Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters











Publication year range
2.
Curr Opin Hematol ; 8(4): 218-23, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11561159

ABSTRACT

The in-frame fusion of mixed lineage leukemia to CREB binding protein has been cloned from several patients with t-acute myeloid leukemia and a t(11;16)(q23;p13). A murine retroviral transduction model of mixed lineage leukemia fused to CREB binding protein successfully recapitulates the disease. Interestingly, the mice also develop a preleukemic phase reminiscent of what is often seen in patients with t(11;16). From this work, it was determined that minimally, the amino terminus of mixed lineage leukemia fused to the bromodomain and histone acetyltransferase domain of CREB binding protein are necessary for developing acute myeloid leukemia. This model provides a useful tool for understanding the biologic basis of mixed lineage leukemia leukemogenesis and for developing and testing potential therapeutic agents.


Subject(s)
DNA-Binding Proteins/genetics , Disease Models, Animal , Leukemia, Myeloid/etiology , Nuclear Proteins/genetics , Proto-Oncogenes , Retroviridae/genetics , Trans-Activators/genetics , Transcription Factors , Transduction, Genetic , Animals , Artificial Gene Fusion , CREB-Binding Protein , Histone-Lysine N-Methyltransferase , Mice , Myeloid-Lymphoid Leukemia Protein , Myeloproliferative Disorders/etiology
3.
Cancer Res ; 61(1): 59-63, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11196198

ABSTRACT

A partial nontandem duplication (PNTD) of mixed lineage leukemia (MLL) gene is described in B-cell acute lymphoid leukemia without structural cytogenetic abnormalities at 11q23 and 9p22. A duplicated portion of MLL is interrupted by the insertion of a region of 9p22 that includes the 3'-end of the AF9 gene. The PNTD encodes: (a) a PNTD transcript; (b) a partial tandem duplication of MLL; and (c) a chimeric transcript fusing MLL to the 3'-end of AF9, mimicking the t(9;11)(p22;q23) and expressed 1024-fold higher than the other two. The MLL PNTD, therefore, contributes toward leukemogenesis through simultaneous production of fusion transcripts that are otherwise encoded by three distinct genetic defects.


Subject(s)
Burkitt Lymphoma/genetics , DNA-Binding Proteins/genetics , Gene Rearrangement , Proto-Oncogenes , RNA, Messenger/genetics , Transcription Factors , Alternative Splicing/genetics , Blotting, Southern , Chromosome Breakage , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 9/genetics , Exons , Histone-Lysine N-Methyltransferase , Humans , Myeloid-Lymphoid Leukemia Protein , Nuclear Proteins/genetics , Recombinant Fusion Proteins/genetics , Repetitive Sequences, Nucleic Acid , Reverse Transcriptase Polymerase Chain Reaction , Translocation, Genetic , Tumor Cells, Cultured
4.
Genes Chromosomes Cancer ; 29(4): 333-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11066077

ABSTRACT

Cell lines derived from patients with leukemia are used in many molecular biology studies. Here we report the cytogenetic analysis of the THP-1 cell line using G-banding, fluorescence in situ hybridization (FISH), and spectral karyotyping (SKY), and the molecular characterization of the MLL-AF9 rearrangement by RT-PCR. The THP-1 cell line was established from the peripheral blood of a 1-year-old boy with acute monocytic leukemia (AML-M5). THP-1 is near-diploid and consists of two related subclones with a number of aberrations, including the t(9;11), associated with AML M5. The use of FISH allowed us to identify and characterize otherwise hidden cytogenetic rearrangements, which include duplication of the 3' portion of MLL in the derivative 9 chromosome and a deletion of the 5' portion of the AF9 gene involved in the translocation. In addition to confirming the FISH results, SKY allowed for a more precise characterization of the karyotype of THP-1 and allowed us to identify other abnormalities in this cell line, including der(1)t(1;12), der(20)t(1;20), deletions 6p, 12p, and 17p, trisomy 8, and monosomy 10. Sequencing of the RT-PCR product showed a direct in-frame fusion product on the derivative chromosome 11 between exon 6 (exon 9) of MLL and exon 5 of AF9, which is most commonly involved in MLL-AF9 translocations. This study demonstrates that combining different techniques to achieve a more precise characterization of the THP-1 cell line provides important information that will be valuable for understanding the critical events required for leukemogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Leukemia, Monocytic, Acute/genetics , Nuclear Proteins/genetics , Proto-Oncogenes , Transcription Factors , Translocation, Genetic/genetics , Amino Acid Sequence , Base Sequence , Chromosome Aberrations/genetics , Chromosome Banding , Chromosome Disorders , Chromosomes, Human, Pair 9/genetics , Histone-Lysine N-Methyltransferase , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Oncogene Proteins, Fusion/genetics , Tumor Cells, Cultured
5.
EMBO J ; 19(17): 4655-64, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10970858

ABSTRACT

As a result of the recurring translocation t(11;16) (q23;p13.3), MLL (mixed-lineage leukemia) is fused in frame to CBP (CREB binding protein). This translocation has been documented almost exclusively in cases of acute leukemia or myelodysplasia secondary to therapy with drugs that target DNA topo isomerase II. The minimal chimeric protein that is produced fuses MLL to the bromodomain, histone acetyltransferase (HAT) domain, EIA-binding domain and steroid-receptor coactivator binding domains of CBP. We show that transplantation of bone marrow retrovirally transduced with MLL-CBP induces myeloid leukemias in mice that are preceded by a long preleukemic phase similar to the myelodysplastic syndrome (MDS) seen in many t(11;16) patients but unusual for other MLL translocations. Structure-function analysis demonstrated that fusion of both the bromodomain and HAT domain of CBP to the amino portion of MLL is required for full in vitro transformation and is sufficient to induce the leukemic phenotype in vivo. This suggests that the leukemic effect of MLL-CBP results from the fusion of the chromatin association and modifying activities of CBP with the DNA binding activities of MLL.


Subject(s)
DNA-Binding Proteins/metabolism , Leukemia, Myeloid/pathology , Myelodysplastic Syndromes/metabolism , Nuclear Proteins/metabolism , Proto-Oncogenes , Recombinant Fusion Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors , Animals , Base Sequence , CREB-Binding Protein , DNA Primers , DNA-Binding Proteins/genetics , Histone-Lysine N-Methyltransferase , Mice , Myelodysplastic Syndromes/pathology , Myeloid-Lymphoid Leukemia Protein , Nuclear Proteins/genetics , Recombinant Fusion Proteins/genetics , Trans-Activators/genetics
6.
Hum Mol Genet ; 9(11): 1671-9, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10861294

ABSTRACT

The human AF9 gene at 9p22 is one of the most common fusion partner genes with the MLL gene at 11q23, resulting in the t(9;11)(p22;q23). The MLL-AF9 fusion gene is associated with de novo acute myelo-genous leukemia (AML), rarely with acute lymphocytic leukemia (ALL) and with therapy related leukemia (t-AML). The AF9 gene is >100 kb and two patient breakpoint cluster regions (BCRs) have been identified; BCR1 is within intron 4, previously called site A, whereas BCR2 or site B spans introns 7 and 8. Patient breakpoint locations were determined previously by RT-PCR and by genomic DNA cloning. In this study, we defined the exon-intron boundaries and identified several different structural elements in AF9 including a co-localizing in vivo DNA topo II cleavage site and an in vitro DNase I hypersensitive (DNase 1 HS) site in intron 7 in BCR2. Reversibility experiments demonstrated a religation of the topo II cleavage sites. The location of the in vivo topo II cleavage site was confirmed in vitro using a topo II cleavage assay. In addition, two scaffold associated regions (SARs) are located centromeric to the topo II and DNase I HS cleavage sites and border both patient breakpoint regions: SAR1 is located in intron 4, whereas SAR2 encompasses parts of exons 5-7. This study demonstrates that the patient breakpoint regions of AF9 share the same structural elements as the MLL BCR. We describe a DNA breakage and repair model for non-homologous recombination between MLL and its partner genes, particularly AF9.


Subject(s)
DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Leukemia/genetics , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogenes , Transcription Factors , Binding Sites , Cell Line , Chromatin/genetics , DNA Topoisomerases, Type II/metabolism , DNA, Neoplasm/chemistry , Deoxyribonuclease I/metabolism , Histone-Lysine N-Methyltransferase , Humans , Introns , Jurkat Cells , K562 Cells , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Nuclear Proteins/metabolism , Recombination, Genetic , Sequence Analysis, DNA , Translocation, Genetic , Tumor Cells, Cultured
7.
Proc Natl Acad Sci U S A ; 96(11): 6428-33, 1999 May 25.
Article in English | MEDLINE | ID: mdl-10339604

ABSTRACT

MLL (ALL1, Htrx, HRX), which is located on chromosome band 11q23, frequently is rearranged in patients with therapy-related acute myeloid leukemia who previously were treated with DNA topoisomerase II inhibitors. In this study, we have identified a fusion partner of MLL in a 10-year-old female who developed therapy-related acute myeloid leukemia 17 months after treatment for Hodgkin's disease. Leukemia cells of this patient had a t(11;17)(q23;q25), which involved MLL as demonstrated by Southern blot analysis. The partner gene was cloned from cDNA of the leukemia cells by use of a combination of adapter reverse transcriptase-PCR, rapid amplification of 5' cDNA ends, and BLAST database analysis to identify expressed sequence tags. The full-length cDNA of 2.8 kb was found to be an additional member of the septin family, therefore it was named MSF (MLL septin-like fusion). Members of the septin family conserve the GTP binding domain, localize in the cytoplasm, and interact with cytoskeletal filaments. A major 4-kb transcript of MSF was expressed ubiquitously; a 1.7-kb transcript was found in most tissues. An additional 3-kb transcript was found only in hematopoietic tissues. By amplification with MLL exon 5 forward primer and reverse primers in MSF, the appropriately sized products were obtained. MSF is highly homologous to hCDCrel-1, which is a partner gene of MLL in leukemias with a t(11;22)(q23;q11.2). Further analysis of MSF may help to delineate the function of MLL partner genes in leukemia, particularly in therapy-related leukemia.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 17 , DNA-Binding Proteins/genetics , GTP Phosphohydrolases , GTP-Binding Proteins/genetics , Hodgkin Disease/drug therapy , Leukemia, Myeloid/chemically induced , Leukemia, Myeloid/genetics , Proto-Oncogenes , Transcription Factors , Translocation, Genetic , Acute Disease , Amino Acid Sequence , Animals , Base Sequence , Child , Chromosome Mapping , Cyclophosphamide/administration & dosage , Drosophila/genetics , Female , GTP-Binding Proteins/chemistry , Histone-Lysine N-Methyltransferase , Humans , Male , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Organ Specificity , Prednisone/administration & dosage , Procarbazine/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Septins , Sequence Alignment , Sequence Homology, Amino Acid , Vincristine/administration & dosage , Zinc Fingers
8.
Leukemia ; 12(12): 1895-902, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9844920

ABSTRACT

The t(9;11)(p22;q23) is the most common chromosomal translocation in topoisomerase II inhibitor therapy-related acute myeloid leukemia (tAML). This translocation fuses the MLL and AF9 proto-oncogenes producing a novel chimeric protein. In order to gain insight into the mechanism generating the t(9;11) and to clarify the role topoisomerase II inhibition may play in that mechanism we have cloned and sequenced the breakpoints from four tAML patients with the t(9;11). This sequence analysis identifies topoisomerase II consensus binding sequences near or at the chromosome 11 and chromosome 9 breakpoints in all four patients. One patient also had the consensus binding sequence for the TRANSLIN DNA-binding protein at the 9p22 and 11q23 breakpoints. Our results further support a direct role for topoisomerase II in the genesis of these tAML translocations.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 9/genetics , DNA-Binding Proteins/genetics , Leukemia, Myeloid/genetics , Neoplasm Proteins/genetics , Neoplasms, Second Primary/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogenes , Topoisomerase II Inhibitors , Transcription Factors , Translocation, Genetic , Acute Disease , Adolescent , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , Cloning, Molecular , DNA-Binding Proteins/analysis , Female , Histone-Lysine N-Methyltransferase , Humans , Leukemia, Myeloid/chemically induced , Male , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Neoplasm Proteins/analysis , Neoplasms, Second Primary/chemically induced , Oncogene Proteins, Fusion/analysis , Polymerase Chain Reaction/methods , Sensitivity and Specificity , Sequence Analysis, DNA
9.
Blood ; 92(10): 3793-803, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9808573

ABSTRACT

The human myeloid-lymphoid leukemia gene, MLL (also called ALL-1, Htrx, or HRX ), maps to chromosomal band 11q23. MLL is involved in translocations that result in de novo acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), mixed lineage leukemia, and also in therapy AML (t-AML) and therapy ALL (t-ALL) resulting from treatment with DNA topoisomerase II (topo II) targeting drugs. MLL can recombine with more than 30 other chromosomal bands, of which 16 of the partner genes have been cloned. Breaks in MLL occur in an 8. 3-kb breakpoint cluster region (BCR) encompassing exons 5 through 11. We recently demonstrated that 75% of de novo patient breakpoints in MLL mapped in the centromeric half of the BCR between two scaffold-associated regions (SAR), whereas 75% of the t-AML patient breakpoints mapped to the telomeric half of the BCR within a strong SAR. We have mapped additional structural elements in the BCR. An in vivo DNA topo II cleavage site (induced with several different drugs that target topo II) mapped near exon 9 in three leukemia cell lines. A strong DNase I hypersensitive site (HS) also mapped near exon 9 in four leukemia cell lines, including two in which MLL was rearranged [a t(6;11) and a t(9;11)], and in two lymphoblastoid cell lines with normal MLL. Two of the leukemia cell lines also showed an in vivo topo II cleavage site. Our results suggest that the chromatin structure of the MLL BCR may influence the location of DNA breaks in both de novo and therapy-related leukemias. We propose that topo II is enriched in the MLL telomeric SAR and that it cleaves the DNase I HS site after treatment with topo II inhibitors. These events may be involved in recombination associated with t-AML/t-ALL breakpoints mapping in the MLL SAR.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 11/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , Deoxyribonuclease I/metabolism , Leukemia/genetics , Proto-Oncogenes , Transcription Factors , Cell Transformation, Neoplastic/genetics , Chromosome Mapping , Chromosomes, Human, Pair 11/ultrastructure , Histone-Lysine N-Methyltransferase , Humans , Myeloid-Lymphoid Leukemia Protein , Recombination, Genetic , Substrate Specificity , Translocation, Genetic , Tumor Cells, Cultured
10.
Leukemia ; 12(7): 1119-27, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9665199

ABSTRACT

A new cell line with megakaryoblastic features, designated UoC-M1, was established from the malignant cells of a 68-year-old patient with acute myeloid leukemia. The patient's leukemic cells reacted with alpha-naphthyl acetate esterase and acid phosphatase and expressed CD7, CD24, CD34, CD38, CD45, HLA-DR and CD61. Cytogenetic analysis of the patient's malignant cells (and of the UoC-M1 cells) showed a human, male hypodiploid karyotype with many chromosome rearrangements and marker chromosomes. Spectral karyotyping (SKY) analysis complemented the G-banded karyotyping and clarified several chromosomal translocations and identified the marker chromosomes. Fluorescence in situ hybridization (FISH) and SKY analysis demonstrated that one marker chromosome contained three segments of chromosome 9 interspersed with three segments of chromosome 11, as well as a portion of chromosome 19. FISH analysis with a probe for MLL revealed that the UoC-M1 cells contained four copies of the MLL gene. Southern blot analysis determined that the MLL gene had a germline profile while Northern and Western analyses showed that the MLL mRNAs and protein were of the appropriate sizes. This is the first report of amplification of the MLL gene which may be an additional mechanism of leukemogenesis or disease progression.


Subject(s)
DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Megakaryocytes/cytology , Proto-Oncogenes , Transcription Factors , Tumor Cells, Cultured , Aged , Blotting, Northern , Blotting, Southern , Blotting, Western , Gene Amplification , Gene Rearrangement , Histone-Lysine N-Methyltransferase , Humans , Immunophenotyping , In Situ Hybridization, Fluorescence , Karyotyping , Male , Megakaryocytes/physiology , Microscopy, Electron , Myeloid-Lymphoid Leukemia Protein
11.
Genes Chromosomes Cancer ; 20(2): 185-95, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9331569

ABSTRACT

The MLL gene at chromosome 11, band q23, is involved in translocations with as many as 40 different chromosomal bands. Virtually all breakpoints occur within an 8.3 kb BamHI fragment and result in 5' MLL fused to partner genes in a 5'-3' orientation. The translocation t(9;11)(p22;q23), which results in the fusion of MLL to AF9, is the most common of the 11q23 chromosomal abnormalities observed in de novo acute myeloid leukemia (AML), in therapy related leukemia (t-AML), and rarely in acute lymphoblastic leukemia (ALL). We have studied 24 patients with a t(9;11) and an MLL rearrangement, including 19 patients with AML, four with t-AML, and one with ALL. To understand the mechanisms of this illegitimate recombination, we cloned and sequenced the t(9;11) translocation breakpoint junctions on both derivative chromosomes from one AML patient and from the Mono Mac 6 (MM6) cell line, which was derived from a patient with AML. Two different complex junctions were noted. In the AML patient, both chromosome 11 and 9 breaks were staggered, occurred in Alu DNA sequences, and resulted in a 331 bp duplication. In the MM6 cell line, breaks in chromosomes 11 and 9 were also staggered, but, in contrast to the finding in the AML patient, the breaks did not involve Alu DNA sequences and resulted in a 664 bp deletion at the breakpoints. Using reverse transcriptase (RT-) PCR, we analyzed 11 patient samples, including the two just described, for MML-AF9 fusions. The fusion occurred in six of seven AML patients, two of two t-AML patients, one patient with ALL, and in the MM6 cell line. Interestingly, all of the breaks within the AF9 gene in AML patients occurred in the central AF9 exon, called Site A by others, whereas in the single ALL patient the breakpoint mapped to a more 3' region of the AF9 gene. Our data, when combined with those of others, suggest that the fusion point within the AF9 gene, and thus the amount of AF9 material included in the MLL-AF9 fusion gene product, may influence the phenotype of the resulting leukemia. This further supports the proposal that the MML translocation partner genes play a critical role in the leukemogenic process.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 9/genetics , DNA-Binding Proteins/genetics , Leukemia, Myeloid/genetics , Nuclear Proteins/genetics , Proto-Oncogenes , Transcription Factors , Translocation, Genetic/genetics , Acute Disease , Adolescent , Adult , Aged , Animals , Artificial Gene Fusion , Blotting, Southern , Child , Child, Preschool , Chromosome Breakage , Chromosome Mapping , Cloning, Molecular , Cricetinae , DNA, Neoplasm/analysis , Female , Gene Rearrangement , Genomic Library , Histone-Lysine N-Methyltransferase , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence , Infant , Male , Middle Aged , Myeloid-Lymphoid Leukemia Protein , Polymerase Chain Reaction , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA-Directed DNA Polymerase , Tumor Cells, Cultured
12.
Proc Natl Acad Sci U S A ; 94(16): 8732-7, 1997 Aug 05.
Article in English | MEDLINE | ID: mdl-9238046

ABSTRACT

The recurring translocation t(11;16)(q23;p13.3) has been documented only in cases of acute leukemia or myelodysplasia secondary to therapy with drugs targeting DNA topoisomerase II. We show that the MLL gene is fused to the gene that codes for CBP (CREB-binding protein), the protein that binds specifically to the DNA-binding protein CREB (cAMP response element-binding protein) in this translocation. MLL is fused in-frame to a different exon of CBP in two patients producing chimeric proteins containing the AT-hooks, methyltransferase homology domain, and transcriptional repression domain of MLL fused to the CREB binding domain or to the bromodomain of CBP. Both fusion products retain the histone acetyltransferase domain of CBP and may lead to leukemia by promoting histone acetylation of genomic regions targeted by the MLL AT-hooks, leading to transcriptional deregulation via aberrant chromatin organization. CBP is the first partner gene of MLL containing well defined structural and functional motifs that provide unique insights into the potential mechanisms by which these translocations contribute to leukemogenesis.


Subject(s)
Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 16 , DNA-Binding Proteins/genetics , Leukemia, Myeloid/genetics , Nuclear Proteins/genetics , Proto-Oncogenes , Trans-Activators , Transcription Factors/genetics , Translocation, Genetic , Acute Disease , Amino Acid Sequence , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Base Sequence , CREB-Binding Protein , Histone-Lysine N-Methyltransferase , Humans , Leukemia, Myeloid/drug therapy , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein
13.
Blood ; 90(2): 535-41, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9226152

ABSTRACT

The involvement of 11q23-balanced translocations in acute leukemia after treatment with drugs that inhibit the function of DNA topoisomerase II (topo II) is being recognized with increasing frequency. We and others have shown that the gene at 11q23 that is involved in all of these treatment-related leukemias is MLL (also called ALL1, Htrx, and HRX). In general, the translocations in these leukemias are the same as those occurring in de novo leukemia [eg, t(9;11), t(11;19), and t(4;11)], with the treatment-related leukemias accounting for no more than 5% to 10% of any particular translocation type. We have cloned the t(11;16)(q23;p13.3) and have shown that it involves MLL and CBP (CREB binding protein). The CBP gene was recently identified as a partner gene in the t(8;16) that occurs in acute myelomonocytic leukemia (AML-M4) de novo and rarely in treatment-related acute myeloid leukemia. We have studied eight t(11;16) patients, all of whom had prior therapy with drugs targetting topo II with fluorescence in situ hybridization (FISH) using a probe for MLL and a cosmid contig covering the CBP gene. Both probes were split in all eight patients and the two derivative (der) chromosomes were each labeled with both probes. Use of an approximately 100-kb PAC located at the breakpoint of chromosome 16 from one patient revealed some variability in the breakpoint because it was on the der(16) in three patients, on the der(11) in another, and split in four others. We assume that the critical fusion gene is 5'MLL/3'CBP. Our series of patients is unusual because three of them presented with a myelodysplastic syndrome (MDS) most similar to chronic myelomonocytic leukemia (CMMoL) and one other had dyserythropoiesis; MDS is rarely seen in 11q23 translocations either de novo or with t-AML. Using FISH and these same probes to analyze the lineage of bone marrow cells from one patient with CMMoL, we showed that all the mature monocytes contained the fusion genes as did some of the granulocytes and erythroblasts; none of the lymphocytes contained the fusion gene. The function of MLL is not well understood, but many domains could target the MLL protein to particular chromatin complexes. CBP is an adapter protein that is involved in regulating transcription. It is also involved in histone acetylation, which is thought to contribute to an increased level of gene expression. The fusion gene could alter the CBP protein such that it is constitutively active; alternatively, it could modify the chromatin-association functions of MLL.


Subject(s)
Antineoplastic Agents/adverse effects , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 16 , DNA-Binding Proteins/genetics , Leukemia/drug therapy , Leukemia/genetics , Myelodysplastic Syndromes/genetics , Neoplasms, Second Primary/genetics , Neoplasms/drug therapy , Nuclear Proteins/genetics , Proto-Oncogenes , Trans-Activators , Transcription Factors/genetics , Translocation, Genetic , Adolescent , Adult , CREB-Binding Protein , Child , Child, Preschool , Chromosome Mapping , Female , Histone-Lysine N-Methyltransferase , Humans , In Situ Hybridization, Fluorescence , Leukemia/chemically induced , Leukemia/pathology , Male , Middle Aged , Myelodysplastic Syndromes/chemically induced , Myelodysplastic Syndromes/pathology , Myeloid-Lymphoid Leukemia Protein , Neoplasms/radiotherapy , Neoplasms, Second Primary/chemically induced , Topoisomerase II Inhibitors , Zinc Fingers
14.
Genes Chromosomes Cancer ; 19(1): 14-21, 1997 May.
Article in English | MEDLINE | ID: mdl-9135990

ABSTRACT

The gene BCL6 encodes a zinc finger protein with similarities to transcription factors. We previously reported that a number of viral genomes, including human immunodeficiency virus type I (HIV-1), contain sequences which are similar to the BCL6 DNA-binding consensus in their promoter regions. Electrophoretic mobility shift assays showed that the full-length BCL6 protein extracted from transfected COS cells and a bacterially expressed truncated protein containing the BCL6 zinc fingers can bind specifically to DNA from the U3 promoter/enhancer region of HIV-1. Transient transfections were performed to analyze the effects of the BCL6 protein on luciferase expression driven by the HIV-1 long terminal repeat (LTR) sequences. Full-length BCL6 significantly repressed luciferase activity compared with multiple controls. We conclude that the BCL6 protein can bind to the HIV-1 promoter-enhancer region and contains a domain upstream of its zinc fingers that can repress transcription from the HIV-1 LTR.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Viral , HIV Enhancer , HIV-1/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins/physiology , Transcription Factors/physiology , Transcription, Genetic , Zinc Fingers , Animals , Base Sequence , Binding Sites , Blotting, Western , COS Cells , Consensus Sequence , DNA/metabolism , DNA, Complementary/chemistry , DNA-Binding Proteins/genetics , Electrophoresis, Polyacrylamide Gel , Genes, Reporter , HeLa Cells , Humans , Luciferases/genetics , Molecular Sequence Data , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-6 , Transcription Factors/genetics
15.
Blood ; 88(9): 3535-45, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8896421

ABSTRACT

Invins(10;11)(p12;q23q12) is one of the rare but recurring chromosome rearrangements seen in acute monoblastic leukemia. We cloned the proximal 10p breakpoint from one patient and showed that the MLL gene at 11q23 was fused to the 3' portion of AF10 at 10p12. In addition, we cloned the telomeric 10p junction and we found that the 5' portion of AF10 was juxtaposed to a previously unidentified gene at 11q12, which we call HEAB (a human homolog to a hypothetical Caenorhabditis elegans ATP/GTP-binding protein). These results indicate that the AF10 gene is split into a 5' AF10 and a 3' AF10 portion by the 11q23q12 chromosome segment and that both breakpoint junctions result in fusion transcripts of 5' AF10/HEAB and MLL/3' AF10. Only the MLL/3' AF10 fusion mRNA results in an in-frame fusion. Northern blot analysis of HEAB expression shows that a 2.0-kb major transcript is expressed ubiquitously in human tissues and is especially abundant in testis and skeletal muscle, whereas a 3.2-kb minor transcript is noted with the highest level of expression in thymus and peripheral blood leukocytes. The HEAB gene encodes a 425-amino acid protein that is rich in valine and leucine. HEAB protein shows high homology in its entire amino acid sequence to a putative C elegans protein and contains an adenosine triphosphate (ATP)/guanosine triphosphate (GTP)-binding motif that has homology to the ATP-binding transporter superfamily or to GTP-binding proteins. Our results could explain the high frequency of complex insertion and other rearrangement events that involve 10p12 and 11q12 and 11q23. The finding that different portions of a single gene are involved in fusions with two independent genes in the same leukemic cell is unique in the analysis of chromosome translocations.


Subject(s)
Caenorhabditis elegans/genetics , Chromosomes, Human, Pair 11 , DNA-Binding Proteins/genetics , GTP-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Proto-Oncogenes , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Caenorhabditis elegans/metabolism , Chromosome Mapping , Histone-Lysine N-Methyltransferase , Humans , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Sequence Analysis , Sequence Homology, Amino Acid , Translocation, Genetic
16.
Genes Chromosomes Cancer ; 15(4): 206-16, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8703846

ABSTRACT

The t(6;11)(q27;23) is one of the most common translocations observed in patients with acute myeloid leukemia (AML). The translocation breakpoint involves the MLL gene, which is the human homolog of the Drosophila trithorax gene, at 11q23 and the AF6 gene at 6q27. Reverse transcriptase-polymerase chain reaction (RT-PCR) using an MLL sense primer and an AF6 antisense primer detected the MLL/AF6 fusion cDNA from three leukemia patients with the t(6;11) [two AML and one T-acute lymphoblastic leukemia (ALL)] and one cell line. The fusion point in the AF6 cDNA from these cases is identical, regardless of the leukemia phenotype. The ML-2 cell line, which was established from a patient with AML that developed after complete remission of T-cell lymphoma, has retained an 11q23-24 deletion from the lymphoma stage and has acquired the t(6;11) with development of AML. The ML-2 cells have no normal MLL gene on Southern blot analysis, which indicates that an intact MLL gene is not necessary for survival of leukemic cells.


Subject(s)
Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 6 , Leukemia, Myeloid, Acute/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , Proto-Oncogenes , Transcription Factors , Translocation, Genetic , Adolescent , Adult , Amino Acid Sequence , Base Sequence , Cell Line , Chromosome Mapping , Cloning, Molecular , DNA, Complementary , DNA-Binding Proteins/genetics , Histone-Lysine N-Methyltransferase , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein
17.
Blood ; 87(5): 1912-22, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8634439

ABSTRACT

A major unresolved question for 11q23 translocations involving MLL is the chromosomal mechanism(s) leading to these translocations. We have mapped breakpoints within the 8.3-kb BamHI breakpoint cluster region in 31 patients with acute lymphoblastic leukemia and acute myeloid leukemia (AML) de novo and in 8 t-AML patients. In 23 of 31 leukemia de novo patients, MLL breakpoints mapped to the centromeric half (4.57 kb) of the breakpoint cluster region, whereas those in eight de novo patients mapped to the telomeric half (3.87 kb). In contrast, only two t-AML breakpoints mapped in the centromeric half, whereas six mapped in the telomeric half. The difference in distribution of the leukemia de novo breakpoints is statistically significant (P = .02). A similar difference in distribution of breakpoints between de novo patients and t-AML patients has been reported by others. We identified a low- or weak-affinity scaffold attachment region (SAR) mapping just centromeric to the breakpoint cluster region, and a high-affinity SAR mapping within the telomeric half of the breakpoint cluster region. Using high stringency criteria to define in vitro vertebrate topoisomerase II (topo II) consensus sites, one topo II site mapped adjacent to the telomeric SAR, whereas six mapped within the SAR. Therefore, 74% of leukemia de novo and 25% of t-AML breakpoints map to the centromeric half of the breakpoint cluster region map between the two SARs; in contrast, 26% of the leukemia de novo and 75% of the t-AML patient breakpoints map to the telomeric half of the breakpoint cluster region that contains both the telomeric SAR and the topo II sites. Thus, the chromatin structure of the MLL breakpoint cluster region may be important in determining the distribution of the breakpoints. The data suggest that the mechanism(s) leading to translocations may differ in leukemia de novo and in t-AML.


Subject(s)
Chromatin/ultrastructure , Chromosomes, Human, Pair 11/ultrastructure , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , Leukemia/genetics , Neoplasms, Second Primary/genetics , Proto-Oncogenes , Transcription Factors , Translocation, Genetic , Acute Disease , Adolescent , Adult , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/therapeutic use , Base Sequence , Binding Sites , Centromere/ultrastructure , Child , Child, Preschool , Consensus Sequence , DNA, Neoplasm/genetics , Etoposide/adverse effects , Etoposide/therapeutic use , Female , Histone-Lysine N-Methyltransferase , Humans , Infant , Leukemia/chemically induced , Male , Middle Aged , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Neoplasms, Second Primary/chemically induced , Telomere/ultrastructure , Teniposide/adverse effects , Teniposide/therapeutic use , Topoisomerase II Inhibitors , Tumor Cells, Cultured
18.
Proc Natl Acad Sci U S A ; 93(4): 1642-7, 1996 Feb 20.
Article in English | MEDLINE | ID: mdl-8643684

ABSTRACT

The EVI1 gene, located at chromosome band 3q26, is overexpressed in some myeloid leukemia patients with breakpoints either 5' of the gene in the t(3;3)(q21;q26) or 3' of the gene in the inv(3)(q21q26). EVI1 is also expressed as part of a fusion transcript with the transcription factor AML1 in the t(3;21)(q26;q22), associated with myeloid leukemia. In cells with t(3;21), additional fusion transcripts are AML1-MDS1 and AML1-MDS1-EVI1. MDS1 is located at 3q26 170-400 kb upstream (telomeric) of EVI1 in the chromosomal region in which some of the breakpoints 5' of EVI1 have been mapped. MDS1 has been identified as a single gene as well as a previously unreported exon(s) of EVI1 We have analyzed the relationship between MDS1 and EVI1 to determine whether they are two separate genes. In this report, we present evidence indicating that MDS1 exists in normal tissues both as a unique transcript and as a normal fusion transcript with EVI1, with an additional 188 codons at the 5' end of the previously reported EVI1 open reading frame. This additional region has about 40% homology at the amino acid level with the PR domain of the retinoblastoma-interacting zinc-finger protein RIZ. These results are important in view of the fact that EVI1 and MDS1 are involved in leukemia associated with chromosomal translocation breakpoints in the region between these genes.


Subject(s)
DNA-Binding Proteins/genetics , Leukemia, Myeloid/genetics , Multigene Family , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion , Proteins/genetics , Proto-Oncogene Proteins , Proto-Oncogenes , Recombinant Fusion Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Base Sequence , Chromosomes, Human, Pair 21/ultrastructure , Chromosomes, Human, Pair 3/ultrastructure , Core Binding Factor Alpha 2 Subunit , DNA, Complementary/genetics , DNA-Binding Proteins/biosynthesis , Exons/genetics , Gene Expression Regulation, Leukemic , Gene Library , Histone-Lysine N-Methyltransferase , Humans , Kidney/metabolism , MDS1 and EVI1 Complex Locus Protein , Molecular Sequence Data , Neoplasm Proteins/biosynthesis , Nuclear Proteins/chemistry , Open Reading Frames , Pancreas/metabolism , Protein Biosynthesis , RNA Splicing , Recombinant Fusion Proteins/biosynthesis , Sequence Alignment , Sequence Homology , Transcription Factors/biosynthesis , Transcription, Genetic , Translocation, Genetic , Zinc Fingers/genetics
19.
Curr Top Microbiol Immunol ; 211: 259-68, 1996.
Article in English | MEDLINE | ID: mdl-8585957

ABSTRACT

Translocations involving chromosome band 11q23, found in acute lymphoid and myeloid leukemias, disrupt the MLL gene. This gene encodes a putative transcription factor with regions of homology to several other proteins including the zinc fingers and other domains of the Drosophila trithorax gene product, and the "AT-hook" DNA-binding motif of high mobility group proteins. We have previously demonstrated that MLL contains transcriptional activation and repression domains using a GAL4 fusion protein system (21). The repression domain, which is capable of repressing transcription 3-5-fold, is located centromeric to the breakpoint region of MLL. The activation domain, located telomeric to the breakpoint region, activated transcription from a variety of promoters including ones containing only basal promoter elements. The level of activation was very high, ranging from 10-fold to more than 300-fold, depending on the promoter and cell line used for transient transfection. In translocations involving MLL, the protein produced from the der(11) chromosome which contains the critical junction for leukemogenesis includes the AT-hook domain and the repression domain. We assessed the DNA binding capability of the MLL AT-hook domain using bacterially expressed and purified AT-hook protein. In a gel mobility shift assay, the MLL AT-hook domain could bind cruciform DNA, recognizing structure rather than sequence of the target DNA. This binding could be specifically competed with Hoechst 33258 dye and with distamycin. In a nitrocellulose protein-DNA binding assay, the MLL AT-hook domain could bind to AT-rich SARs, but not to non-SAR DNA fragments. The role that the AT-hook binding to DNA may play in vivo is unclear, but it is likely that DNA binding could affect downstream gene regulation. The AT-hook domain retained on the der(11) would potentially recognize a different DNA target than the one normally recognized by the intact MLL protein. Furthermore, loss of an activation domain while retaining a repression domain on the der(11) chromosome could alter the expression of various downstream target genes, suggesting potential mechanisms of action for MLL in leukemia.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Proto-Oncogenes , Transcription Factors , Amino Acid Sequence , DNA/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Histone-Lysine N-Methyltransferase , Humans , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Nuclear Matrix/metabolism , Nucleic Acid Conformation , Protein Binding , Recombinant Proteins/metabolism
20.
Blood ; 86(9): 3613-4, 1995 Nov 01.
Article in English | MEDLINE | ID: mdl-7579475
SELECTION OF CITATIONS
SEARCH DETAIL