Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Sci Rep ; 12(1): 18656, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333382

ABSTRACT

Advanced computational methods exploit gene expression and epigenetic datasets to predict gene regulatory networks controlled by transcription factors (TFs). These methods have identified cell fate determining TFs but require large amounts of reference data and experimental expertise. Here, we present an easy to use network-based computational framework that exploits enhancers defined by bidirectional transcription, using as sole input CAGE sequencing data to correctly predict TFs key to various human cell types. Next, we applied this Analysis Algorithm for Networks Specified by Enhancers based on CAGE (ANANSE-CAGE) to predict TFs driving red and white blood cell development, and THP-1 leukemia cell immortalization. Further, we predicted TFs that are differentially important to either cell line- or primary- associated MLL-AF9-driven gene programs, and in primary MLL-AF9 acute leukemia. Our approach identified experimentally validated as well as thus far unexplored TFs in these processes. ANANSE-CAGE will be useful to identify transcription factors that are key to any cell fate change using only CAGE-seq data as input.


Subject(s)
Gene Regulatory Networks , Leukemia, Myeloid, Acute , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Leukemia, Myeloid, Acute/genetics , Algorithms , Blood Cells/metabolism , Computational Biology
2.
Ann Hematol ; 101(3): 571-579, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35024892

ABSTRACT

Acute myeloid leukemia (AML) is a highly heterogeneous disease showing dynamic clonal evolution patterns over time. Various subclones may be present simultaneously and subclones may show a different expansion pattern and respond differently to applied therapies. It is already clear that immunophenotyping and genetic analyses may yield overlapping, but also complementary information. Detailed information on the genetic make-up of immunophenotypically defined subclones is however scarce. We performed error-corrected sequencing for 27 myeloid leukemia driver genes in 86, FACS-sorted immunophenotypically characterized normal and aberrant subfractions in 10 AML patients. We identified three main scenarios. In the first group of patients, the two techniques were equally well characterizing the malignancy. In the second group, most of the isolated populations did not express aberrant immunophenotypes but still harbored several genetic aberrancies, indicating that the information obtained only by immunophenotyping would be incomplete. Vice versa, one patient was identified in which genetic mutations were found only in a small fraction of the immunophenotypically defined malignant populations, indicating that the genetic analysis gave an incomplete picture of the disease. We conclude that currently, characterization of leukemic cells in AML by molecular and immunophenotypic techniques is complementary, and infer that both techniques should be used in parallel in order to obtain the most complete view on the disease.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Clonal Evolution , Gene Expression Regulation, Leukemic , Genetic Variation , Humans , Immunophenotyping , Mutation
3.
Blood ; 138(1): 86-90, 2021 07 08.
Article in English | MEDLINE | ID: mdl-33690840

ABSTRACT

Mutations in the transcription factors GATA binding factor 1 (GATA1), growth factor independence 1B (GFI1B), and Runt-related transcription factor 1 (RUNX1) cause familial platelet and bleeding disorders. Mutant platelets exhibit common abnormalities including an α-granule reduction resulting in a grayish appearance in blood smears. This suggests that similar pathways are deregulated by different transcription factor mutations. To identify common factors, full platelet proteomes from 11 individuals with mutant GATA1R216Q, GFI1BQ287*, RUNX1Q154Rfs, or RUNX1TD2-6 and 28 healthy controls were examined by label-free quantitative mass spectrometry. In total, 2875 platelet proteins were reliably quantified. Clustering analysis of more than 300 differentially expressed proteins revealed profound differences between cases and controls. Among cases, 44 of 143 significantly downregulated proteins were assigned to platelet function, hemostasis, and granule biology, in line with platelet dysfunction and bleedings. Remarkably, none of these proteins were significantly diminished in all affected cases. Similarly, no proteins were commonly overrepresented in all affected cases compared with controls. These data indicate that the studied transcription factor mutations alter platelet proteomes in distinct largely nonoverlapping manners. This work provides the quantitative landscape of proteins that affect platelet function when deregulated by mutated transcription factors in inherited bleeding disorders.


Subject(s)
Blood Platelet Disorders/metabolism , Blood Platelets/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , GATA1 Transcription Factor/metabolism , Proteome/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Homeostasis , Humans , Mutation/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Leukemia ; 32(3): 828-836, 2018 03.
Article in English | MEDLINE | ID: mdl-28871137

ABSTRACT

Overexpression of the BRE (brain and reproductive organ-expressed) gene defines a distinct pediatric and adult acute myeloid leukemia (AML) subgroup. Here we identify a promoter enriched for active chromatin marks in BRE intron 4 causing strong biallelic expression of a previously unknown C-terminal BRE transcript. This transcript starts with BRE intron 4 sequences spliced to exon 5 and downstream sequences, and if translated might code for an N terminally truncated BRE protein. Remarkably, the new BRE transcript was highly expressed in over 50% of 11q23/KMT2A (lysine methyl transferase 2A)-rearranged and t(8;16)/KAT6A-CREBBP cases, while it was virtually absent from other AML subsets and normal tissues. In gene reporter assays, the leukemia-specific fusion protein KMT2A-MLLT3 transactivated the intragenic BRE promoter. Further epigenome analyses revealed 97 additional intragenic promoter marks frequently bound by KMT2A in AML with C-terminal BRE expression. The corresponding genes may be part of a context-dependent KMT2A-MLLT3-driven oncogenic program, because they were higher expressed in this AML subtype compared with other groups. C-terminal BRE might be an important contributor to this program because in a case with relapsed AML, we observed an ins(11;2) fusing CHORDC1 to BRE at the region where intragenic transcription starts in KMT2A-rearranged and KAT6A-CREBBP AML.


Subject(s)
Gene Rearrangement , Leukemia, Myeloid, Acute/genetics , Nerve Tissue Proteins/genetics , Protein Interaction Domains and Motifs/genetics , Transcriptional Activation , Translocation, Genetic , Cell Line , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 16 , Epigenesis, Genetic , Exons , Gene Expression Regulation, Leukemic , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Introns , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic
6.
Leukemia ; 31(11): 2315-2325, 2017 11.
Article in English | MEDLINE | ID: mdl-28216661

ABSTRACT

Deregulation of epigenetic mechanisms, including microRNA, contributes to leukemogenesis and drug resistance by interfering with cancer-specific molecular pathways. Here, we show that the balance between miR-194-5p and its newly discovered target BCL2-associated transcription factor 1 (BCLAF1) regulates differentiation and survival of normal hematopoietic progenitors. In acute myeloid leukemias this balance is perturbed, locking cells into an immature, potentially 'immortal' state. Enhanced expression of miR-194-5p by treatment with the histone deacetylase inhibitor SAHA or by exogenous miR-194-5p expression re-sensitizes cells to differentiation and apoptosis by inducing BCLAF1 to shuttle between nucleus and cytosol. miR-194-5p/BCLAF1 balance was found commonly deregulated in 60 primary acute myeloid leukemia patients and was largely restored by ex vivo SAHA treatment. Our findings link treatment responsiveness to re-instatement of miR-194-5p/BCLAF1 balance.


Subject(s)
Gene Expression Regulation , Leukemia, Myeloid, Acute/pathology , MicroRNAs/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Apoptosis , Cell Cycle , Cell Differentiation , Cell Line, Tumor , Down-Regulation , Humans , Leukemia, Myeloid, Acute/genetics
8.
Oncogene ; 36(23): 3346-3356, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28114278

ABSTRACT

In 11q23 leukemias, the N-terminal part of the mixed lineage leukemia (MLL) gene is fused to >60 different partner genes. In order to define a core set of MLL rearranged targets, we investigated the genome-wide binding of the MLL-AF9 and MLL-AF4 fusion proteins and associated epigenetic signatures in acute myeloid leukemia (AML) cell lines THP-1 and MV4-11. We uncovered both common as well as specific MLL-AF9 and MLL-AF4 target genes, which were all marked by H3K79me2, H3K27ac and H3K4me3. Apart from promoter binding, we also identified MLL-AF9 and MLL-AF4 binding at specific subsets of non-overlapping active distal regulatory elements. Despite this differential enhancer binding, MLL-AF9 and MLL-AF4 still direct a common gene program, which represents part of the RUNX1 gene program and constitutes of CD34+ and monocyte-specific genes. Comparing these data sets identified several zinc finger transcription factors (TFs) as potential MLL-AF9 co-regulators. Together, these results suggest that MLL fusions collaborate with specific subsets of TFs to deregulate the RUNX1 gene program in 11q23 AMLs.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Adult , Core Binding Factor Alpha 2 Subunit/genetics , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Infant , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Staging , Oncogene Proteins, Fusion/genetics , Prognosis , Promoter Regions, Genetic
9.
Oncogene ; 35(15): 1965-76, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26148230

ABSTRACT

The ETS transcription factor ERG has been implicated as a major regulator of both normal and aberrant hematopoiesis. In acute myeloid leukemias harboring t(16;21), ERG function is deregulated due to a fusion with FUS/TLS resulting in the expression of a FUS-ERG oncofusion protein. How this oncofusion protein deregulates the normal ERG transcription program is unclear. Here, we show that FUS-ERG acts in the context of a heptad of proteins (ERG, FLI1, GATA2, LYL1, LMO2, RUNX1 and TAL1) central to proper expression of genes involved in maintaining a stem cell hematopoietic phenotype. Moreover, in t(16;21) FUS-ERG co-occupies genomic regions bound by the nuclear receptor heterodimer RXR:RARA inhibiting target gene expression and interfering with hematopoietic differentiation. All-trans retinoic acid treatment of t(16;21) cells as well as FUS-ERG knockdown alleviate the myeloid-differentiation block. Together, the results suggest that FUS-ERG acts as a transcriptional repressor of the retinoic acid signaling pathway.


Subject(s)
Chromosomes, Human, Pair 16/genetics , Chromosomes, Human, Pair 21/genetics , Gene Expression Regulation, Neoplastic/genetics , Hematopoiesis/physiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myelomonocytic, Acute/genetics , Neoplasm Proteins/physiology , Oncogene Proteins, Fusion/physiology , RNA-Binding Protein FUS/physiology , Signal Transduction/physiology , Translocation, Genetic , Tretinoin/physiology , Amino Acid Motifs , Cell Line, Tumor , Chromosomes, Human, Pair 16/ultrastructure , Chromosomes, Human, Pair 21/ultrastructure , Dimerization , Enhancer Elements, Genetic , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/physiopathology , Leukemia, Myelomonocytic, Acute/pathology , Leukemia, Myelomonocytic, Acute/physiopathology , Multiprotein Complexes , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Promoter Regions, Genetic , Protein Binding , Protein Interaction Mapping , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA-Binding Protein FUS/antagonists & inhibitors , RNA-Binding Protein FUS/genetics , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptors/metabolism , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology , U937 Cells
10.
Genom Data ; 2: 170-2, 2014 Dec.
Article in English | MEDLINE | ID: mdl-26484088

ABSTRACT

The inv(16) translocation is associated with 5% of AML cases and gives rise to expression of the oncofusion protein CBFß-MYH11. Although different molecular mechanisms for the oncogenic activity of this fusion protein have been proposed these were mostly based on in vitro experiments or single loci analysis. Recently, we investigated the genome-wide action of this fusion protein in the context of other hematopoietic transcription factors (Mandoli et al., 2014). Here, we describe in detail the ChIP-seq and RNA-seq methods used to generate the data associated with this study. Our analysis of CBFß-MYH11 as well as multiple other hematopoietic transcription factors using ChIP-seq data revealed RUNX1 dependent binding of CBFß-MYH11 as well as interaction of the RUNX1/CBFß-MYH11 complex with other hematopoietic regulators. Further RNA-seq based analysis suggested that CBFß-MYH11 can act both as activator and repressor.

11.
Leukemia ; 28(4): 770-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24002588

ABSTRACT

Different mechanisms for CBFß-MYH11 function in acute myeloid leukemia with inv(16) have been proposed such as tethering of RUNX1 outside the nucleus, interference with transcription factor complex assembly and recruitment of histone deacetylases, all resulting in transcriptional repression of RUNX1 target genes. Here, through genome-wide CBFß-MYH11-binding site analysis and quantitative interaction proteomics, we found that CBFß-MYH11 localizes to RUNX1 occupied promoters, where it interacts with TAL1, FLI1 and TBP-associated factors (TAFs) in the context of the hematopoietic transcription factors ERG, GATA2 and PU.1/SPI1 and the coregulators EP300 and HDAC1. Transcriptional analysis revealed that upon fusion protein knockdown, a small subset of the CBFß-MYH11 target genes show increased expression, confirming a role in transcriptional repression. However, the majority of CBFß-MYH11 target genes, including genes implicated in hematopoietic stem cell self-renewal such as ID1, LMO1 and JAG1, are actively transcribed and repressed upon fusion protein knockdown. Together these results suggest an essential role for CBFß-MYH11 in regulating the expression of genes involved in maintaining a stem cell phenotype.


Subject(s)
Chromosome Inversion , Chromosomes, Human, Pair 16 , Core Binding Factor Alpha 2 Subunit/physiology , Core Binding Factor beta Subunit/physiology , Leukemia, Myeloid, Acute/genetics , Myosin Heavy Chains/physiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Binding Sites , GATA2 Transcription Factor/physiology , Histone Deacetylases/physiology , Humans , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1/physiology , Proto-Oncogene Proteins/physiology , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcriptional Activation
12.
Br J Cancer ; 104(4): 554-8, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21245861

ABSTRACT

PML-RAR (retinoic acid receptor)α is the hallmark protein of acute promyelocytic leukaemia, a highly malignant subtype of acute myeloid leukaemia that accounts for approximately 10% of all AML cases. Recently, several studies have been set out to obtain a comprehensive genome-wide view of the molecular actions of this chimeric protein. In this review, we highlight the new insights that arose from these studies, in particular focussing on newly identified PML-RARα target genes, its interplay with RXR and deregulation of epigenetic modifications.


Subject(s)
Genome, Human , Leukemia, Promyelocytic, Acute/genetics , Oncogene Proteins, Fusion/physiology , Animals , Epigenesis, Genetic/physiology , Humans , Models, Biological , Oncogene Proteins, Fusion/genetics , Signal Transduction/genetics , Signal Transduction/physiology
13.
Article in English | MEDLINE | ID: mdl-16568946

ABSTRACT

Epigenetic mechanisms control eukaryotic development beyond DNA-stored information. DNA methylation, histone modifications and variants, nucleosome remodeling and noncoding RNAs all contribute to the dynamic make-up of chromatin under distinct developmental options. In particular, the great diversity of covalent histone tail modifications has been proposed to be ideally suited for imparting epigenetic information. While most of the histone tail modifications represent transient marks at transcriptionally permissive chromatin, some modifications appear more robust at silent chromatin regions, where they index repressive epigenetic states with functions also outside transcriptional regulation. Under-representation of repressive histone marks could be indicative of epigenetic plasticity in stem, young and tumor cells, while committed and senescent (old) cells often display increased levels of these more stable modifications. Here, we discuss profiles of normal and aberrant histone lysine methylation patterns, as they occur during the transition of an embryonic to a differentiated cell or in controlled self-renewal vs pro-neoplastic or metastatic conditions. Elucidating these histone modification patterns promises to have important implications for novel advances in stem cell research, nuclear reprogramming and cancer, and may offer novel targets for the combat of tumor cells, potentially leading to new diagnostic and therapeutic avenues in human biology and disease.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Epigenesis, Genetic/genetics , Histones/genetics , Histones/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Transformation, Neoplastic/genetics , Embryonic Development/genetics , Humans , Methylation , Protein Structure, Tertiary/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...