Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Nat Commun ; 14(1): 6185, 2023 10 04.
Article En | MEDLINE | ID: mdl-37794021

The myeloid transcription factor CEBPA is recurrently biallelically mutated (i.e., double mutated; CEBPADM) in acute myeloid leukemia (AML) with a combination of hypermorphic N-terminal mutations (CEBPANT), promoting expression of the leukemia-associated p30 isoform, and amorphic C-terminal mutations. The most frequently co-mutated genes in CEBPADM AML are GATA2 and TET2, however the molecular mechanisms underlying this co-mutational spectrum are incomplete. By combining transcriptomic and epigenomic analyses of CEBPA-TET2 co-mutated patients with models thereof, we identify GATA2 as a conserved target of the CEBPA-TET2 mutational axis, providing a rationale for the mutational spectra in CEBPADM AML. Elevated CEBPA levels, driven by CEBPANT, mediate recruitment of TET2 to the Gata2 distal hematopoietic enhancer thereby increasing Gata2 expression. Concurrent loss of TET2 in CEBPADM AML induces a competitive advantage by increasing Gata2 promoter methylation, thereby rebalancing GATA2 levels. Of clinical relevance, demethylating treatment of Cebpa-Tet2 co-mutated AML restores Gata2 levels and prolongs disease latency.


Dioxygenases , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/pathology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Mutation , Regulatory Sequences, Nucleic Acid , Promoter Regions, Genetic/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism
2.
Haematologica ; 106(4): 1000-1007, 2021 04 01.
Article En | MEDLINE | ID: mdl-32381577

ASXL1 is one of the most commonly mutated genes in myeloid malignancies, including Myelodysplastic Syndrome (MDS) and Acute Myeloid Leukemia (AML). In order to further our understanding of the role of ASXL1 lesions in malignant hematopoiesis, we generated a novel knock-in mouse model carrying the most frequent ASXL1 mutation identified in MDS patients, p.G643WfsX12. Mutant mice did not display any major hematopoietic defects nor developed any apparent hematological disease. In AML patients, ASXL1 mutations co-occur with mutations in CEBPA and we therefore generated compound Cebpa and Asxl1 mutated mice. Using a transplantation model, we found that the mutated Asxl1 allele significantly accelerated disease development in a CEBPA mutant context. Importantly, we demonstrated that, similar to the human setting, Asxl1 mutated mice responded poorly to chemotherapy. This model therefore constitutes an excellent experimental system for further studies into the clinically important question of chemotherapy resistance mediated by mutant ASXL1.


Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Myeloproliferative Disorders , Animals , CCAAT-Enhancer-Binding Proteins , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Mutation , Myelodysplastic Syndromes/genetics , Repressor Proteins/genetics
3.
Sci Adv ; 5(7): eaaw4304, 2019 07.
Article En | MEDLINE | ID: mdl-31309149

The key myeloid transcription factor (TF), CEBPA, is frequently mutated in acute myeloid leukemia (AML), but the direct molecular effects of this leukemic driver mutation remain elusive. To investigate CEBPA mutant AML, we performed microscale, in vivo chromatin immunoprecipitation sequencing and identified a set of aberrantly activated enhancers, exclusively occupied by the leukemia-associated CEBPA-p30 isoform. Comparing gene expression changes in human CEBPA mutant AML and the corresponding Cebpa Lp30 mouse model, we identified Nt5e, encoding CD73, as a cross-species AML gene with an upstream leukemic enhancer physically and functionally linked to the gene. Increased expression of CD73, mediated by the CEBPA-p30 isoform, sustained leukemic growth via the CD73/A2AR axis. Notably, targeting of this pathway enhanced survival of AML-transplanted mice. Our data thus indicate a first-in-class link between a cancer driver mutation in a TF and a druggable, direct transcriptional target.


5'-Nucleotidase/genetics , CCAAT-Enhancer-Binding Proteins/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Mutation , Animals , Binding Sites , CCAAT-Enhancer-Binding Proteins/metabolism , Enhancer Elements, Genetic , Epigenesis, Genetic , GPI-Linked Proteins/genetics , Gene Expression Profiling , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Mice , Nucleotide Motifs , Prognosis , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/genetics
4.
Eur J Haematol ; 103(4): 319-328, 2019 Oct.
Article En | MEDLINE | ID: mdl-31254415

OBJECTIVES: Familial cases of hematological malignancies are associated with germline mutations. In particular, heterozygous mutations of SRP72 correlate with the development of myelodysplasia and bone marrow aplasia in two families. The signal recognition particle 72 kDa protein (SRP72) is part of the SRP complex, responsible for targeting of proteins to the endoplasmic reticulum. The main objective of this study is to investigate the role of SRP72 in the hematopoietic system, thus explaining why a reduced dose could increase susceptibility to hematological malignancies. METHODS: We developed an Srp72 null mouse model and characterized its hematopoietic system using flow cytometry, bone marrow transplantations, and gene expression analysis. RESULTS: Heterozygous loss of Srp72 in mice is not associated with major changes in hematopoiesis, although causes mild reductions in blood and BM cellularity and minor changes within the stem/progenitor compartment. We did not observe any hematological disorder. Interestingly, gene expression analysis demonstrated that genes encoding secreted factors, including cytokines and receptors, were transcriptionally down-regulated in Srp72+/- animals. CONCLUSIONS: The Srp72+/- mouse model only partially recapitulates the phenotype observed in families with inherited SRP72 lesions. Nonetheless, these results can provide mechanistic insights into why SRP72 mutations are associated with aplasia and myelodysplasia in humans.


Genetic Association Studies , Genetic Predisposition to Disease , Hematopoiesis/genetics , Loss of Heterozygosity , Mutation , Phenotype , Signal Recognition Particle/genetics , Animals , Biomarkers , Blood Cell Count , Bone Marrow/pathology , Disease Models, Animal , Gene Editing , Gene Expression , Genes, Lethal , Genotype , Mice , Mice, Knockout
5.
BMC Genomics ; 16: 46, 2015 Feb 05.
Article En | MEDLINE | ID: mdl-25652644

BACKGROUND: Chromatin-Immunoprecipitation coupled with deep sequencing (ChIP-seq) is used to map transcription factor occupancy and generate epigenetic profiles genome-wide. The requirement of nano-scale ChIP DNA for generation of sequencing libraries has impeded ChIP-seq on in vivo tissues of low cell numbers. RESULTS: We describe a robust, simple and scalable methodology for ChIP-seq of low-abundant cell populations, verified down to 10,000 cells. By employing non-mammalian genome mapping bacterial carrier DNA during amplification, we reliably amplify down to 50 pg of ChIP DNA from transcription factor (CEBPA) and histone mark (H3K4me3) ChIP. We further demonstrate that genomic profiles are highly resilient to changes in carrier DNA to ChIP DNA ratios. CONCLUSIONS: This represents a significant advance compared to existing technologies, which involve either complex steps of pre-selection for nucleosome-containing chromatin or pre-amplification of precipitated DNA, making them prone to introduce experimental biases.


Chromatin Immunoprecipitation , Nucleic Acid Amplification Techniques , Oligonucleotide Array Sequence Analysis , Transcription Factors/genetics , Animals , Chromatin/genetics , DNA, Bacterial/genetics , Genome, Bacterial/genetics , High-Throughput Nucleotide Sequencing , Sequence Analysis, DNA
6.
PLoS Genet ; 10(1): e1004079, 2014 Jan.
Article En | MEDLINE | ID: mdl-24415956

Transcription factors are key regulators of hematopoietic stem cells (HSCs) and act through their ability to bind DNA and impact on gene transcription. Their functions are interpreted in the complex landscape of chromatin, but current knowledge on how this is achieved is very limited. C/EBPα is an important transcriptional regulator of hematopoiesis, but its potential functions in HSCs have remained elusive. Here we report that C/EBPα serves to protect adult HSCs from apoptosis and to maintain their quiescent state. Consequently, deletion of Cebpa is associated with loss of self-renewal and HSC exhaustion. By combining gene expression analysis with genome-wide assessment of C/EBPα binding and epigenetic configurations, we show that C/EBPα acts to modulate the epigenetic states of genes belonging to molecular pathways important for HSC function. Moreover, our data suggest that C/EBPα acts as a priming factor at the HSC level where it actively promotes myeloid differentiation and counteracts lymphoid lineage choice. Taken together, our results show that C/EBPα is a key regulator of HSC biology, which influences the epigenetic landscape of HSCs in order to balance different cell fate options.


CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Differentiation/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Animals , Apoptosis , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Lineage , Cell Proliferation , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/metabolism , Mice
7.
Exp Hematol ; 41(10): 882-893.e16, 2013 Oct.
Article En | MEDLINE | ID: mdl-23831605

Acute myeloid leukemia (AML) develops via a multistep process involving several genetic and epigenetic events, which ultimately leads to the formation of a heterogeneous population of malignant cells, of which only a small subpopulation termed the leukemia initiating cell (LIC) is able to sustain the leukemia. The identity of the LIC is highly diverse and ranges from populations resembling hematopoietic stem cells or multipotent progenitors (MPPs) to more committed myeloid progenitors, and the question still remains whether this is a direct consequence of which cells are targets of the final transforming events. In this study, we use premalignant cells from a Cebpa mutant AML model, in which the LIC population resembles granulocyte-macrophage progenitors (GMPs), to show that premalignant GMPs undergo spontaneous immortalization with a high clonal frequency when cultured in vitro, suggesting that these cells constitute the target of the final transforming events. Furthermore, we show that premalignant GMPs are characterized by a distinct T cell gene expression signature correlating with an increased potential for differentiation toward the T cell lineage. These findings have implications for our understanding of the transcriptional wiring in premalignant myeloid progenitors and how this contributes to the development of AML.


CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Lineage , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/pathology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Genetic Variation , Leukemia, Myeloid, Acute/pathology , Mice , Mutation , Phenotype , Time Factors
8.
PLoS One ; 7(6): e38841, 2012.
Article En | MEDLINE | ID: mdl-22715416

BACKGROUND: Transcription factors play a key role in lineage commitment and differentiation of stem cells into distinct mature cells. In hematopoiesis, they regulate lineage-specific gene expression in a stage-specific manner through various physical and functional interactions with regulatory proteins that are simultanously recruited and activated to ensure timely gene expression. The transcription factor CCAAT/enhancer binding protein α (C/EBPα) is such a factor and is essential for the development of granulocytic/monocytic cells. The activity of C/EBPα is regulated on several levels including gene expression, alternative translation, protein interactions and posttranslational modifications, such as phosphorylation. In particular, the phosphorylation of serine 248 of the transactivation domain has been shown to be of crucial importance for granulocytic differentiation of 32Dcl3 cells in vitro. METHODOLOGY/PRINCIPAL FINDINGS: Here, we use mouse genetics to investigate the significance of C/EBPα serine 248 in vivo through the construction and analysis of Cebpa(S248A/S248A) knock-in mice. Surprisingly, 8-week old Cebpa(S248A/S248A) mice display normal steady-state hematopoiesis including unaltered development of mature myeloid cells. However, over time some of the animals develop a hematopoietic disorder with accumulation of multipotent, megakaryocytic and erythroid progenitor cells and a mild impairment of differentiation along the granulocytic-monocytic lineage. Furthermore, BM cells from Cebpa(S248A/S248A) animals display a competitive advantage compared to wild type cells in a transplantation assay. CONCLUSIONS/SIGNIFICANCE: Taken together, our data shows that the substitution of C/EBPα serine 248 to alanine favors the selection of the megakaryocytic/erythroid lineage over the monocytic/granulocytic compartment in old mice and suggests that S248 phosphorylation may be required to maintain proper hematopoietic homeostasis in response to changes in the wiring of cellular signalling networks. More broadly, the marked differences between the phenotype of the S248A variant in vivo and in vitro highlight the need to exert caution when extending in vitro phenotypes to the more appropriate in vivo context.


CCAAT-Enhancer-Binding Proteins/metabolism , Erythroid Precursor Cells/metabolism , Megakaryocyte Progenitor Cells/metabolism , Mutation, Missense , Myelopoiesis , Myeloproliferative Disorders/metabolism , Amino Acid Substitution , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cell Line , Erythroid Precursor Cells/pathology , Megakaryocyte Progenitor Cells/pathology , Mice , Mice, Mutant Strains , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Phosphorylation , Serine/genetics , Serine/metabolism
9.
Cancer Cell ; 13(4): 299-310, 2008 Apr.
Article En | MEDLINE | ID: mdl-18394553

Mutations in the CEBPA gene are present in 7%-10% of human patients with acute myeloid leukemia (AML). However, no genetic models exist that demonstrate their etiological relevance. To mimic the most common mutations affecting CEBPA-that is, those leading to loss of the 42 kDa C/EBPalpha isoform (p42) while retaining the 30kDa isoform (p30)-we modified the mouse Cebpa locus to express only p30. p30 supported the formation of granulocyte-macrophage progenitors. However, p42 was required for control of myeloid progenitor proliferation, and p42-deficient mice developed AML with complete penetrance. p42-deficient leukemia could be transferred by a Mac1+c-Kit+ population that gave rise only to myeloid cells in recipient mice. Expression profiling of this population against normal Mac1+c-Kit+ progenitors revealed a signature shared with MLL-AF9-transformed AML.


CCAAT-Enhancer-Binding Protein-alpha/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myelomonocytic, Acute/genetics , Leukemia, Myelomonocytic, Acute/pathology , Models, Biological , Mutant Proteins/metabolism , Neoplastic Stem Cells/pathology , Animals , CCAAT-Enhancer-Binding Protein-alpha/deficiency , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Differentiation , Disease Progression , Gene Expression Profiling , Granulocytes/cytology , Macrophage-1 Antigen/metabolism , Mice , Mice, Knockout , Myeloid Progenitor Cells/pathology , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Phenotype , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-kit/metabolism
10.
Blood ; 111(8): 4309-21, 2008 Apr 15.
Article En | MEDLINE | ID: mdl-18212247

The CCAAT enhancer binding protein alpha (C/EBPalpha) is an important myeloid tumor suppressor that is frequently mutated in human acute myeloid leukemia (AML). We have previously shown that mice homozygous for the E2F repression-deficient Cebpa(BRM2) allele develop nonfatal AML with long latency and incomplete penetrance, suggesting that accumulation of secondary mutations is necessary for disease progression. Here, we use SRS19-6-driven retroviral insertional mutagenesis to compare the phenotypes of leukemias arising in Cebpa(+/+), Cebpa(+/BRM2), and Cebpa(BRM2/BRM2) mice, with respect to disease type, latency of tumor development, and identity of the retroviral insertion sites (RISs). Both Cebpa(+/BRM2) and Cebpa(BRM2/BRM2) mice preferentially develop myeloid leukemias, but with differing latencies, thereby demonstrating the importance of gene dosage. Determination of RISs led to the identification of several novel candidate oncogenes, some of which may collaborate specifically with the E2F repression-deficient allele of Cebpa. Finally, we used an in silico pathway analysis approach to extract additional information from single RISs, leading to the identification of signaling pathways which were preferentially deregulated in a disease- and/or genotype-specific manner.


CCAAT-Enhancer-Binding Protein-alpha/genetics , Genetic Predisposition to Disease , Leukemia, Myeloid/genetics , Leukemia, Myeloid/pathology , Mutagenesis, Insertional , Mutation/genetics , Retroviridae/genetics , Alleles , Animals , Clone Cells , Computational Biology , Gene Expression Regulation, Leukemic , Gene Rearrangement , Genes, Neoplasm , Genomic Instability , Immunoglobulins/genetics , Injections , Mice , Mice, Inbred C57BL , Phenotype , Precancerous Conditions/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , Retroviridae/physiology , Virus Latency
...