Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Leukemia ; 37(9): 1850-1859, 2023 09.
Article in English | MEDLINE | ID: mdl-37532789

ABSTRACT

The transcription factor CCAAT-enhancer binding factor alpha (C/ebpα) is a master controller of myeloid differentiation that is expressed as long (p42) and short (p30) isoform. Mutations within the CEBPA gene selectively deleting p42 are frequent in human acute myeloid leukemia. Here we investigated the individual genomics and transcriptomics of p42 and p30. Both proteins bound to identical sites across the genome. For most targets, they induced a highly similar transcriptional response with the exception of a few isoform specific genes. Amongst those we identified early growth response 1 (Egr1) and tribbles1 (Trib1) as key targets selectively induced by p42 that are also underrepresented in CEBPA-mutated AML. Egr1 executed a program of myeloid differentiation and growth arrest. Oppositely, Trib1 established a negative feedback loop through activation of Erk1/2 kinase thus placing differentiation under control of signaling. Unexpectedly, differentiation elicited either by removal of an oncogenic input or by G-CSF did not peruse C/ebpα as mediator but rather directly affected the cell cycle core by upregulation of p21/p27 inhibitors. This points to functions downstream of C/ebpα as intersection point where transforming and differentiation stimuli converge and this finding offers a new perspective for therapeutic intervention.


Subject(s)
Granulocyte Precursor Cells , Leukemia, Myeloid, Acute , Humans , Granulocyte Precursor Cells/metabolism , Leukemia, Myeloid, Acute/genetics , Cell Differentiation , Protein Isoforms/genetics , Mutation , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism
2.
Haematologica ; 107(11): 2601-2616, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35546301

ABSTRACT

The homeobox transcription factors HoxA9 and Meis1 are causally involved in the etiology of acute myeloid leukemia. While HoxA9 alone immortalizes cells, cooperation with Meis1 is necessary to induce a full leukemic phenotype. Here, we applied degron techniques to elucidate the leukemogenic contribution of Meis1. Chromatin immunoprecipitation experiments revealed that Meis1 localized mainly to H3K27 acetylated and H3K4 mono-methylated enhancers preactivated by HoxA9. Chromatin association of Meis1 required physical presence of HoxA9 and all Meis1 DNA interactions were rapidly lost after HoxA9 degradation. Meis1 controlled a gene expression pattern dominated by Myc, ribosome biogenesis and ribosomal RNA synthesis genes. While Myc accounted for the cell cycle stimulating effect of Meis1, overexpression of this oncogene alone did not accelerate leukemogenesis. Besides its effect on Myc, Meis1 induced transcription of ribosomal biogenesis genes. This was accompanied by an elevated resistance against inhibition of ribosomal RNA synthesis and translation, but without affecting steady-state protein synthesis. Finally, we demonstrate that HoxA9 and Meis1 proteins are stabilized by post-translational modification. Mutation of HoxA9/Meis1 phosphorylation sites or inhibition of casein kinase 2 lead to rapid protein degradation suggesting a potential pathway for pharmacological intervention.


Subject(s)
Leukemia, Myeloid, Acute , Neoplasm Proteins , Carcinogenesis/genetics , Gene Expression Regulation, Leukemic , Homeodomain Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/genetics , RNA, Ribosomal , Animals , Mice
3.
Blood Adv ; 4(15): 3626-3638, 2020 08 11.
Article in English | MEDLINE | ID: mdl-32766858

ABSTRACT

Mixed-lineage leukemia (MLL) fusions are transcriptional activators that induce leukemia, with a dismal prognosis that mandates further elucidation of their transformation mechanism. In this study, knockdown of the direct MLL-ENL target gene polypyrimidine tract binding protein-1 (PTBP1) was rate limiting for cell proliferation and caused a metabolic phenotype associated with reduced glucose consumption and lactate production. This effect was accompanied by a reduction of splice isoform-2 of pyruvate kinase M (PKM2). Because PKM2 restricts glycolytic outflow to provide anabolic intermediates, we tested the consequences of glucose, energy, and Ser/Gly starvation for cell physiology. Administration of deoxyglucose, energetic decoupling with rotenone, and inhibition of Ser biosynthesis by CBR5884 had a significantly stronger influence on self-renewal and survival of transformed cells than on normal controls. In particular, inhibition of Ser synthesis, which branches off glycolysis caused accumulation of reactive oxygen species, DNA damage, and apoptosis, predominantly in leukemic cells. Depletion of exogenous Ser/Gly affected proliferation and self-renewal of murine and human leukemia samples, even though they are classified as nonessential amino acids. Response to Ser/Gly starvation correlated with glucose transport, but did not involve activation of the AMPK energy homeostasis system. Finally, survival times in transplantation experiments were significantly extended by feeding recipients a Ser/Gly-free diet. These results suggest selective starvation as an option for supportive leukemia treatment.


Subject(s)
Leukemia , Animals , Cell Proliferation , Glycolysis , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Leukemia/genetics , Mice , Polypyrimidine Tract-Binding Protein/metabolism , Protein Isoforms , Transcription Factors
4.
Blood Adv ; 2(22): 3137-3148, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30463913

ABSTRACT

Ectopic expression of the oncogenic transcription factor HoxA9 is a major cause of acute myeloid leukemia (AML). Here, we demonstrate that HoxA9 is a specific substrate of granule proteases. Protease knockout allowed the comprehensive determination of genome-wide HoxA9 binding sites by chromatin immunoprecipitation sequencing in primary murine cells and a human AML cell line. The kinetics of enhancer activity and transcription rates in response to alterations of an inducible HoxA9 were determined. This permitted identification of HoxA9-controlled enhancers and promoters, allocation to their respective transcription units, and discrimination against HoxA9-bound, but unresponsive, elements. HoxA9 triggered an elaborate positive-feedback loop that drove expression of the complete Hox-A locus. In addition, it controlled key oncogenic transcription factors Myc and Myb and directly induced the cell cycle regulators Cdk6 and CyclinD1, as well as telomerase, drawing the essential blueprint for perturbation of proliferation by leukemogenic HoxA9 expression.


Subject(s)
Cell Cycle Checkpoints , Cyclin-Dependent Kinase 6/metabolism , Homeodomain Proteins/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 6/genetics , Enhancer Elements, Genetic , Gene Editing , Histones/genetics , Histones/metabolism , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myc/genetics , Transcription, Genetic
5.
Blood ; 131(6): 662-673, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29217648

ABSTRACT

Eleven-nineteen leukemia (ENL) is a chromatin reader present in complexes stimulating transcriptional elongation. It is fused to mixed-lineage leukemia (MLL) in leukemia, and missense mutations have been identified in Wilms tumor and acute myeloid leukemia. Here we demonstrate that ENL overcomes polycomb silencing through recruitment of PAF1 via the conserved YEATS domain, which recognizes acetylated histone H3. PAF1 was responsible for antirepressive activities of ENL in vitro, and it determined the transforming potential of MLL-ENL. MLL-ENL target loci showed supraphysiological PAF1 binding, hyperubiquitination of histone H2B and hypomodification with H2AUb, resulting in accelerated transcription rates. YEATS mutations induced a gain of function, transforming primary hematopoietic cells in vitro and in transplantation assays through aberrant transcription and H2B ubiquitination of Hoxa9 and Meis1 Mechanistically, H3 and PAF1 competed for ENL interaction, with activating mutations favoring PAF1 binding, whereas the MLL moiety provided a constitutive PAF1 tether allowing MLL fusions to circumvent H3 competition.


Subject(s)
Carrier Proteins/metabolism , Cell Transformation, Neoplastic , DNA-Binding Proteins/metabolism , Gene Silencing , Leukemia/genetics , Polycomb-Group Proteins/genetics , Transcription Factors/metabolism , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , HEK293 Cells , Histones/metabolism , Humans , Leukemia/metabolism , Leukemia/pathology , Mice , Mice, Inbred BALB C , Mutation , Protein Binding/genetics , Protein Domains/genetics , Protein Processing, Post-Translational , Transcription Factors/chemistry , Transcription Factors/genetics
6.
Nat Cell Biol ; 19(7): 844-855, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28650479

ABSTRACT

Leukaemogenesis requires enhanced self-renewal, which is induced by oncogenes. The underlying molecular mechanisms remain incompletely understood. Here, we identified C/D box snoRNAs and rRNA 2'-O-methylation as critical determinants of leukaemic stem cell activity. Leukaemogenesis by AML1-ETO required expression of the groucho-related amino-terminal enhancer of split (AES). AES functioned by inducing snoRNA/RNP formation via interaction with the RNA helicase DDX21. Similarly, global loss of C/D box snoRNAs with concomitant loss of rRNA 2'-O-methylation resulted in decreased leukaemia self-renewal potential. Genomic deletion of either C/D box snoRNA SNORD14D or SNORD35A suppressed clonogenic potential of leukaemia cells in vitro and delayed leukaemogenesis in vivo. We further showed that AML1-ETO9a, MYC and MLL-AF9 all enhanced snoRNA formation. Expression levels of C/D box snoRNAs in AML patients correlated closely with in vivo frequency of leukaemic stem cells. Collectively, these findings indicate that induction of C/D box snoRNA/RNP function constitutes an important pathway in leukaemogenesis.


Subject(s)
Cell Proliferation , Cell Self Renewal , Cell Transformation, Neoplastic/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Leukemia/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , RNA, Small Nucleolar/metabolism , Ribonucleoproteins/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Co-Repressor Proteins , Core Binding Factor Alpha 2 Subunit/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation, Leukemic , Genetic Predisposition to Disease , HEK293 Cells , HL-60 Cells , Humans , K562 Cells , Leukemia/genetics , Leukemia/pathology , Methylation , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Phenotype , Protein Interaction Maps , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , RNA, Small Nucleolar/genetics , RUNX1 Translocation Partner 1 Protein , Repressor Proteins/genetics , Repressor Proteins/metabolism , Ribonucleoproteins/genetics , Signal Transduction , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , U937 Cells
7.
Cell Rep ; 15(2): 310-22, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27050521

ABSTRACT

MLL fusions are leukemogenic transcription factors that enhance transcriptional elongation through modification of chromatin and RNA Pol II. Global transcription rates and chromatin changes accompanying the transformation process induced by MLL-ENL were monitored by nascent RNA-seq and ChIP-seq, revealing 165 direct target genes separated into two distinct clades. ME5 genes bound MLL-ENL at the promoter, relied on DOT1L-mediated histone methylation, and coded preferentially for transcription factors, including many homeobox genes. A distinct ME3 group accumulated MLL-ENL beyond the termination site, was dependent on P-TEFb-mediated phosphorylation of RNA Pol II for transcription, and translated mainly into proteins involved in RNA biology and ribosome assembly. This dichotomy was reflected by a differential sensitivity toward small molecule inhibitors, suggesting the possibility of a combinatorial strategy for treatment of MLL-induced leukemia.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation, Leukemic , Genes, Neoplasm , Oncogene Proteins, Fusion/metabolism , Animals , Gene Expression Profiling , Mice , Sequence Analysis, RNA , Transcription, Genetic
8.
Haematologica ; 100(7): 905-13, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25911551

ABSTRACT

Hox homeobox transcription factors drive leukemogenesis efficiently only in the presence of Meis or Pbx proteins. Here we show that Pbx3 and Meis1 need to dimerize to support Hox-induced leukemia and we analyze the molecular details of this cooperation. In the absence of Pbx3, Meis1 was highly unstable. As shown by a deletion analysis Meis1 degradation was contingent on a motif coinciding with the Pbx-binding domain. Either deletion of this sequence or binding to Pbx3 prolonged the half-life of Meis1 by preventing its ubiquitination. Meis1 break-down could also be blocked by inhibition of the ubiquitin proteasome system, indicating tight post-transcriptional control. In addition, Meis1 and Pbx3 cooperated genetically as overexpression of Pbx3 induced endogenous Meis1 transcription. These functional interactions translated into in vivo activity. Blocking Meis1/Pbx3 dimerization abrogated the ability to enhance proliferation and colony-forming cell numbers in primary cells transformed by Hoxa9. Furthermore, expression of Meis1 target genes Flt3 and Trib2 was dependent on Pbx3/Meis1 dimerization. This correlated with the requirement of Meis1 to bind Pbx3 in order to form high affinity DNA/Hoxa9/Meis1/Pbx3 complexes in vitro. Finally, kinetics and severity of disease in transplantation assays indicated that Pbx3/Meis1 dimers are rate-limiting factors for Hoxa9-induced leukemia.


Subject(s)
Gene Expression Regulation, Leukemic , Homeodomain Proteins/genetics , Leukemia/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , Amino Acid Motifs , Animals , Binding Sites , Disease Models, Animal , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia/metabolism , Leukemia/pathology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Multimerization , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Proteolysis , Proto-Oncogene Proteins/metabolism , Signal Transduction , Ubiquitination , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
9.
Cell Rep ; 3(5): 1553-66, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23623499

ABSTRACT

Stimulation of transcriptional elongation is a key activity of leukemogenic MLL fusion proteins. Here, we provide evidence that MLL-ENL also inhibits Polycomb-mediated silencing as a prerequisite for efficient transformation. Biochemical studies identified ENL as a scaffold that contacted the elongation machinery as well as the Polycomb repressive complex 1 (PRC1) component CBX8. These interactions were mutually exclusive in vitro, corresponding to an antagonistic behavior of MLL-ENL and CBX8 in vivo. CBX8 inhibited elongation in a specific reporter assay, and this effect was neutralized by direct association with ENL. Correspondingly, CBX8-binding-defective MLL-ENL could not fully activate gene loci necessary for transformation. Finally, we demonstrate dimerization of MLL-ENL as a neomorphic activity that may augment Polycomb inhibition and transformation.


Subject(s)
Myeloid-Lymphoid Leukemia Protein/metabolism , Polycomb Repressive Complex 1/metabolism , Transcriptional Elongation Factors/metabolism , Cell Transformation, Neoplastic , Dimerization , HEK293 Cells , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase , Humans , Myeloid-Lymphoid Leukemia Protein/genetics , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transcriptional Elongation Factors/genetics
10.
Blood ; 120(19): 4018-27, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22990017

ABSTRACT

HOX proteins are widely involved in hematopoietic development. These transcription factors combine a conserved DNA-binding homeobox with a divergent N-terminus that mediates interaction with variable cofactors. The resulting combinatorial diversity is thought to be responsible for mammalian HOX specificity. Contrasting this proposed mechanism for normal HOX function, here we demonstrate that, in the context of hematopoietic immortalization and leukemogenesis, individual HOX properties are governed almost exclusively by the homeodomain. Swap experiments between HOXA1 and HOXA9, 2 members of nonrelated paralog groups, revealed that gene expression patterns of HOX transformed cells in vitro are determined by the nature of the homeodomain. Similar results were seen in vivo during HOX-mediated leukemogenesis. An exchange of the homeodomains was sufficient to convert the slow, low-penetrance phenotype of HOXA1-induced leukemia to the aggressive fast-acting disease elicited by HOXA9 and vice versa. Mutation and deletion studies identified several subregions within the DNA binding domain responsible for paralog specificity. Previously defined binding sites for PBX cofactors within the exchangeable, nonhomeobox segment were dispensable for in vitro oncogenic HOX activity but affected in vivo disease development. The transcriptional activator domain shared by HOXA1 and HOXA9 at the very N-terminus proved essential for all transformation.


Subject(s)
Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Leukemia/genetics , Phenotype , Amino Acid Sequence , Animals , Cell Line , Cell Transformation, Neoplastic/genetics , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genetic Vectors/genetics , Homeodomain Proteins/chemistry , Humans , Leukemia/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Protein Conformation , Protein Interaction Domains and Motifs , Sequence Alignment
11.
Blood ; 115(14): 2910-8, 2010 Apr 08.
Article in English | MEDLINE | ID: mdl-20130239

ABSTRACT

HOX homeobox genes are important regulators of normal and malignant hematopoiesis. Abdominal-type HOXA genes like HOXA9 are highly leukemogenic. However, little is known about transformation by anterior HOXA genes. Here we performed a comprehensive assessment of the oncogenic potential of every HOXA gene in primary hematopoietic cells. With exception of HOXA2 and HOXA5, all HOXA genes caused a block or delay of hematopoietic differentiation and cooperated with Meis1. No evidence for the alleged tumor-suppressor function of HOXA5 could be found. Whereas all active HOXA genes immortalized mixed granulocytic/monocytic populations, HOXA13 preferentially specified monocytoid development. The anterior HOXA genes HOXA1, HOXA4, and HOXA6 transformed cells, generating permanent cell lines, although they did so less potently than HOXA9. Upon transplantation these lines induced myeloproliferation and acute myeloid leukemia in recipient animals. Kinetic studies with inducible HOX derivatives demonstrated that anterior HOXA genes autonomously contributed to cellular transformation. This function was not mediated by endogenous Hoxa9, which was persistently expressed in cells transformed by anterior HOX genes. In summary our results demonstrate a hitherto unexpected role of anterior HOXA genes in hematopoietic malignancy.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Hematologic Neoplasms/metabolism , Homeodomain Proteins/biosynthesis , Leukemia, Myeloid, Acute/metabolism , Multigene Family , Neoplasm Proteins/metabolism , Animals , Cell Transformation, Neoplastic/pathology , Hematologic Neoplasms/genetics , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred BALB C , Neoplasm Proteins/genetics
12.
PLoS Biol ; 7(11): e1000249, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19956800

ABSTRACT

Fusion proteins composed of the histone methyltransferase mixed-lineage leukemia (MLL) and a variety of unrelated fusion partners are highly leukemogenic. Despite their prevalence, particularly in pediatric acute leukemia, many molecular details of their transforming mechanism are unknown. Here, we provide mechanistic insight into the function of MLL fusions, demonstrating that they capture a transcriptional elongation complex that has been previously found associated with the eleven-nineteen leukemia protein (ENL). We show that this complex consists of a tight core stabilized by recursive protein-protein interactions. This central part integrates histone H3 lysine 79 methylation, RNA Polymerase II (RNA Pol II) phosphorylation, and MLL fusion partners to stimulate transcriptional elongation as evidenced by RNA tethering assays. Coimmunoprecipitations indicated that MLL fusions are incorporated into this complex, causing a constitutive recruitment of elongation activity to MLL target loci. Chromatin immunoprecipitations (ChIP) of the homeobox gene A cluster confirmed a close relationship between binding of MLL fusions and transcript levels. A time-resolved ChIP utilizing a conditional MLL fusion singled out H3K79 methylation as the primary parameter correlated with target expression. The presence of MLL fusion proteins also kept RNA Pol II in an actively elongating state and prevented accumulation of inhibitory histone methylation on target chromatin. Hox loci remained open and productive in the presence of MLL fusion activity even under conditions of forced differentiation. Finally, MLL-transformed cells were particularly sensitive to pharmacological inhibition of RNA Pol II phosphorylation, pointing to a potential treatment for MLL. In summary, we show aberrant transcriptional elongation as a novel mechanism for oncogenic transformation.


Subject(s)
Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Leukemia/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Transcriptional Elongation Factors/metabolism , Animals , Cell Line, Tumor , Chromatin Assembly and Disassembly , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Flavonoids , Histone-Lysine N-Methyltransferase , Humans , Mice , Mice, Inbred BALB C , Piperidines
13.
Methods Mol Biol ; 538: 347-67, 2009.
Article in English | MEDLINE | ID: mdl-19277583

ABSTRACT

The two-hybrid system is a genetic method to search for and to identify direct interaction partners of a protein of interest. This method is instrumental to elucidate the transformation mechanism of several oncogenes that play a role in childhood leukaemia. With respect to mixed lineage leukaemia gene (MLL) fusions, two-hybrid screening was applied to discover proteins that bind to various MLL fusion partners. Here we describe a streamlined protocol that enables any average molecular biology laboratory to conduct and evaluate a standard two-hybrid screen. Starting with a general explanation of the biological background of the two-hybrid method, this chapter covers the construction of bait vectors and two comprehensive protocols for screening either by yeast mating or yeast transformation. In addition, it also gives guidelines for the evaluation of two-hybrid results.


Subject(s)
Protein Interaction Mapping/methods , Proteins/metabolism , Saccharomyces cerevisiae/genetics , Two-Hybrid System Techniques , Blotting, Western , Cloning, Molecular , Gene Library , Genetic Vectors , Histone-Lysine N-Methyltransferase , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Protein Binding , Proteins/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Transformation, Genetic
14.
Blood ; 110(13): 4445-54, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17855633

ABSTRACT

Chimeric proteins joining the histone methyltransferase MLL with various fusion partners trigger distinctive lymphoid and myeloid leukemias. Here, we immunopurified proteins associated with ENL, a protein commonly fused to MLL. Identification of these ENL-associated proteins (EAPs) by mass spectrometry revealed enzymes with a known role in transcriptional elongation (RNA polymerase II C-terminal domain kinase [RNAPolII CTD] positive transcription elongation factor b [pTEFb]), and in chromatin modification (histone-H3 methyltransferase DOT1L) as well as other frequent MLL partners (AF4, AF5q31, and LAF4), and polycomb group members (RING1, CBX8, and BCoR). The composition of EAP was further verified by coimmunoprecipitation, 2-hybrid analysis, pull-down, and colocalization experiments. Purified EAP showed a histone H3 lysine 79-specific methylase activity, displayed a robust RNAPolII CTD kinase function, and counteracted the effect of the pTEFb inhibitor 5,6-dichloro-benzimidazole-riboside. In vivo, an ENL knock-down diminished genome-wide as well as gene-specific H3K79 dimethylation, reduced global run-on elongation, and inhibited transient transcriptional reporter activity. According to structure-function data, DOT1L recruitment was important for transformation by the MLL-ENL fusion derivative. These results suggest a function of ENL in histone modification and transcriptional elongation.


Subject(s)
Chromatin Assembly and Disassembly , Methyltransferases/metabolism , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Positive Transcriptional Elongation Factor B/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Transcription, Genetic , Animals , Cell Line , Histone-Lysine N-Methyltransferase , Humans , Immunoprecipitation , Mice , Mitochondrial Membrane Transport Proteins , Multiprotein Complexes/analysis , Myeloid-Lymphoid Leukemia Protein , Neoplasm Proteins/isolation & purification , Nuclear Proteins/isolation & purification , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Protein Binding , Recombinant Fusion Proteins , Repressor Proteins/metabolism , Transcription Factors/isolation & purification
15.
Oncogene ; 24(35): 5525-32, 2005 Aug 18.
Article in English | MEDLINE | ID: mdl-15856011

ABSTRACT

Mixed lineage leukemia (MLL) fusion proteins are derived from translocations at 11q23 that occur in aggressive subtypes of leukemia. As a consequence, MLL is joined to different unrelated proteins to form oncogenic transcription factors. Here we demonstrate a direct interaction between several nuclear MLL fusion partners and present evidence for a role of these proteins in histone binding. In two-hybrid studies, ENL interacted with AF4 and AF5q31 as well as with a fragment of AF10. A structure-function analysis revealed that the AF4/AF5q31/AF10 binding domain in ENL coincided with the C-terminus that is essential for transformation by MLL-ENL. The ENL/AF4 association was corroborated by GST-pulldown experiments and by mutual coprecipitation. Both proteins colocalized in vivo in a nuclear speckled pattern. Moreover, AF4 and ENL coeluted on sizing columns together with the known ENL binding partner Polycomb3, suggesting the presence of a multiprotein complex. The overexpression of ENL alone activated a reporter construct and a mutational screen indicated the conserved YEATS domain as essential for this function. Overlay and pulldown-assays finally showed a specific and YEATS domain-dependent association of ENL with histones H3 and H1. In summary, our studies support a common role for nuclear MLL fusion partners in chromatin biology.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , Protein Biosynthesis/physiology , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism , Animals , Chromatin/chemistry , DNA-Binding Proteins/chemistry , Histone-Lysine N-Methyltransferase , Histones/chemistry , Histones/metabolism , Humans , Myeloid-Lymphoid Leukemia Protein , Proto-Oncogenes , Recombinant Fusion Proteins/chemistry , Structure-Activity Relationship , Transcription Factors/chemistry , Transcriptional Activation , Two-Hybrid System Techniques
16.
Mol Cell Biol ; 24(2): 617-28, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14701735

ABSTRACT

MLL fusion proteins are oncogenic transcription factors that are associated with aggressive lymphoid and myeloid leukemias. We constructed an inducible MLL fusion, MLL-ENL-ERtm, that rendered the transcriptional and transforming properties of MLL-ENL strictly dependent on the presence of 4-hydroxy-tamoxifen. MLL-ENL-ERtm-immortalized hematopoietic cells required 4-hydroxy-tamoxifen for continuous growth and differentiated terminally upon tamoxifen withdrawal. Microarray analysis performed on these conditionally transformed cells revealed Hoxa9 and Hoxa7 as well as the Hox coregulators Meis1 and Pbx3 among the targets upregulated by MLL-ENL-ERtm. Overexpression of the Hox repressor Bmi-1 inhibited the growth-transforming activity of MLL-ENL. Moreover, the enforced expression of Hoxa9 in combination with Meis1 was sufficient to substitute for MLL-ENL-ERtm function and to maintain a state of continuous proliferation and differentiation arrest. These results suggest that MLL fusion proteins impose a reversible block on myeloid differentiation through aberrant activation of a limited set of homeobox genes and Hox coregulators that are consistently expressed in MLL-associated leukemias.


Subject(s)
Cell Transformation, Neoplastic/genetics , Homeodomain Proteins/genetics , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/metabolism , Tamoxifen/analogs & derivatives , Animals , Cell Line , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Down-Regulation , Genes, Homeobox , Humans , Leukemia/genetics , Leukemia/metabolism , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Myeloid-Lymphoid Leukemia Protein , Oncogene Proteins, Fusion/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Tamoxifen/pharmacology , Up-Regulation
17.
Nucleic Acids Res ; 30(4): 958-65, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11842107

ABSTRACT

Alterations of the proto-oncogene MLL (mixed lineage leukemia) are characteristic for a high proportion of acute leukemias, especially those occurring in infants. The activation of MLL is achieved either by an internal tandem duplication of 5' MLL exons or by chromosomal translocations that create chimeric proteins with the N-terminus of MLL fused to a variety of different partner proteins. A domain of MLL with significant homology to the eukaryotic DNA methyltransferases (MT domain) has been found to be essential for the transforming potential of the oncogenic MLL derivatives. Here we demonstrate that this domain specifically recognizes DNA with unmethylated CpG sequences. In gel mobility shifts, the presence of CpG was sufficient for binding of recombinant GST-MT protein to DNA. The introduction of 5-methylCpG on one or both DNA strands precluded an efficient interaction. In surface plasmon resonance a KD of approximately 3.3 x 10(-8) M was determined for the GST-MT/DNA complex formation. Site selection experiments and DNase I footprinting confirmed CpG as the target of the MT domain. Finally, this interaction was corroborated in vivo in reporter assays utilizing the DNA-binding properties of the MT domain in a hybrid MT-VP16 transactivator construct.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Oligodeoxyribonucleotides/metabolism , Proto-Oncogenes , Transcription Factors , Amino Acid Sequence , Base Sequence , Binding Sites , DNA Footprinting , DNA Methylation , DNA Modification Methylases/chemistry , DNA-Binding Proteins/genetics , Deoxyribonuclease I/chemistry , Electrophoretic Mobility Shift Assay , Glutathione Transferase/genetics , Histone-Lysine N-Methyltransferase , Humans , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein , Protein Structure, Tertiary , Proto-Oncogene Mas , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Surface Plasmon Resonance , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...