph1-negative. One of the Ph1-negative samples was also PCR-negative. This approach is fast and sensitive, and provides potential for determining the frequency of the abnormality in different cell lineages.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Protein-Tyrosine Kinases , Chromosomes, Human, Pair 22 , Chromosomes, Human, Pair 9 , Genes, abl , Humans , Interphase , Metaphase , Nucleic Acid Hybridization , Philadelphia Chromosome , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcr , Translocation, Genetic
10.
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
11.
Mol Cell Biol ; 16(7): 3884-92, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8668206

ABSTRACT

Using the yeast two-hybrid system, we have isolated a cDNA (designated BBP, for Bcl2-binding protein) for a protein (Bbp) that interacts with Bcl2. Bbp is identical to 53BP2, a partial clone of which was previously isolated in a two-hybrid screen for proteins that interact with p53. In this study, we show that specific interactions of Bbp/53BP2 with either Bcl2 or p53 require its ankyrin repeats and SH3 domain. These interactions can be reproduced in vitro with bacterially expressed fusion proteins, and competition experiments indicate that Bcl2 prevents p53 from binding to Bbp/53BP2. BBP/53BP2 mRNA is abundant in most cell lines examined, but the protein cannot be stably expressed in a variety of cell types by transfection. In transiently transfected cells, Bbp partially colocalizes with Bcl2 in the cytoplasm and results in an increased number of cells at G2/M, possibly accounting for the inability to obtain stable transfectants expressing the protein. These results demonstrate that a single protein can interact with either Bcl2 or p53 both in yeast cells and in vitro. The in vivo significance of these interactions and their potential consequences for cell cycle progression and cell death remain to be determined.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle , Tumor Suppressor Protein p53/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Ankyrin Repeat , Apoptosis , Apoptosis Regulatory Proteins , Carrier Proteins/biosynthesis , Cell Line , Chlorocebus aethiops , Cloning, Molecular , Escherichia coli , Glutathione Transferase/biosynthesis , Mice , Molecular Sequence Data , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2 , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae , Sequence Tagged Sites , Transfection , src Homology Domains
12.
Mol Cell Biol ; 14(9): 5986-96, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8065331

ABSTRACT

The t(17;19) translocation in acute lymphoblastic leukemias results in creation of E2A-hepatic leukemia factor (HLF) chimeric proteins that contain the DNA-binding and protein dimerization domains of the basic leucine zipper (bZIP) protein HLF fused to a portion of E2A proteins with transcriptional activation properties. An in vitro binding site selection procedure was used to determine DNA sequences preferentially bound by wild-type HLF and chimeric E2A-HLF proteins isolated from various t(17;19)-bearing leukemias. All were found to selectively bind the consensus sequence 5'-GTTACGTAAT-3' with high affinity. Wild-type and chimeric HLF proteins also bound closely related sites identified previously for bZIP proteins of both the proline- and acidic amino acid-rich (PAR) and C/EBP subfamilies; however, E2A-HLF proteins were significantly less tolerant of certain deviations from the HLF consensus binding site. These differences were directly attributable to loss of an HLF ancillary DNA-binding domain in all E2A-HLF chimeras and were further exacerbated by a zipper mutation in one isolate. Both wild-type and chimeric HLF proteins displayed transcriptional activator properties in lymphoid and nonlymphoid cells on reporter genes containing HLF or C/EBP consensus binding sites. But on reporter genes with nonoptimal binding sites, their transcriptional properties diverged and E2A-HLF competitively inhibited activation by wild-type PAR proteins. These findings establish a spectrum of binding site-specific transcriptional properties for E2A-HLF which may preferentially activate expression of select subordinate genes as a homodimer and potentially antagonize expression of others through heteromeric interactions.


Subject(s)
DNA-Binding Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Recombinant Fusion Proteins/metabolism , Trans-Activators , Transcription Factors/metabolism , Base Sequence , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 19 , Consensus Sequence , DNA-Binding Proteins/genetics , Gene Expression Regulation , Humans , Leucine Zippers , Lymphocytes/physiology , Macromolecular Substances , Molecular Sequence Data , Oligonucleotide Probes/chemistry , Oncogene Proteins, Fusion/genetics , RNA, Messenger/genetics , Recombinant Fusion Proteins/genetics , Transcription, Genetic , Translocation, Genetic
13.
Mol Cell Biol ; 18(1): 122-9, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9418860

ABSTRACT

The HRX gene (also called MLL, ALL-1, and Htrx) at chromosome band 11q23 is associated with specific subsets of acute leukemias through translocations that result in its fusion with a variety of heterologous partners. Two of these partners, ENL and AF9, code for proteins that are highly similar to each other and as fusions with HRX induce myeloid leukemias in mice as demonstrated by retroviral gene transfer and knock-in experiments, respectively. In the present study, a structure-function analysis was performed to determine the molecular requirements for in vitro immortalization of murine myeloid cells by HRX-ENL. Deletions of either the AT hook motifs or the methyltransferase homology domain of HRX substantially impaired the transforming effects of HRX-ENL. The methyltransferase homology domain was shown to bind non-sequence specifically to DNA in vitro, providing evidence that the full transforming activity of HRX-ENL requires multiple DNA binding structures in HRX. The carboxy-terminal 84 amino acids of ENL, which encode two predicted helical structures highly conserved in AF9, were necessary and sufficient for transformation when they were fused to HRX. Similarly, mutations that deleted one or both of these conserved helices completely abrogated the transcriptional activation properties of ENL. This finding correlates, for the first time, a biological function of an HRX fusion partner with the transforming activity of the chimeric proteins. Our studies support a model in which HRX-ENL induces myeloid transformation by deregulating subordinate genes through a gain of function contributed by the transcriptional effector properties of ENL.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Leukemia/genetics , Neoplasm Proteins , Nuclear Proteins/genetics , Proto-Oncogenes , Recombinant Fusion Proteins/genetics , Transcription Factors , Transcriptional Activation , Amino Acid Sequence , Animals , Cell Line , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase , Humans , Mice , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Nuclear Proteins/metabolism , Structure-Activity Relationship , Translocation, Genetic
14.
Mol Cell Biol ; 17(1): 81-8, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8972188

ABSTRACT

E2a-Pbx1 chimeric oncoproteins result from fusion of the E2A and PBX1 genes at the sites of t(1;19) chromosomal translocations in a subset acute lymphoblastic leukemias. Experimentally, E2a-Pbx1 transforms a variety of cell types, including fibroblasts, myeloid progenitors, and lymphoblasts. Structure-function studies have shown that contributions from both E2a and Pbx1 are necessary for oncogenesis, but the Pbx1 homeodomain is dispensable and the required portion of Pbx1 has not been delineated. In this study, we used deletional and site-directed mutagenesis to identify portions of Pbx1 necessary for oncogenic and transcriptional activities of E2a-Pbx1. These studies defined a motif (named the Hox cooperativity motif [HCM]) carboxy terminal to the Pbx homeodomain that is required for cooperative DNA binding, cellular transcriptional activity, and the oncogenic potential of E2a-Pbx1. The HCM is highly conserved throughout the Pbx/exd subfamily of divergent homeodomain proteins and functions in DNA-binding assays as a potential contact site for Hox dimerization. E2a-Pbx1 proteins with interstitial deletion or single-point mutations in the HCM could neither activate transcription in cellular assays nor transform NIH 3T3 cells. An E2a-Pbx1 mutant containing 50 amino acids of Pbx1b spanning the HCM but lacking the homeodomain was capable of inducing fibroblast transformation. Thus, the HCM is a necessary and sufficient contribution of Pbx1 for oncogenesis induced by E2a-Pbx1 and accounts for its homeodomain-independent transforming properties. Since subtle alterations of the Pbx HCM result in complete abrogation of transforming activity whereas the homeodomain is entirely dispensable, we conclude that interactions mediated by the HCM are more important for transformation by E2a-Pbx1 than interactions with cognate Pbx DNA sites.


