Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Blood ; 139(7): 1080-1097, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34695195

ABSTRACT

In an effort to identify novel drugs targeting fusion-oncogene-induced acute myeloid leukemia (AML), we performed high-resolution proteomic analysis. In AML1-ETO (AE)-driven AML, we uncovered a deregulation of phospholipase C (PLC) signaling. We identified PLCgamma 1 (PLCG1) as a specific target of the AE fusion protein that is induced after AE binding to intergenic regulatory DNA elements. Genetic inactivation of PLCG1 in murine and human AML inhibited AML1-ETO dependent self-renewal programs, leukemic proliferation, and leukemia maintenance in vivo. In contrast, PLCG1 was dispensable for normal hematopoietic stem and progenitor cell function. These findings are extended to and confirmed by pharmacologic perturbation of Ca++-signaling in AML1-ETO AML cells, indicating that the PLCG1 pathway poses an important therapeutic target for AML1-ETO+ leukemic stem cells.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/pathology , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/pathology , Oncogene Proteins, Fusion/metabolism , Phospholipase C gamma/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , Animals , Cell Self Renewal , Core Binding Factor Alpha 2 Subunit/genetics , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Phospholipase C gamma/genetics , Proteome , RUNX1 Translocation Partner 1 Protein/genetics , Transcriptome , Translocation, Genetic
2.
Blood ; 140(17): 1875-1890, 2022 10 27.
Article in English | MEDLINE | ID: mdl-35839448

ABSTRACT

The fusion gene MLL/AF4 defines a high-risk subtype of pro-B acute lymphoblastic leukemia. Relapse can be associated with a lineage switch from acute lymphoblastic to acute myeloid leukemia, resulting in poor clinical outcomes caused by resistance to chemotherapies and immunotherapies. In this study, the myeloid relapses shared oncogene fusion breakpoints with their matched lymphoid presentations and originated from various differentiation stages from immature progenitors through to committed B-cell precursors. Lineage switching is linked to substantial changes in chromatin accessibility and rewiring of transcriptional programs, including alternative splicing. These findings indicate that the execution and maintenance of lymphoid lineage differentiation is impaired. The relapsed myeloid phenotype is recurrently associated with the altered expression, splicing, or mutation of chromatin modifiers, including CHD4 coding for the ATPase/helicase of the nucleosome remodelling and deacetylation complex. Perturbation of CHD4 alone or in combination with other mutated epigenetic modifiers induces myeloid gene expression in MLL/AF4+ cell models, indicating that lineage switching in MLL/AF4 leukemia is driven and maintained by disrupted epigenetic regulation.


Subject(s)
Myeloid-Lymphoid Leukemia Protein , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Epigenesis, Genetic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Genes, Regulator , Chromatin
3.
PLoS Comput Biol ; 15(11): e1007337, 2019 11.
Article in English | MEDLINE | ID: mdl-31682597

ABSTRACT

Gene expression governs cell fate, and is regulated via a complex interplay of transcription factors and molecules that change chromatin structure. Advances in sequencing-based assays have enabled investigation of these processes genome-wide, leading to large datasets that combine information on the dynamics of gene expression, transcription factor binding and chromatin structure as cells differentiate. While numerous studies focus on the effects of these features on broader gene regulation, less work has been done on the mechanisms of gene-specific transcriptional control. In this study, we have focussed on the latter by integrating gene expression data for the in vitro differentiation of murine ES cells to macrophages and cardiomyocytes, with dynamic data on chromatin structure, epigenetics and transcription factor binding. Combining a novel strategy to identify communities of related control elements with a penalized regression approach, we developed individual models to identify the potential control elements predictive of the expression of each gene. Our models were compared to an existing method and evaluated using the existing literature and new experimental data from embryonic stem cell differentiation reporter assays. Our method is able to identify transcriptional control elements in a gene specific manner that reflect known regulatory relationships and to generate useful hypotheses for further testing.


Subject(s)
Cell Differentiation/genetics , High-Throughput Screening Assays/methods , Regulatory Elements, Transcriptional/genetics , Animals , Cell Differentiation/physiology , Chromatin/metabolism , Databases, Genetic , Epigenesis, Genetic , Epigenomics , Gene Expression Regulation/genetics , Genome , Macrophages/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism
4.
Development ; 143(23): 4324-4340, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27802171

ABSTRACT

The transmission of extracellular signals into the nucleus involves inducible transcription factors, but how different signalling pathways act in a cell type-specific fashion is poorly understood. Here, we studied the regulatory role of the AP-1 transcription factor family in blood development using embryonic stem cell differentiation coupled with genome-wide transcription factor binding and gene expression analyses. AP-1 factors respond to MAP kinase signalling and comprise dimers of FOS, ATF and JUN proteins. To examine genes regulated by AP-1 and to examine how it interacts with other inducible transcription factors, we abrogated its global DNA-binding activity using a dominant-negative FOS peptide. We show that FOS and JUN bind to and activate a specific set of vascular genes and that AP-1 inhibition shifts the balance between smooth muscle and hematopoietic differentiation towards blood. Furthermore, AP-1 is required for de novo binding of TEAD4, a transcription factor connected to Hippo signalling. Our bottom-up approach demonstrates that AP-1- and TEAD4-associated cis-regulatory elements form hubs for multiple signalling-responsive transcription factors and define the cistrome that regulates vascular and hematopoietic development by extrinsic signals.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Muscle Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Activating Transcription Factors/metabolism , Animals , Binding Sites/genetics , Cell Line , DNA-Binding Proteins/genetics , Gene Expression/genetics , Gene Expression Profiling , Mice , Muscle, Smooth, Vascular/metabolism , Protein Binding , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/physiology , TEA Domain Transcription Factors , Transcription Factor AP-1/antagonists & inhibitors
5.
Blood ; 130(10): 1213-1222, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28710059

ABSTRACT

Understanding and blocking the self-renewal pathway of preleukemia stem cells could prevent acute myeloid leukemia (AML) relapse. In this study, we show that increased FOXO1 represents a critical mechanism driving aberrant self-renewal in preleukemic cells expressing the t(8;21)-associated oncogene AML1-ETO (AE). Although generally considered as a tumor suppressor, FOXO1 is consistently upregulated in t(8;21) AML. Expression of FOXO1 in human CD34+ cells promotes a preleukemic state with enhanced self-renewal and dysregulated differentiation. The DNA binding domain of FOXO1 is essential for these functions. FOXO1 activates a stem cell molecular signature that is also present in AE preleukemia cells and preserved in t(8;21) patient samples. Genome-wide binding studies show that AE and FOXO1 share the majority of their binding sites, whereby FOXO1 binds to multiple crucial self-renewal genes and is required for their activation. In agreement with this observation, genetic and pharmacological ablation of FOXO1 inhibited the long-term proliferation and clonogenicity of AE cells and t(8;21) AML cell lines. Targeting of FOXO1 therefore provides a potential therapeutic strategy for elimination of stem cells at both preleukemic and leukemic stages.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Forkhead Box Protein O1/metabolism , Gene Regulatory Networks , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/metabolism , Precancerous Conditions/genetics , Animals , Antigens, CD34/metabolism , Cell Line, Tumor , Cell Proliferation , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genome, Human , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , Mice, SCID , Oncogene Proteins, Fusion/genetics , Precancerous Conditions/pathology , RUNX1 Translocation Partner 1 Protein , Up-Regulation/genetics
6.
Nucleic Acids Res ; 45(17): 9874-9888, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28973433

ABSTRACT

LMO2 is a bridging factor within a DNA binding complex and is required for definitive haematopoiesis to occur. The developmental stage of the block in haematopoietic specification is not known. We show that Lmo2-/- mouse embryonic stem cells differentiated to Flk-1+ haemangioblasts, but less efficiently to haemogenic endothelium, which only produced primitive haematopoietic progenitors. Genome-wide approaches indicated that LMO2 is required at the haemangioblast stage to position the TAL1/LMO2/LDB1 complex to regulatory elements that are important for the establishment of the haematopoietic developmental program. In the absence of LMO2, the target site recognition of TAL1 is impaired. The lack of LMO2 resulted in altered gene expression levels already at the haemangioblast stage, with transcription factor genes accounting for ∼15% of affected genes. Comparison of Lmo2-/- with Tal1-/- Flk-1+ cells further showed that TAL1 was required to initiate or sustain Lmo2 expression.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA-Binding Proteins/genetics , DNA/genetics , Genome , Hemangioblasts/metabolism , LIM Domain Proteins/genetics , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins/genetics , Adaptor Proteins, Signal Transducing/deficiency , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/deficiency , Cell Differentiation , Cell Line , DNA/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Hemangioblasts/cytology , Hematopoiesis/genetics , LIM Domain Proteins/deficiency , LIM Domain Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Protein Binding , Proto-Oncogene Proteins/deficiency , Regulatory Elements, Transcriptional , Signal Transduction , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription, Genetic , Vascular Endothelial Growth Factor Receptor-2/deficiency , Vascular Endothelial Growth Factor Receptor-2/genetics
7.
Development ; 141(12): 2391-401, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24850855

ABSTRACT

Mammalian development is regulated by the interplay of tissue-specific and ubiquitously expressed transcription factors, such as Sp1. Sp1 knockout mice die in utero with multiple phenotypic aberrations, but the underlying molecular mechanism of this differentiation failure has been elusive. Here, we have used conditional knockout mice as well as the differentiation of mouse ES cells as a model with which to address this issue. To this end, we examined differentiation potential, global gene expression patterns and Sp1 target regions in Sp1 wild-type and Sp1-deficient cells representing different stages of hematopoiesis. Sp1(-/-) cells progress through most embryonic stages of blood cell development but cannot complete terminal differentiation. This failure to fully differentiate is not seen when Sp1 is knocked out at later developmental stages. For most Sp1 target and non-target genes, gene expression is unaffected by Sp1 inactivation. However, Cdx genes and multiple Hox genes are stage-specific targets of Sp1 and are downregulated at an early stage. As a consequence, expression of genes involved in hematopoietic specification is progressively deregulated. Our work demonstrates that the early absence of active Sp1 sets a cascade in motion that culminates in a failure of terminal hematopoietic differentiation and emphasizes the role of ubiquitously expressed transcription factors for tissue-specific gene regulation. In addition, our global side-by-side analysis of the response of the transcriptional network to perturbation sheds a new light on the regulatory hierarchy of hematopoietic specification.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/cytology , Sp1 Transcription Factor/physiology , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cell Lineage , Embryonic Stem Cells/cytology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Macrophages/cytology , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phenotype , Protein Binding , Stem Cells/cytology
8.
EMBO J ; 31(22): 4318-33, 2012 Nov 14.
Article in English | MEDLINE | ID: mdl-23064151

ABSTRACT

Cell fate decisions during haematopoiesis are governed by lineage-specific transcription factors, such as RUNX1, SCL/TAL1, FLI1 and C/EBP family members. To gain insight into how these transcription factors regulate the activation of haematopoietic genes during embryonic development, we measured the genome-wide dynamics of transcription factor assembly on their target genes during the RUNX1-dependent transition from haemogenic endothelium (HE) to haematopoietic progenitors. Using a Runx1-/- embryonic stem cell differentiation model expressing an inducible Runx1 gene, we show that in the absence of RUNX1, haematopoietic genes bind SCL/TAL1, FLI1 and C/EBPß and that this early priming is required for correct temporal expression of the myeloid master regulator PU.1 and its downstream targets. After induction, RUNX1 binds to numerous de novo sites, initiating a local increase in histone acetylation and rapid global alterations in the binding patterns of SCL/TAL1 and FLI1. The acquisition of haematopoietic fate controlled by Runx1 therefore does not represent the establishment of a new regulatory layer on top of a pre-existing HE program but instead entails global reorganization of lineage-specific transcription factor assemblies.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Epigenesis, Genetic/physiology , Hematopoiesis/physiology , Acetylation , Animals , Base Sequence , Cell Line , Core Binding Factor Alpha 2 Subunit/genetics , Embryonic Stem Cells/physiology , Epigenesis, Genetic/genetics , Hematopoiesis/genetics , Histones/metabolism , Mice , Molecular Sequence Data , Protein Binding , Transcription Factors/physiology
9.
BMC Genomics ; 16: 1000, 2015 Nov 25.
Article in English | MEDLINE | ID: mdl-26608661

ABSTRACT

BACKGROUND: The analysis of differential gene expression is a fundamental tool to relate gene regulation with specific biological processes. Differential binding of transcription factors (TFs) can drive differential gene expression. While DNase-seq data can provide global snapshots of TF binding, tools for detecting differential binding from pairs of DNase-seq data sets are lacking. RESULTS: In order to link expression changes with changes in TF binding we introduce the concept of differential footprinting alongside a computational tool. We demonstrate that differential footprinting is associated with differential gene expression and can be used to define cell types by their specific TF occupancy patterns. CONCLUSIONS: Our new tool, Wellington-bootstrap, will enable the detection of differential TF binding facilitating the study of gene regulatory systems.


Subject(s)
Binding Sites , Computational Biology/methods , DNA Footprinting , Deoxyribonucleases/metabolism , High-Throughput Nucleotide Sequencing , Transcription Factors/metabolism , Antigens, CD19/metabolism , B-Lymphocyte Subsets/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cluster Analysis , DNA Footprinting/methods , Gene Expression Regulation , Humans , Organ Specificity/genetics , Protein Binding
10.
Nat Commun ; 15(1): 1359, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38355578

ABSTRACT

Acute Myeloid Leukemia (AML) is caused by multiple mutations which dysregulate growth and differentiation of myeloid cells. Cells adopt different gene regulatory networks specific to individual mutations, maintaining a rapidly proliferating blast cell population with fatal consequences for the patient if not treated. The most common treatment option is still chemotherapy which targets such cells. However, patients harbour a population of quiescent leukemic stem cells (LSCs) which can emerge from quiescence to trigger relapse after therapy. The processes that allow such cells to re-grow remain unknown. Here, we examine the well characterised t(8;21) AML sub-type as a model to address this question. Using four primary AML samples and a novel t(8;21) patient-derived xenograft model, we show that t(8;21) LSCs aberrantly activate the VEGF and IL-5 signalling pathways. Both pathways operate within a regulatory circuit consisting of the driver oncoprotein RUNX1::ETO and an AP-1/GATA2 axis allowing LSCs to re-enter the cell cycle while preserving self-renewal capacity.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mutation , Stem Cells/metabolism , Neoplastic Stem Cells/metabolism
11.
bioRxiv ; 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37503022

ABSTRACT

AML is a heterogenous disease caused by different mutations. We have previously shown that each mutational sub-type develops its specific gene regulatory network (GRN) with transcription factors interacting with multiple gene modules, many of which are transcription factor genes themselves. Here we hypothesized that highly connected nodes within such networks comprise crucial regulators of AML maintenance. We tested this hypothesis using FLT3-ITD mutated AML as a model and conducted an shRNA drop-out screen informed by this analysis. We show that AML-specific GRNs predict identifying crucial regulatory modules required for AML but not normal cellular growth. Furthermore, our work shows that all modules are highly connected and regulate each other. The careful multi-omic analysis of the role of one (RUNX1) module by shRNA and chemical inhibition shows that this transcription factor and its target genes stabilize the GRN of FLT3-ITD AML and that its removal leads to GRN collapse and cell death.

12.
Leukemia ; 37(1): 102-112, 2023 01.
Article in English | MEDLINE | ID: mdl-36333583

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy caused by mutations in genes encoding transcriptional and epigenetic regulators together with signaling genes. It is characterized by a disturbance of differentiation and abnormal proliferation of hematopoietic progenitors. We have previously shown that each AML subtype establishes its own core gene regulatory network (GRN), consisting of transcription factors binding to their target genes and imposing a specific gene expression pattern that is required for AML maintenance. In this study, we integrate gene expression, open chromatin and ChIP data with promoter-capture Hi-C data to define a refined core GRN common to all patients with CEBPA-double mutant (CEBPAN/C) AML. These mutations disrupt the structure of a major regulator of myelopoiesis. We identify the binding sites of mutated C/EBPα proteins in primary cells, we show that C/EBPα, AP-1 factors and RUNX1 colocalize and are required for AML maintenance, and we employ single cell experiments to link important network nodes to the specific differentiation trajectory from leukemic stem to blast cells. Taken together, our study provides an important resource which predicts the specific therapeutic vulnerabilities of this AML subtype in human cells.


Subject(s)
Gene Regulatory Networks , Leukemia, Myeloid, Acute , Humans , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Mutation , Cell Differentiation/genetics , Leukemia, Myeloid, Acute/pathology
13.
Cell Rep ; 42(12): 113568, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38104314

ABSTRACT

Acute myeloid leukemia (AML) is a heterogeneous disease caused by different mutations. Previously, we showed that each mutational subtype develops its specific gene regulatory network (GRN) with transcription factors interacting within multiple gene modules, many of which are transcription factor genes themselves. Here, we hypothesize that highly connected nodes within such networks comprise crucial regulators of AML maintenance. We test this hypothesis using FLT3-ITD-mutated AML as a model and conduct an shRNA drop-out screen informed by this analysis. We show that AML-specific GRNs predict crucial regulatory modules required for AML growth. Furthermore, our work shows that all modules are highly connected and regulate each other. The careful multi-omic analysis of the role of one (RUNX1) module by shRNA and chemical inhibition shows that this transcription factor and its target genes stabilize the GRN of FLT3-ITD+ AML and that its removal leads to GRN collapse and cell death.


Subject(s)
Gene Regulatory Networks , Leukemia, Myeloid, Acute , Humans , Regulon , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mutation/genetics , RNA, Small Interfering , fms-Like Tyrosine Kinase 3/genetics
14.
Nat Commun ; 14(1): 6947, 2023 11 07.
Article in English | MEDLINE | ID: mdl-37935654

ABSTRACT

Disease-causing mutations in genes encoding transcription factors (TFs) can affect TF interactions with their cognate DNA-binding motifs. Whether and how TF mutations impact upon the binding to TF composite elements (CE) and the interaction with other TFs is unclear. Here, we report a distinct mechanism of TF alteration in human lymphomas with perturbed B cell identity, in particular classic Hodgkin lymphoma. It is caused by a recurrent somatic missense mutation c.295 T > C (p.Cys99Arg; p.C99R) targeting the center of the DNA-binding domain of Interferon Regulatory Factor 4 (IRF4), a key TF in immune cells. IRF4-C99R fundamentally alters IRF4 DNA-binding, with loss-of-binding to canonical IRF motifs and neomorphic gain-of-binding to canonical and non-canonical IRF CEs. IRF4-C99R thoroughly modifies IRF4 function by blocking IRF4-dependent plasma cell induction, and up-regulates disease-specific genes in a non-canonical Activator Protein-1 (AP-1)-IRF-CE (AICE)-dependent manner. Our data explain how a single mutation causes a complex switch of TF specificity and gene regulation and open the perspective to specifically block the neomorphic DNA-binding activities of a mutant TF.


Subject(s)
Interferon Regulatory Factors , Lymphoma , Humans , B-Lymphocytes/metabolism , DNA , Gene Expression Regulation , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Lymphoma/genetics
15.
Nucleic Acids Res ; 38(6): e86, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20008102

ABSTRACT

Protein-protein interactions (PPIs) are ubiquitous in Biology, and thus offer an enormous potential for the discovery of novel therapeutics. Although protein interfaces are large and lack defining physiochemical traits, is well established that only a small portion of interface residues, the so-called hot spot residues, contribute the most to the binding energy of the protein complex. Moreover, recent successes in development of novel drugs aimed at disrupting PPIs rely on targeting such residues. Experimental methods for describing critical residues are lengthy and costly; therefore, there is a need for computational tools that can complement experimental efforts. Here, we describe a new computational approach to predict hot spot residues in protein interfaces. The method, called Presaging Critical Residues in Protein interfaces (PCRPi), depends on the integration of diverse metrics into a unique probabilistic measure by using Bayesian Networks. We have benchmarked our method using a large set of experimentally verified hot spot residues and on a blind prediction on the protein complex formed by HRAS protein and a single domain antibody. Under both scenarios, PCRPi delivered consistent and accurate predictions. Finally, PCRPi is able to handle cases where some of the input data is either missing or not reliable (e.g. evolutionary information).


Subject(s)
Protein Interaction Mapping/methods , Amino Acids/chemistry , Bayes Theorem , Computational Biology , Immunoglobulin Variable Region/chemistry , Models, Molecular , Protein Conformation , Proteins/chemistry , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/immunology , Software
16.
Cell Rep ; 35(3): 109010, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882316

ABSTRACT

Acute myeloid leukemia (AML) is caused by recurrent mutations in members of the gene regulatory and signaling machinery that control hematopoietic progenitor cell growth and differentiation. Here, we show that the transcription factor WT1 forms a major node in the rewired mutation-specific gene regulatory networks of multiple AML subtypes. WT1 is frequently either mutated or upregulated in AML, and its expression is predictive for relapse. The WT1 protein exists as multiple isoforms. For two main AML subtypes, we demonstrate that these isoforms exhibit differential patterns of binding and support contrasting biological activities, including enhanced proliferation. We also show that WT1 responds to oncogenic signaling and is part of a signaling-responsive transcription factor hub that controls AML growth. WT1 therefore plays a central and widespread role in AML biology.


Subject(s)
Chromatin/chemistry , Core Binding Factor Alpha 2 Subunit/genetics , Gene Regulatory Networks , Leukemia, Myeloid, Acute/genetics , Lung Neoplasms/genetics , WT1 Proteins/genetics , Base Sequence , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chromatin/metabolism , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/classification , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RUNX1 Translocation Partner 1 Protein/genetics , RUNX1 Translocation Partner 1 Protein/metabolism , Signal Transduction , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Translocation, Genetic , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/metabolism , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
17.
Nat Commun ; 12(1): 520, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33483506

ABSTRACT

The fusion oncogene RUNX1/RUNX1T1 encodes an aberrant transcription factor, which plays a key role in the initiation and maintenance of acute myeloid leukemia. Here we show that the RUNX1/RUNX1T1 oncogene is a regulator of alternative RNA splicing in leukemic cells. The comprehensive analysis of RUNX1/RUNX1T1-associated splicing events identifies two principal mechanisms that underlie the differential production of RNA isoforms: (i) RUNX1/RUNX1T1-mediated regulation of alternative transcription start site selection, and (ii) direct or indirect control of the expression of genes encoding splicing factors. The first mechanism leads to the expression of RNA isoforms with alternative structure of the 5'-UTR regions. The second mechanism generates alternative transcripts with new junctions between internal cassettes and constitutive exons. We also show that RUNX1/RUNX1T1-mediated differential splicing affects several functional groups of genes and produces proteins with unique conserved domain structures. In summary, this study reveals alternative splicing as an important component of transcriptome re-organization in leukemia by an aberrant transcriptional regulator.


Subject(s)
Alternative Splicing , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid/genetics , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein/genetics , Acute Disease , Cell Line, Tumor , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Profiling/methods , Humans , Leukemia, Myeloid/pathology , Models, Genetic , Oncogene Proteins, Fusion/metabolism , RNA Interference , RNA Isoforms/genetics , RNA Isoforms/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , Transcription Initiation Site
18.
Exp Hematol ; 92: 62-74, 2020 12.
Article in English | MEDLINE | ID: mdl-33152396

ABSTRACT

Acute myeloid leukemia development occurs in a stepwise fashion whereby an original driver mutation is followed by additional mutations. The first type of mutations tends to be in genes encoding members of the epigenetic/transcription regulatory machinery (i.e., RUNX1, DNMT3A, TET2), while the secondary mutations often involve genes encoding members of signaling pathways that cause uncontrolled growth of such cells such as the growth factor receptors c-KIT of FLT3. Patients usually present with both types of mutations, but it is currently unclear how both mutational events shape the epigenome in developing AML cells. To this end we generated an in vitro model of t(8;21) AML by expressing its driver oncoprotein RUNX1-ETO with or without a mutated (N822K) KIT protein. Expression of N822K-c-KIT strongly increases the self-renewal capacity of RUNX1-ETO-expressing cells. Global analysis of gene expression changes and alterations in the epigenome revealed that N822K-c-KIT expression profoundly influences the open chromatin landscape and transcription factor binding. However, our experiments also revealed that double mutant cells still differ from their patient-derived counterparts, highlighting the importance of studying patient cells to obtain a true picture of how gene regulatory networks have been reprogrammed during tumorigenesis.


Subject(s)
Chromatin/metabolism , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit/metabolism , Leukemia, Myeloid, Acute , Mutation, Missense , Proto-Oncogene Proteins c-kit/metabolism , RUNX1 Translocation Partner 1 Protein/metabolism , Transcription, Genetic , Translocation, Genetic , Amino Acid Substitution , Chromatin/pathology , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 21/metabolism , Chromosomes, Human, Pair 8/genetics , Chromosomes, Human, Pair 8/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Female , Gene Expression Regulation, Leukemic , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Proto-Oncogene Proteins c-kit/genetics , RUNX1 Translocation Partner 1 Protein/genetics
19.
Cancer Inform ; 18: 1176935119859863, 2019.
Article in English | MEDLINE | ID: mdl-31263370

ABSTRACT

Acute myeloid leukemia (AML) is a highly heterogeneous cancer associated with different patterns of gene expression determined by the nature of their DNA mutations. These mutations mostly act to deregulate gene expression by various mechanisms at the level of the nucleus. By performing genome-wide epigenetic profiling of cis-regulatory elements, we found that AML encompasses different mutation-specific subclasses associated with the rewiring of the gene regulatory networks that drive differentiation into different directions away from normal myeloid development. By integrating epigenetic profiles with gene expression and chromatin conformation data, we defined pathways within gene regulation networks that were differentially rewired within each mutation-specific subclass of AML. This analysis revealed 2 major classes of AML: one class defined by mutations in signaling molecules that activate AP-1 via the mitogen-activated protein (MAP) kinase pathway and a second class defined by mutations within genes encoding transcription factors such as RUNX1/CBFß and C/EBPα. By identifying specific DNA motifs protected from DNase I digestion at cis-regulatory elements, we were able to infer candidate transcription factors bound to these motifs. These integrated analyses allowed the identification of AML subtype-specific core regulatory networks that are required for AML development and maintenance, which could now be targeted in personalized therapies.

20.
Leukemia ; 33(6): 1463-1474, 2019 06.
Article in English | MEDLINE | ID: mdl-30546079

ABSTRACT

Long terminal repeat (LTR) elements are wide-spread in the human genome and have the potential to act as promoters and enhancers. Their expression is therefore under tight epigenetic control. We previously reported in classical Hodgkin Lymphoma (cHL) that a member of the THE1B class of LTR elements acted as a promoter for the proto-oncogene and growth factor receptor gene CSF1R and that expression of this gene is required for cHL tumour survival. However, to which extent and how such elements participate in globally shaping the unique cHL gene expression programme is unknown. To address this question we mapped the genome-wide activation of THE1-LTRs in cHL cells using a targeted next generation sequencing approach (RACE-Seq). Integration of these data with global gene expression data from cHL and control B cell lines showed a unique pattern of LTR activation impacting on gene expression, including genes associated with the cHL phenotype. We also show that global LTR activation is induced by strong inflammatory stimuli. Together these results demonstrate that LTR activation provides an additional layer of gene deregulation in classical Hodgkin lymphoma and highlight the potential impact of genome-wide LTR activation in other inflammatory diseases.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Genome, Human , Hodgkin Disease/genetics , Regulatory Sequences, Nucleic Acid , Terminal Repeat Sequences , Transcriptional Activation , Gene Expression Profiling , Hodgkin Disease/pathology , Humans , Promoter Regions, Genetic , Proto-Oncogene Mas , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL