Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nat Immunol ; 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992254

ABSTRACT

Upregulation of diverse self-antigens that constitute components of the inflammatory response overlaps spatially and temporally with the emergence of pathogen-derived foreign antigens. Therefore, discrimination between these inflammation-associated self-antigens and pathogen-derived molecules represents a unique challenge for the adaptive immune system. Here, we demonstrate that CD8+ T cell tolerance to T cell-derived inflammation-associated self-antigens is efficiently induced in the thymus and supported by redundancy in cell types expressing these molecules. In addition to thymic epithelial cells, this included thymic eosinophils and innate-like T cells, a population that expressed molecules characteristic for all major activated T cell subsets. We show that direct T cell-to-T cell antigen presentation by minute numbers of innate-like T cells was sufficient to eliminate autoreactive CD8+ thymocytes. Tolerance to such effector molecules was of critical importance, as its breach caused by decreased thymic abundance of a single model inflammation-associated self-antigen resulted in autoimmune elimination of an entire class of effector T cells.

3.
Comput Struct Biotechnol J ; 21: 21-33, 2023.
Article in English | MEDLINE | ID: mdl-36514338

ABSTRACT

Hematopoietic stem cell (HSC) aging is a multifactorial event leading to changes in HSC properties and functions, which are intrinsically coordinated and affect the early hematopoiesis. To better understand the mechanisms and factors controlling these changes, we developed an original strategy to construct a Boolean model of HSC differentiation. Based on our previous scRNA-seq data, we exhaustively characterized active transcription modules or regulons along the differentiation trajectory and constructed an influence graph between 15 selected components involved in the dynamics of the process. Then we defined dynamical constraints between observed cellular states along the trajectory and using answer set programming with in silico perturbation analysis, we obtained a Boolean model explaining the early priming of HSCs. Finally, perturbations of the model based on age-related changes revealed important deregulations, such as the overactivation of Egr1 and Junb or the loss of Cebpa activation by Gata2. These new regulatory mechanisms were found to be relevant for the myeloid bias of aged HSC and explain the decreased transcriptional priming of HSCs to all mature cell types except megakaryocytes.

4.
Cells ; 11(19)2022 10 04.
Article in English | MEDLINE | ID: mdl-36231086

ABSTRACT

Single-cell transcriptomic technologies enable the uncovering and characterization of cellular heterogeneity and pave the way for studies aiming at understanding the origin and consequences of it. The hematopoietic system is in essence a very well adapted model system to benefit from this technological advance because it is characterized by different cellular states. Each cellular state, and its interconnection, may be defined by a specific location in the global transcriptional landscape sustained by a complex regulatory network. This transcriptomic signature is not fixed and evolved over time to give rise to less efficient hematopoietic stem cells (HSC), leading to a well-documented hematopoietic aging. Here, we review the advance of single-cell transcriptomic approaches for the understanding of HSC heterogeneity to grasp HSC deregulations upon aging. We also discuss the new bioinformatics tools developed for the analysis of the resulting large and complex datasets. Finally, since hematopoiesis is driven by fine-tuned and complex networks that must be interconnected to each other, we highlight how mathematical modeling is beneficial for doing such interconnection between multilayered information and to predict how HSC behave while aging.


Subject(s)
Hematopoietic Stem Cells , Transcriptome , Hematopoiesis/genetics , Models, Biological , Transcriptome/genetics
5.
Blood ; 140(22): 2358-2370, 2022 12 01.
Article in English | MEDLINE | ID: mdl-35984905