Subject(s)
Cell Transformation, Neoplastic/genetics , Homeodomain Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Transcription, Genetic/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Cell Line , Conserved Sequence , DNA/metabolism , Homeodomain Proteins/genetics , Humans , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Oncogene Proteins, Fusion/genetics , Point Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Protein Binding , Recombinant Fusion Proteins/metabolism , Sequence Deletion/genetics , Tumor Cells, Cultured
15.
Mol Cell Biol ; 16(5): 2394-401, 1996 May.
Article in English | MEDLINE | ID: mdl-8628307

ABSTRACT

LYL1 is a basic helix-loop-helix (HLH) protein that was originally discovered because of its translocation into the beta T-cell receptor locus in an acute lymphoblastic leukemia. LYL1 is expressed in many hematolymphoid cells, with the notable exceptions of thymocytes and T cells. Using the yeast two-hybrid system to screen a cDNA library constructed from B cells, we identified the E-box-binding proteins E12 and E47 as potential lymphoid dimerization partners for LYL1. The interaction of LYL1 with E2a proteins was further characterized in vitro and shown to require the HLH motifs of both proteins. Immunoprecipitation analyses showed that in T-ALL and other cell lines, endogenous LYL1 exists in a complex with E2a proteins. A preferred DNA-binding sequence, 5'-AACAGATG(T/g)T-3', for the LYL1-E2a heterodimer was determined by PCR-assisted site selection. Endogenous protein complexes containing both LYL1 and E2a bound this sequence in various LYL1-expressing cell lines and could distinguish between the LYL1 consensus and muE2 sites. These data demonstrate that E2a proteins serve as dimerization partners for the basic HLH protein LYL1 to form complexes with distinctive DNA-binding properties and support the hypothesis that the leukemic properties of the LYL1 and TAL subfamily of HLH proteins could be mediated by recognition of a common set of target genes as heterodimeric complexes with class I HLH proteins.


Subject(s)
DNA-Binding Proteins/metabolism , Helix-Loop-Helix Motifs , Neoplasm Proteins/metabolism , Transcription Factors , Animals , B-Lymphocytes/metabolism , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , Blotting, Western , Cell Line , Chlorocebus aethiops , Cloning, Molecular , Consensus Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , Gene Library , Humans , Macromolecular Substances , Molecular Sequence Data , Mutagenesis, Site-Directed , Neoplasm Proteins/chemistry , Neoplasm Proteins/isolation & purification , Oligodeoxyribonucleotides , Polymerase Chain Reaction , Protein Biosynthesis , Saccharomyces cerevisiae , Sequence Homology, Nucleic Acid , TCF Transcription Factors , Transcription Factor 7-Like 1 Protein , Transcription, Genetic , Transfection
16.
Mol Cell Biol ; 19(7): 5134-42, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10373562

ABSTRACT

Pbx/exd proteins modulate the DNA binding affinities and specificities of Hox proteins and contribute to the execution of Hox-dependent developmental programs in arthropods and vertebrates. Pbx proteins also stably heterodimerize and bind DNA with Meis and Pknox1-Prep1, additional members of the TALE (three-amino-acid loop extension) superclass of homeodomain proteins that function on common genetic pathways with a subset of Hox proteins. In this study, we demonstrated that Pbx and Meis bind DNA as heterotrimeric complexes with Hoxb1 on a genetically defined Hoxb2 enhancer, r4, that mediates the cross-regulatory transcriptional effects of Hoxb1 in vivo. The DNA binding specificity of the heterotrimeric complex for r4 is mediated by a Pbx-Hox site in conjunction with a distal Meis site, which we showed to be required for ternary complex formation and Meis-enhanced transcription. Formation of heterotrimeric complexes in which all three homeodomains bind their cognate DNA sites is topologically facilitated by the ability of Pbx and Meis to interact through their amino termini and bind DNA without stringent half-site orientation and spacing requirements. Furthermore, Meis site mutation in the Hoxb2 enhancer phenocopies Pbx-Hox site mutation to abrogate enhancer-directed expression of a reporter transgene in the murine embryonic hindbrain, demonstrating that DNA binding by all three proteins is required for trimer function in vivo. Our data provide in vitro and in vivo evidence for the combinatorial regulation of Hox and TALE protein functions that are mediated, in part, by their interdependent DNA binding activities as ternary complexes. As a consequence, Hoxb1 employs Pbx and Meis-related proteins, as a pair of essential cofactors in a higher-order molecular complex, to mediate its transcriptional effects on an endogenous Hox response element.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/genetics , Animals , Binding Sites , COS Cells , Consensus Sequence , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dimerization , Homeodomain Proteins/chemistry , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Pre-B-Cell Leukemia Transcription Factor 1 , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Rhombencephalon/metabolism , Solutions , Transcription, Genetic
17.
Mol Cell Biol ; 9(2): 701-10, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2651903

ABSTRACT

We have identified a 24-kilodalton protein that is the product of the human bcl-2 gene, implicated as an oncogene because of its presence at the site of t(14;18) translocation breakpoints. The Bcl-2 protein was detected by specific, highly sensitive rabbit antibodies and was shown to be present in a number of human lymphoid cell lines and tissues, as well as in mouse B cells transfected with a bcl-2 cDNA construct. Characterization of the Bcl-2 protein demonstrated that it has a lipophilic nature and is associated with membrane structures, probably by means of its hydrophobic carboxy-terminal membrane-spanning domain. In t(14;18)-carrying cell lines, the protein is predominantly localized to the perinuclear endoplasmic reticulum, with a minor fraction in the plasma membrane. These properties, together with the observations that Bcl-2 does not have a characteristic signal peptide and is not glycosylated, suggest that it is an integral-membrane protein that spans the bilayer at its C-terminal hydrophobic region but is exposed only at the cytoplasmic surface. The relative abundance of the Bcl-2 protein in various human lymphoid cell lines correlated with transcription of the bcl-2 gene. The protein was abundant in all t(14;18)-carrying cell lines and lymphomas and was also found at lower levels in pre-B-cell lines and nonmalignant lymphoid tissues that do not carry t(14;18) translocations. These results suggest that the Bcl-2 protein is functional in normal B lymphocytes and that a quantitative difference in its expression may play a role in the pathogenesis of lymphomas carrying the t(14;18) translocation.


Subject(s)
Lymphocytes/metabolism , Lymphoma/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Antibody Specificity , Cell Line , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 18 , Humans , Immunohistochemistry , Lymphoma/genetics , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-bcl-2 , Transcription, Genetic , Transfection , Translocation, Genetic
18.
Mol Cell Biol ; 20(13): 4900-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10848615

ABSTRACT

Mammalian SET domain-containing proteins define a distinctive class of chromatin-associated factors that are targets for growth control signals and oncogenic activation. SUV39H1, a mammalian ortholog of Drosophila Su(var)3-9, contains both SET and chromo domains, signature motifs for proteins that contribute to epigenetic control of gene expression through effects on the regional organization of chromatin structure. In this report we demonstrate that SUV39H1 represses transcription in a transient transcriptional assay when tethered to DNA through the GAL4 DNA binding domain. Under these conditions, SUV39H1 displays features of a long-range repressor capable of acting over several kilobases to silence basal promoters. A possible role in chromatin-mediated gene silencing is supported by the localization of exogenously expressed SUV39H1 to nuclear bodies with morphologic features suggestive of heterochromatin in interphase cells. In addition, we show that SUV39H1 is phosphorylated specifically at the G(1)/S cell cycle transition and when forcibly expressed suppresses cell growth. Growth suppression as well as the ability of SUV39H1 to form nuclear bodies and silence transcription are antagonized by the oncogenic antiphosphatase Sbf1 that when hyperexpressed interacts with the SET domain and stabilizes the phosphorylated form of SUV39H1. These studies suggest a phosphorylation-dependent mechanism for regulating the chromatin organizing activity of a mammalian su(var) protein and implicate the SET domain as a gatekeeper motif that integrates upstream signaling pathways to epigenetic regulation and growth control.


Subject(s)
Carrier Proteins/metabolism , Cell Division/physiology , Drosophila Proteins , Intracellular Signaling Peptides and Proteins , Methyltransferases , Repressor Proteins/metabolism , 3T3 Cells , Amino Acid Motifs , Animals , Carrier Proteins/genetics , DNA/metabolism , Drosophila/genetics , G1 Phase , Gene Expression Regulation , Gene Silencing , Humans , Mice , Mutation , Organelles/metabolism , Phosphorylation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/genetics , S Phase , Transcription, Genetic
19.
Mol Cell Biol ; 14(12): 8304-14, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7969166

ABSTRACT

The t(1;19) chromosomal translocation in acute lymphoblastic leukemias creates chimeric E2a-Pbx1 oncoproteins that can act as DNA-binding activators of transcription. A structural analysis of the functional domains of E2a-Pbx1 showed that portions of both E2a and Pbx1 were essential for transformation of NIH 3T3 cells and transcriptional activation of synthetic reporter genes containing PBX1 consensus binding sites. Hyperexpression of wild-type or experimentally truncated Pbx1 proteins was insufficient for transformation, consistent with their inability to activate transcription. When fused with E2a, the Pbx-related proteins Pbx2 and Pbx3 were also transformation competent, demonstrating that all known members of this highly similar subfamily of homeodomain proteins have latent oncogenic potential. The oncogenic contributions of E2a to the chimeras were localized to transactivation motifs AD1 and AD2, as their mutation significantly impaired transformation. Either the homeodomain or Pbx1 amino acids flanking this region could mediate transformation when fused to E2a. However, the homeodomain was not essential for transformation, since a mutant E2a-Pbx1 protein (E2a-Pbx delta HD) lacking the homeodomain efficiently transformed fibroblasts and induced malignant lymphomas in transgenic mice. Thus, transformation mediated by the chimeric oncoprotein E2a-Pbx1 is absolutely dependent on motifs acquired from E2a but the Pbx1 homeodomain is optional. The latter finding suggests that E2a-Pbx1 may interact with cellular proteins that assist or mediate alterations in gene expression responsible for oncogenesis even in the absence of homeodomain-DNA interactions.


Subject(s)
Adenovirus E2 Proteins/physiology , Cell Transformation, Neoplastic , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Developmental , Genes, Homeobox , Homeodomain Proteins/chemistry , Homeodomain Proteins/physiology , Oncogene Proteins, Fusion/physiology , Proto-Oncogene Proteins/chemistry , Transcription Factors/physiology , 3T3 Cells , Animals , DNA-Binding Proteins/physiology , Homeodomain Proteins/metabolism , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Transgenic , Pre-B-Cell Leukemia Transcription Factor 1 , Proto-Oncogene Proteins/metabolism , Recombinant Fusion Proteins , Structure-Activity Relationship
20.
Mol Cell Biol ; 19(6): 4443-51, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10330184

ABSTRACT

The hepatic leukemia factor (HLF) gene codes for a basic region-leucine zipper (bZIP) protein that is disrupted by chromosomal translocations in a subset of pediatric acute lymphoblastic leukemias. HLF undergoes fusions with the E2A gene, resulting in chimeric E2a-Hlf proteins containing the E2a transactivation domains and the Hlf bZIP DNA binding and dimerization motifs. To investigate the in vivo role of this chimeric bZIP protein in oncogenic transformation, its expression was directed to the lymphoid compartments of transgenic mice. Within the thymus, E2a-Hlf induced profound hypoplasia, premature involution, and progressive accumulation of a T-lineage precursor population arrested at an early stage of maturation. In the spleen, mature T cells were present but in reduced numbers, and they lacked expression of the transgene, suggesting further that E2a-Hlf expression was incompatible with T-cell differentiation. In contrast, mature splenic B cells expressed E2a-Hlf but at lower levels and without apparent adverse or beneficial effects on their survival. Approximately 60% of E2A-HLF mice developed lymphoid malignancies with a mean latency of 10 months. Tumors were monoclonal, consistent with a requirement for secondary genetic events, and displayed phenotypes of either mid-thymocytes or, rarely, B-cell progenitors. We conclude that E2a-Hlf disrupts the differentiation of T-lymphoid progenitors in vivo, leading to profound postnatal thymic depletion and rendering B- and T-cell progenitors susceptible to malignant transformation.


Subject(s)
Cell Transformation, Neoplastic , DNA-Binding Proteins/physiology , Lymphocytes/physiology , Oncogene Proteins, Fusion/physiology , Age Factors , Animals , B-Lymphocytes/metabolism , Basic-Leucine Zipper Transcription Factors , DNA-Binding Proteins/analysis , Flow Cytometry , Immunohistochemistry , Lymphoma/pathology , Mice , Mice, Inbred Strains , Mice, Transgenic , Models, Biological , Models, Genetic , Neoplasms, Experimental , Oncogene Proteins, Fusion/analysis , Stem Cells/physiology , Survival , T-Lymphocytes/metabolism , Thymus Gland/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL