Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Hemasphere ; 8(6): e83, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38836099
2.
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
3.
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
4.
Life Sci Alliance ; 4(1)2021 01.
Article in English | MEDLINE | ID: mdl-33144337

ABSTRACT

Chromosomal rearrangements of the mixed-lineage leukemia gene MLL1 are the hallmark of infant acute leukemia. The granulocyte-macrophage progenitor state forms the epigenetic basis for myelomonocytic leukemia stemness and transformation by MLL-type oncoproteins. Previously, it was shown that the establishment of murine myelomonocytic MLL-ENL transformation, but not its maintenance, depends on the transcription factor C/EBPα, suggesting an epigenetic hit-and-run mechanism of MLL-driven oncogenesis. Here, we demonstrate that compound deletion of Cebpa/Cebpb almost entirely abrogated the growth and survival of MLL-ENL-transformed cells. Rare, slow-growing, and apoptosis-prone MLL-ENL-transformed escapees were recovered from compound Cebpa/Cebpb deletions. The escapees were uniformly characterized by high expression of the resident Cebpe gene, suggesting inferior functional compensation of C/EBPα/C/EBPß deficiency by C/EBPε. Complementation was augmented by ectopic C/EBPß expression and downstream activation of IGF1 that enhanced growth. Cebpe gene inactivation was accomplished only in the presence of complementing C/EBPß, but not in its absence, confirming the Cebpe dependency of the Cebpa/Cebpb double knockouts. Our data show that MLL-transformed myeloid cells are dependent on C/EBPs during the initiation and maintenance of transformation.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/deficiency , CCAAT-Enhancer-Binding Protein-beta/deficiency , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Granulocyte Precursor Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Animals , Apoptosis/genetics , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Gene Knockout Techniques , HEK293 Cells , Humans , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Signal Transduction/genetics , Transfection
5.
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
6.
Biochim Biophys Acta Gene Regul Mech ; 1863(3): 194503, 2020 03.
Article in English | MEDLINE | ID: mdl-32061883

ABSTRACT

The highly leukemogenic MLL fusion proteins have a unique mechanism of action. This review summarizes the current knowledge of how MLL fusions interact with the transcriptional machinery and it proposes a hypothesis how these proteins modify transcriptional control to act as transcriptional amplifiers causing runaway production of certain RNAs that transform hematopoietic cells.


Subject(s)
Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Transcription, Genetic , Chromatin/metabolism , Transcriptional Activation
7.
Haematologica ; 105(5): 1294-1305, 2020 05.
Article in English | MEDLINE | ID: mdl-31413090

ABSTRACT

Mixed lineage leukemia (MLL/KMT2A) rearrangements (MLL-r) are one of the most frequent chromosomal aberrations in acute myeloid leukemia. We evaluated the function of Meningioma 1 (MN1), a co-factor of HOXA9 and MEIS1, in human and murine MLL-rearranged leukemia by CRISPR-Cas9 mediated deletion of MN1. MN1 was required for in vivo leukemogenicity of MLL positive murine and human leukemia cells. Loss of MN1 inhibited cell cycle and proliferation, promoted apoptosis and induced differentiation of MLL-rearranged cells. Expression analysis and chromatin immunoprecipitation with sequencing from previously reported data sets demonstrated that MN1 primarily maintains active transcription of HOXA9 and HOXA10, which are critical downstream genes of MLL, and their target genes like BCL2, MCL1 and Survivin. Treatment of MLL-rearranged primary leukemia cells with anti-MN1 siRNA significantly reduced their clonogenic potential in contrast to normal CD34+ hematopoietic progenitor cells, suggesting a therapeutic window for MN1 targeting. In summary, our findings demonstrate that MN1 plays an essential role in MLL fusion leukemias and serve as a therapeutic target in MLL-rearranged acute myeloid leukemia.


Subject(s)
Leukemia, Myeloid, Acute , Trans-Activators/genetics , Tumor Suppressor Proteins/genetics , Animals , Hematopoietic Stem Cells , Humans , Leukemia, Myeloid, Acute/genetics , Mice
8.
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
9.
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
10.
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
11.
PLoS One ; 10(6): e0130495, 2015.
Article in English | MEDLINE | ID: mdl-26098938

ABSTRACT

The oncogenic transcription factor E2A-PBX1 is expressed consequent to chromosomal translocation 1;19 and is an important oncogenic driver in cases of pre-B-cell acute lymphoblastic leukemia (ALL). Elucidating the mechanism by which E2A-PBX1 induces lymphoid leukemia would be expedited by the availability of a tractable experimental model in which enforced expression of E2A-PBX1 in hematopoietic progenitors induces pre-B-cell ALL. However, hematopoietic reconstitution of irradiated mice with bone marrow infected with E2A-PBX1-expressing retroviruses consistently gives rise to myeloid, not lymphoid, leukemia. Here, we elucidate the hematopoietic consequences of forced E2A-PBX1 expression in primary murine hematopoietic progenitors. We show that introducing E2A-PBX1 into multipotent progenitors permits the retention of myeloid potential but imposes a dense barrier to lymphoid development prior to the common lymphoid progenitor stage, thus helping to explain the eventual development of myeloid, and not lymphoid, leukemia in transplanted mice. Our findings also indicate that E2A-PBX1 enforces the aberrant, persistent expression of some genes that would normally have been down-regulated in the subsequent course of hematopoietic maturation. We show that enforced expression of one such gene, Hoxa9, a proto-oncogene associated with myeloid leukemia, partially reproduces the phenotype produced by E2A-PBX1 itself. Existing evidence suggests that the 1;19 translocation event takes place in committed B-lymphoid progenitors. However, we find that retrovirus-enforced expression of E2A-PBX1 in committed pro-B-cells results in cell cycle arrest and apoptosis. Our findings indicate that the neoplastic phenotype induced by E2A-PBX1 is determined by the developmental stage of the cell into which the oncoprotein is introduced.


Subject(s)
Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Lymphopoiesis , Myelopoiesis , Oncogene Proteins, Fusion/metabolism , Animals , Apoptosis , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Humans , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred BALB C , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Mas , Retroviridae/genetics
12.
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
13.
Cancer Cell ; 26(6): 781-782, 2014 Dec 08.
Article in English | MEDLINE | ID: mdl-25490442

ABSTRACT

Epigenetic control mechanisms are central to normal and malignant hematopoiesis. In this issue of Cancer Cell, Deshpande and colleagues demonstrate that AF10, an interaction partner of the histone methyltransferase DOT1L, is essential for efficient H3K79 methylation, thus regulating HOX-gene transcription and transformation in myeloid leukemia.


Subject(s)
Histones/metabolism , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Methyltransferases/metabolism , Transcription Factors/metabolism , Animals , Humans
14.
PLoS One ; 9(8): e104419, 2014.
Article in English | MEDLINE | ID: mdl-25137060

ABSTRACT

Here we present a novel method "Genomic inverse PCR for exploration of ligated breakpoints" (GIPFEL) that allows the sensitive detection of recurrent chromosomal translocations. This technique utilizes limited amounts of DNA as starting material and relies on PCR based quantification of unique DNA sequences that are created by circular ligation of restricted genomic DNA from translocation bearing cells. Because the complete potential breakpoint region is interrogated, a prior knowledge of the individual, specific interchromosomal fusion site is not required. We validated GIPFEL for the five most common gene fusions associated with childhood leukemia (MLL-AF4, MLL-AF9, MLL-ENL, ETV6-RUNX1, and TCF3-PBX1). A workflow of restriction digest, purification, ligation, removal of linear fragments and precipitation enriching for circular DNA was developed. GIPFEL allowed detection of translocation specific signature sequences down to a 10-4 dilution which is close to the theoretical limit. In a blinded proof-of-principle study utilizing DNA from cell lines and 144 children with B-precursor-ALL associated translocations this method was 100% specific with no false positive results. Sensitivity was 83%, 65%, and 24% for t(4;11), t(9;11) and t(11;19) respectively. Translocation t(12;21) was correctly detected in 64% and t(1;19) in 39% of the cases. In contrast to other methods, the characteristics of GIPFEL make it particularly attractive for prospective studies.


Subject(s)
Chromosome Breakpoints , DNA, Circular/genetics , Polymerase Chain Reaction/methods , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Translocation, Genetic , Child , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 4 , Chromosomes, Human, Pair 9 , Core Binding Factor Alpha 2 Subunit/genetics , DNA, Circular/chemistry , Humans , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Sensitivity and Specificity
15.
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
16.
Mol Cell Biol ; 33(7): 1331-44, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23339870

ABSTRACT

CD83 is one of the best-known surface markers for fully mature dendritic cells (mature DCs), and its cell-type- and maturation-specific regulation makes the CD83 promoter an interesting tool for the genetic modulation of DCs. To determine the mechanisms regulating this DC- and maturation-specific CD83 expression, chromatin immunoprecipitation (ChIP)-on-chip microarray, biocomputational, reporter, electrophoretic mobility shift assay (EMSA), and ChIP analyses were performed. These studies led to the identification of a ternary transcriptional activation complex composed of an upstream regulatory element, a minimal promoter, and an enhancer, which have not been reported in this arrangement for any other gene so far. Notably, these DNA regions contain a complex framework of interferon regulatory factor (IRF)- and NF-κB transcription factor-binding sites mediating their arrangement. Mutation of any of the IRF-binding sites resulted in a significant loss of promoter activity, whereas overexpression of NF-κB transcription factors clearly enhanced transcription. We identified IRF-1, IRF-2, IRF-5, p50, p65, and cRel to be involved in regulating maturation-specific CD83 expression in DCs. Therefore, the characterization of this promoter complex not only contributes to the knowledge of DC-specific gene regulation but also suggests the involvement of a transcriptional module with binding sites separated into distinct regions in transcriptional activation as well as cell-type- and maturation-specific transcriptional targeting of DCs.


Subject(s)
Antigens, CD/genetics , Dendritic Cells/metabolism , Immunoglobulins/genetics , Interferon Regulatory Factors/genetics , Membrane Glycoproteins/genetics , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , Transcriptional Activation/genetics , Animals , Antigens, CD/metabolism , Binding Sites , Enhancer Elements, Genetic/genetics , HEK293 Cells , HeLa Cells , Humans , Immunoglobulins/metabolism , Interferon Regulatory Factors/metabolism , Introns , Membrane Glycoproteins/metabolism , Mice , NF-kappa B/metabolism , NIH 3T3 Cells , Transcription Factors/genetics , Transcription Factors/metabolism , CD83 Antigen
17.
Proc Natl Acad Sci U S A ; 109(47): 19397-402, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23132946

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous group of hematopoietic malignancies with variable response to treatment. AMLs bearing MLL (mixed lineage leukemia) rearrangements are associated with intermediate or poor survival. MicroRNAs (miRNAs), a class of small noncoding RNAs, have been postulated to be important gene expression regulators virtually in all biological processes, including leukemogenesis. Through a large-scale, genome-wide miRNA expression profiling assay of 85 human AML and 15 normal control samples, we show that among 48 miRNAs that are significantly differentially expressed between MLL- and non-MLL-rearranged AML samples, only one (miR-495) is expressed at a lower level in MLL-rearranged AML than in non-MLL-rearranged AML; meanwhile, miR-495 is also significantly down-regulated in MLL-rearranged AML samples compared with normal control samples. Through in vitro colony-forming/replating assays and in vivo bone marrow transplantation studies, we show that forced expression of miR-495 significantly inhibits MLL-fusion-mediated cell transformation in vitro and leukemogenesis in vivo. In human leukemic cells carrying MLL rearrangements, ectopic expression of miR-495 greatly inhibits cell viability and increases cell apoptosis. Furthermore, our studies demonstrate that PBX3 and MEIS1 are two direct target genes of miR-495, and forced expression of either of them can reverse the effects of miR-495 overexpression on inhibiting cell viability and promoting apoptosis of human MLL-rearranged leukemic cells. Thus, our data indicate that miR-495 likely functions as a tumor suppressor in AML with MLL rearrangements by targeting essential leukemia-related genes.


Subject(s)
Down-Regulation/genetics , Gene Rearrangement/genetics , Leukemia, Myeloid, Acute/genetics , MicroRNAs/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , Base Sequence , Case-Control Studies , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genes, Neoplasm/genetics , Genetic Association Studies , Homeodomain Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , Mice , MicroRNAs/genetics , Molecular Sequence Data , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins/metabolism
18.
Cancer Cell ; 22(4): 524-35, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23079661

ABSTRACT

Expression of microRNAs (miRNAs) is under stringent regulation at both transcriptional and posttranscriptional levels. Disturbance at either level could cause dysregulation of miRNAs. Here, we show that MLL fusion proteins negatively regulate production of miR-150, an miRNA widely repressed in acute leukemia, by blocking miR-150 precursors from being processed to mature miRNAs through MYC/LIN28 functional axis. Forced expression of miR-150 dramatically inhibited leukemic cell growth and delayed MLL-fusion-mediated leukemogenesis, likely through targeting FLT3 and MYB and thereby interfering with the HOXA9/MEIS1/FLT3/MYB signaling network, which in turn caused downregulation of MYC/LIN28. Collectively, we revealed a MLL-fusion/MYC/LIN28⊣miR-150⊣FLT3/MYB/HOXA9/MEIS1 signaling circuit underlying the pathogenesis of leukemia, where miR-150 functions as a pivotal gatekeeper and its repression is required for leukemogenesis.


Subject(s)
Leukemia/etiology , MicroRNAs/physiology , Myeloid-Lymphoid Leukemia Protein/physiology , Proto-Oncogene Proteins c-myc/physiology , RNA-Binding Proteins/physiology , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA Methylation , Down-Regulation , Gene Dosage , Gene Expression Regulation, Leukemic , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/physiology , Humans , Mice , MicroRNAs/analysis , MicroRNAs/antagonists & inhibitors , Mutation , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/physiology , Nuclear Proteins/genetics , Signal Transduction , fms-Like Tyrosine Kinase 3/genetics
19.
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
20.
Exp Hematol ; 39(1): 77-86.e1-5, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20854876

ABSTRACT

OBJECTIVE: The aim of this study was to better understand how mixed lineage leukemia (MLL) fusion proteins deregulate the expression of genes critical for leukemia. MATERIALS AND METHODS: The transforming domain of one of the most common MLL fusion partners, AF9, was immunopurified after expression in myeloblastic M1 cells, and associating proteins were identified by mass spectrometric analysis. Chromatin immunoprecipitation followed by quantitative polymerase chain reaction was used to determine how binding of associating proteins compare across Hoxa9 and Meis1 in cell lines with and without MLL fusion proteins and how binding is altered during gene down-regulation and differentiation. RESULTS: Consistent with earlier purifications of ENL and AF4 from 293 cells, the 90 amino acid C-terminal domain of AF9 associates with many other MLL translocation partners including Enl, Af4, Laf4, Af5q31, Ell, and Af10. This complex, termed elongation assisting proteins (EAPs), also contains the RNA polymerase II C-terminal domain kinase Cdk9/Cyclin T1/T2 (pTEFb) and the histone H3 lysine 79 methyltransferase Dot1L. Myeloid cells transformed by MLL fusions show higher levels and a broader distribution of EAP components at genes critical for leukemia. Inhibition of EAP components pTEFb and Dot1l show that both contribute significantly to activation of Hoxa9 and Meis1 expression. EAP is dynamically associated with the Hoxa9 and Meis1 loci in hematopoietic cells and rapidly dissociates during induction of differentiation. In the presence of MLL fusion proteins, its dissociation is prevented. CONCLUSIONS: The findings suggest that MLL fusion proteins deregulate genes critical for leukemia by excessive recruitment and impaired dissociation of EAP from target loci.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Leukemia/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Transcription, Genetic/genetics , Blotting, Western , Cell Line , Chromatography, Liquid , Gene Knockdown Techniques , Humans , Immunoprecipitation , Polymerase Chain Reaction , RNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...