ABSTRACT

Cancer cell heterogeneity is a major driver of therapy resistance. To characterize resistant cells and their vulnerabilities, we studied the PLZF-RARA variant of acute promyelocytic leukemia, resistant to retinoic acid (RA), using single-cell multiomics. We uncovered transcriptional and chromatin heterogeneity in leukemia cells. We identified a subset of cells resistant to RA with proliferation, DNA replication, and repair signatures that depend on a fine-tuned E2F transcriptional network targeting the epigenetic regulator enhancer of zeste homolog 2 (EZH2). Epigenomic and functional analyses validated the driver role of EZH2 in RA resistance. Targeting pan-EZH2 activities (canonical/noncanonical) was necessary to eliminate leukemia relapse-initiating cells, which underlies a dependency of resistant cells on an EZH2 noncanonical activity and the necessity to degrade EZH2 to overcome resistance. Our study provides critical insights into the mechanisms of RA resistance that allow us to eliminate treatment-resistant leukemia cells by targeting EZH2, thus highlighting a potential targeted therapy approach. Beyond RA resistance and acute promyelocytic leukemia context, our study also demonstrates the power of single-cell multiomics to identify, characterize, and clear therapy-resistant cells.


Subject(s)
Leukemia, Promyelocytic, Acute , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Tretinoin/pharmacology , Enhancer of Zeste Homolog 2 Protein/genetics , Retinoic Acid Receptor alpha/genetics , Receptors, Retinoic Acid/genetics , Transcription Factors/genetics , Nuclear Proteins/genetics
6.
Nucleic Acids Res ; 50(14): 7938-7958, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35871293

ABSTRACT

Although originally described as transcriptional activator, SPI1/PU.1, a major player in haematopoiesis whose alterations are associated with haematological malignancies, has the ability to repress transcription. Here, we investigated the mechanisms underlying gene repression in the erythroid lineage, in which SPI1 exerts an oncogenic function by blocking differentiation. We show that SPI1 represses genes by binding active enhancers that are located in intergenic or gene body regions. HDAC1 acts as a cooperative mediator of SPI1-induced transcriptional repression by deacetylating SPI1-bound enhancers in a subset of genes, including those involved in erythroid differentiation. Enhancer deacetylation impacts on promoter acetylation, chromatin accessibility and RNA pol II occupancy. In addition to the activities of HDAC1, polycomb repressive complex 2 (PRC2) reinforces gene repression by depositing H3K27me3 at promoter sequences when SPI1 is located at enhancer sequences. Moreover, our study identified a synergistic relationship between PRC2 and HDAC1 complexes in mediating the transcriptional repression activity of SPI1, ultimately inducing synergistic adverse effects on leukaemic cell survival. Our results highlight the importance of the mechanism underlying transcriptional repression in leukemic cells, involving complex functional connections between SPI1 and the epigenetic regulators PRC2 and HDAC1.


Subject(s)
Histone Deacetylase 1 , Leukemia, Erythroblastic, Acute , Polycomb Repressive Complex 2 , Proto-Oncogene Proteins , Trans-Activators , Acetylation , Animals , Chromatin/genetics , Histone Deacetylase 1/genetics , Leukemia, Erythroblastic, Acute/genetics , Mice , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics
7.
Cancer Discov ; 12(3): 774-791, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34862195

ABSTRACT

Cancer cell metabolism is increasingly recognized as providing an exciting therapeutic opportunity. However, a drug that directly couples targeting of a metabolic dependency with the induction of cell death in cancer cells has largely remained elusive. Here we report that the drug-like small-molecule ironomycin reduces the mitochondrial iron load, resulting in the potent disruption of mitochondrial metabolism. Ironomycin promotes the recruitment and activation of BAX/BAK, but the resulting mitochondrial outer membrane permeabilization (MOMP) does not lead to potent activation of the apoptotic caspases, nor is the ensuing cell death prevented by inhibiting the previously established pathways of programmed cell death. Consistent with the fact that ironomycin and BH3 mimetics induce MOMP through independent nonredundant pathways, we find that ironomycin exhibits marked in vitro and in vivo synergy with venetoclax and overcomes venetoclax resistance in primary patient samples. SIGNIFICANCE: Ironomycin couples targeting of cellular metabolism with cell death by reducing mitochondrial iron, resulting in the alteration of mitochondrial metabolism and the activation of BAX/BAK. Ironomycin induces MOMP through a different mechanism to BH3 mimetics, and consequently combination therapy has marked synergy in cancers such as acute myeloid leukemia. This article is highlighted in the In This Issue feature, p. 587.


Subject(s)
Iron , bcl-2 Homologous Antagonist-Killer Protein , Apoptosis , Cell Death , Humans , Iron/metabolism , Mitochondria/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
8.
Blood ; 138(6): 422-423, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34383038
10.
BMC Biol ; 19(1): 19, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33526011

ABSTRACT

BACKGROUND: Hematopoietic stem cells (HSCs) are the guarantor of the proper functioning of hematopoiesis due to their incredible diversity of potential. During aging, heterogeneity of HSCs changes, contributing to the deterioration of the immune system. In this study, we revisited mouse HSC compartment and its transcriptional plasticity during aging at unicellular scale. RESULTS: Through the analysis of 15,000 young and aged transcriptomes, we identified 15 groups of HSCs revealing rare and new specific HSC abilities that change with age. The implantation of new trajectories complemented with the analysis of transcription factor activities pointed consecutive states of HSC differentiation that were delayed by aging and explained the bias in differentiation of older HSCs. Moreover, reassigning cell cycle phases for each HSC clearly highlighted an imbalance of the cell cycle regulators of very immature aged HSCs that may contribute to their accumulation in an undifferentiated state. CONCLUSIONS: Our results establish a new reference map of HSC differentiation in young and aged mice and reveal a potential mechanism that delays the differentiation of aged HSCs and could promote the emergence of age-related hematologic diseases.


Subject(s)
Aging , Cell Cycle , Cell Differentiation , Hematopoietic Stem Cells/physiology , RNA-Seq , Single-Cell Analysis , Animals , Male , Mice
11.
Clin Epigenetics ; 11(1): 141, 2019 10 12.
Article in English | MEDLINE | ID: mdl-31606046

ABSTRACT

BACKGROUND: The epigenetic machinery is frequently altered in acute myeloid leukemia. Focusing on cytogenetically normal (CN) AML, we previously described an abnormal H3K27me3 enrichment covering 70 kb on the HIST1 cluster (6.p22) in CN-AML patient blasts. Here, we further investigate the molecular, functional, and prognosis significance of this epigenetic alteration named H3K27me3 HIST1 in NPM1-mutated (NPM1mut) CN-AML. RESULTS: We found that three quarter of the NPM1mut CN-AML patients were H3K27me3 HIST1high. H3K27me3 HIST1high group of patients was associated with a favorable outcome independently of known molecular risk factors. In gene expression profiling, the H3K27me3 HIST1high mark was associated with lower expression of the histone genes HIST1H1D, HIST1H2BG, HIST1H2AE, and HIST1H3F and an upregulation of genes involved in myelomonocytic differentiation. Mass spectrometry analyses confirmed that the linker histone protein H1d, but not the other histone H1 subtypes, was downregulated in the H3K27me3 HIST1high group of patients. H1d knockdown primed ATRA-mediated differentiation of OCI-AML3 and U937 AML cell lines, as assessed on CD11b/CD11c markers, morphological and gene expression analyses. CONCLUSIONS: Our data suggest that NPM1mut AML prognosis depends on the epigenetic silencing of the HIST1 cluster and that, among the H3K27me3 silenced histone genes, HIST1H1D plays a role in AML blast differentiation.


Subject(s)
Down-Regulation , Histones/genetics , Histones/metabolism , Leukemia, Myeloid, Acute/mortality , Mutation , Nuclear Proteins/genetics , Adult , Aged , Cell Differentiation , Cell Line, Tumor , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genetic Loci , Humans , Leukemia, Myeloid, Acute/genetics , Male , Methylation , Middle Aged , Nucleophosmin , Prognosis , Survival Analysis , Young Adult
12.
Nucleic Acids Res ; 47(9): 4509-4520, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30892634

ABSTRACT

PLZF (promyelocytic leukemia zinc finger) is a transcription factor acting as a global regulator of hematopoietic commitment. PLZF displays an epigenetic specificity by recruiting chromatin-modifying factors but little is known about its role in remodeling chromatin of cells committed toward a given specific hematopoietic lineage. In murine myeloid progenitors, we decipher a new role for PLZF in restraining active genes and enhancers by targeting acetylated lysine 27 of Histone H3 (H3K27ac). Functional analyses reveal that active enhancers bound by PLZF are involved in biological processes related to metabolism and associated with hematopoietic aging. Comparing the epigenome of young and old myeloid progenitors, we reveal that H3K27ac variation at active enhancers is a hallmark of hematopoietic aging. Taken together, these data suggest that PLZF, associated with active enhancers, appears to restrain their activity as an epigenetic gatekeeper of hematopoietic aging.


Subject(s)
Aging/genetics , Hematopoietic Stem Cells/metabolism , Promyelocytic Leukemia Zinc Finger Protein/genetics , Transcription, Genetic , Animals , Cell Differentiation/genetics , Enhancer Elements, Genetic , Epigenesis, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Histones/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Myeloid Progenitor Cells/metabolism , Protein Binding , Regulatory Sequences, Nucleic Acid/genetics
13.
Cell Rep ; 26(12): 3257-3271.e8, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30893599

ABSTRACT

In the bone marrow, CXCL12 and IL-7 are essential for B cell differentiation, whereas hematopoietic stem cell (HSC) maintenance requires SCF and CXCL12. Peri-sinusoidal stromal (PSS) cells are the main source of IL-7, but their characterization as a pro-B cell niche remains limited. Here, we characterize pro-B cell supporting stromal cells and decipher the interaction network allowing pro-B cell retention. Preferential contacts are found between pro-B cells and PSS cells, which homogeneously express HSC and B cell niche genes. Furthermore, pro-B cells are frequently located in the vicinity of HSCs in the same niche. Using an interactome bioinformatics pipeline, we identify Nidogen-1 as essential for pro-B cell retention in the peri-sinusoidal niche as confirmed in Nidogen-1-/- mice. Finally, human pro-B cells and hematopoietic progenitors are observed close to similar IL-7+ stromal cells. Thus, a multispecific niche exists in mouse and human supporting both early progenitors and committed hematopoietic lineages.


Subject(s)
Hematopoietic Stem Cells/immunology , Membrane Glycoproteins/immunology , Precursor Cells, B-Lymphoid/immunology , Stem Cell Niche/immunology , Animals , Hematopoietic Stem Cells/cytology , Interleukin-7/genetics , Interleukin-7/immunology , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Precursor Cells, B-Lymphoid/cytology , Stromal Cells/cytology , Stromal Cells/immunology
14.
Proc Natl Acad Sci U S A ; 116(1): 211-216, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30559202

ABSTRACT

Bone marrow (BM) produces all blood and immune cells deriving from hematopoietic stem cells (HSCs). The decrease of immune cell production during aging is one of the features of immunosenescence. The impact of redox dysregulation in BM aging is still poorly understood. Here we use TP53INP1-deficient (KO) mice endowed with chronic oxidative stress to assess the influence of aging-associated redox alterations in BM homeostasis. We show that TP53INP1 deletion has no impact on aging-related accumulation of HSCs. In contrast, the aging-related contraction of the lymphoid compartment is mitigated in TP53INP1 KO mice. B cells that accumulate in old KO BM are differentiating cells that can mature into functional B cells. Importantly, this phenotype results from B cell-intrinsic events associated with defective redox control. Finally, we show that oxidative stress in aged TP53INP1-deficient mice maintains STAT5 expression and activation in early B cells, driving high Pax5 expression, which provides a molecular mechanism for maintenance of B cell development upon aging.


Subject(s)
B-Lymphocytes/physiology , Bone Marrow/physiology , Lymphopoiesis , Nuclear Proteins/deficiency , Receptors, Interleukin-7/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Aging/physiology , Animals , B-Lymphocytes/metabolism , Bone Marrow/metabolism , Lymphopoiesis/physiology , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Oxidative Stress
15.
Nucleic Acids Res ; 46(7): 3339-3350, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29425303

ABSTRACT

The transcription factor PLZF (promyelocytic leukemia zinc finger protein) acts as an epigenetic regulator balancing self-renewal and differentiation of hematopoietic cells through binding to various chromatin-modifying factors. First described as a transcriptional repressor, PLZF is also associated with active transcription, although the molecular bases underlying the differences are unknown. Here, we reveal that in a hematopoietic cell line, PLZF is predominantly associated with transcribed genes. Additionally, we identify a new association between PLZF and the histone methyltransferase, EZH2 at the genomic level. We find that co-occupancy of PLZF and EZH2 on chromatin at PLZF target genes is not associated with SUZ12 or trimethylated lysine 27 of histone H3 (H3K27me3) but with the active histone mark H3K4me3 and active transcription. Removal of EZH2 leads to an increase of PLZF binding and increased gene expression. Our results suggest a new role of EZH2 in restricting PLZF positive transcriptional activity independently of its canonical PRC2 activity.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Polycomb Repressive Complex 2/genetics , Promyelocytic Leukemia Zinc Finger Protein/genetics , Transcription, Genetic , Binding Sites/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Self Renewal/genetics , Chromatin/genetics , Gene Expression Regulation/genetics , Hematopoietic Stem Cells/metabolism , Histone Methyltransferases/genetics , Histones/genetics , Humans , Neoplasm Proteins , Protein Binding/genetics , Transcription Factors
16.
Med Sci (Paris) ; 33(5): 499-505, 2017 May.
Article in French | MEDLINE | ID: mdl-28612725

ABSTRACT

Polycomb Group proteins (PcG) are repressive epigenetic factors essential for development and involved in numerous cancer processes, yet their modes of action and recruitment to specific genomic loci are not fully understood. Recently, it has been shown that the PcG protein recruitment is a dynamic process, contrary to what was foreseen in the initial hierarchical model. In addition, EZH2, a key PcG protein, can be associated to transcribed genes, challenging the former function of PcG proteins as transcriptional repressors. Furthermore, the dual role of EZH2, which can act as an oncogene or a tumor suppressor depending on the cellular type, illustrates the functional complexity of PcG proteins.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/physiology , Polycomb-Group Proteins/physiology , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Gene Expression Regulation , Gene Regulatory Networks/genetics , Genes, Tumor Suppressor , Humans , Oncogenes/physiology , Signal Transduction/genetics
17.
Blood ; 127(15): 1881-5, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26941402

ABSTRACT

Hematopoietic stem cells (HSCs) give rise to all blood populations due to their long-term self-renewal and multipotent differentiation capacities. Because they have to persist throughout an organism's life span, HSCs tightly regulate the balance between proliferation and quiescence. Here, we investigated the role of the transcription factor promyelocytic leukemia zinc finger (plzf) in HSC fate using the Zbtb16(lu/lu)mouse model, which harbors a natural spontaneous mutation that inactivates plzf. Regenerative stress revealed that Zbtb16(lu/lu)HSCs had a lineage-skewing potential from lymphopoiesis toward myelopoiesis, an increase in the long-term-HSC pool, and a decreased repopulation potential. Furthermore, oldplzf-mutant HSCs present an amplified aging phenotype, suggesting that plzf controls age-related pathway. We found that Zbtb16(lu/lu)HSCs harbor a transcriptional signature associated with a loss of stemness and cell cycle deregulation. Lastly, cell cycle analyses revealed an important role for plzf in the regulation of the G1-S transition of HSCs. Our study reveals a new role for plzf in regulating HSC function that is linked to cell cycle regulation, and positions plzf as a key player in controlling HSC homeostasis.


Subject(s)
Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , Mutation , Animals , Apoptosis , Cell Cycle , Cell Differentiation , Cell Lineage , Cellular Senescence , Epigenesis, Genetic , Gene Expression Profiling , Homeostasis , Lymphopoiesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelopoiesis , Oligonucleotide Array Sequence Analysis , Phenotype , Promyelocytic Leukemia Zinc Finger Protein
18.
Blood ; 125(19): 2985-94, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25805812

ABSTRACT

The gene CXXC5 on 5q31 is frequently deleted in acute myeloid leukemia (AML) with del(5q), suggesting that inactivation of CXXC5 might play a role in leukemogenesis. Here, we investigated the functional and prognostic implications of CXXC5 expression in AML. CXXC5 mRNA was downregulated in AML with MLL rearrangements, t(8;21) and GATA2 mutations. As a mechanism of CXXC5 inactivation, we found evidence for epigenetic silencing by promoter methylation. Patients with CXXC5 expression below the median level had a lower relapse rate (45% vs 59%; P = .007) and a better overall survival (OS, 46% vs 28%; P < .001) and event-free survival (EFS, 36% vs 21%; P < .001) at 5 years, independent of cytogenetic risk groups and known molecular risk factors. In gene-expression profiling, lower CXXC5 expression was associated with upregulation of cell-cycling genes and co-downregulation of genes implicated in leukemogenesis (WT1, GATA2, MLL, DNMT3B, RUNX1). Functional analyses demonstrated CXXC5 to inhibit leukemic cell proliferation and Wnt signaling and to affect the p53-dependent DNA damage response. In conclusion, our data suggest a tumor suppressor function of CXXC5 in AML. Inactivation of CXXC5 is associated with different leukemic pathways and defines an AML subgroup with better outcome.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Wnt Proteins/antagonists & inhibitors , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Cycle , Cohort Studies , DNA Methylation , DNA-Binding Proteins , Down-Regulation , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Prognosis , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Survival Rate , Transcription Factors , Tumor Cells, Cultured , Young Adult
19.
Nat Commun ; 6: 6094, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25615415

ABSTRACT

T-cell acute lymphoblastic leukaemias (T-ALL) are aggressive malignant proliferations characterized by high relapse rates and great genetic heterogeneity. TAL1 is amongst the most frequently deregulated oncogenes. Yet, over half of the TAL1(+) cases lack TAL1 lesions, suggesting unrecognized (epi)genetic deregulation mechanisms. Here we show that TAL1 is normally silenced in the T-cell lineage, and that the polycomb H3K27me3-repressive mark is focally diminished in TAL1(+) T-ALLs. Sequencing reveals that >20% of monoallelic TAL1(+) patients without previously known alterations display microinsertions or RAG1/2-mediated episomal reintegration in a single site 5' to TAL1. Using 'allelic-ChIP' and CrispR assays, we demonstrate that such insertions induce a selective switch from H3K27me3 to H3K27ac at the inserted but not the germline allele. We also show that, despite a considerable mechanistic diversity, the mode of oncogenic TAL1 activation, rather than expression levels, impact on clinical outcome. Altogether, these studies establish site-specific epigenetic desilencing as a mechanism of oncogenic activation.


Subject(s)
Alleles , Gene Expression Regulation, Leukemic , Polycomb-Group Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Acetylation , Adult , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Genetic Loci , Histones/metabolism , Homeodomain Proteins/metabolism , Humans , Jurkat Cells , Methylation , Molecular Sequence Data , Mutagenesis, Insertional , Nuclear Proteins/metabolism , Plasmids/genetics , Polycomb-Group Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Survival Analysis , T-Cell Acute Lymphocytic Leukemia Protein 1 , Treatment Outcome
20.
Crit Rev Oncol Hematol ; 88(2): 231-45, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23838480

ABSTRACT

Dysregulation of cellular epigenetic machinery is considered a major pathogenetic determinant in many malignancies, including myelodysplastic syndromes (MDS). The importance of epigenetic dysfunction in MDS is reflected by the success of hypomethylating agents as standard of care for their treatment. Although these agents improve both survival and quality of life, knowledge gaps remain regarding the precise role of epigenetics in the pathogenesis of MDS and mechanisms by which hypomethylating agents exert their clinical effects. This article reviews the pathogenic role of epigenetic alterations in MDS, including the relationship between genetic and epigenetic abnormalities, and highlights emerging evidence that hypomethylating agents may reprogram the "methylome" while re-establishing hematopoiesis.


Subject(s)
Epigenesis, Genetic , Myelodysplastic Syndromes/genetics , Chromosome Aberrations , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Epigenomics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histones/metabolism , Humans , Leukemia, Myeloid/genetics , Methylation , Myelodysplastic Syndromes/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...