Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell Genom ; 3(12): 100426, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38116120

ABSTRACT

Acute myeloid leukemia (AML) and myeloid neoplasms develop through acquisition of somatic mutations that confer mutation-specific fitness advantages to hematopoietic stem and progenitor cells. However, our understanding of mutational effects remains limited to the resolution attainable within immunophenotypically and clinically accessible bulk cell populations. To decipher heterogeneous cellular fitness to preleukemic mutational perturbations, we performed single-cell RNA sequencing of eight different mouse models with driver mutations of myeloid malignancies, generating 269,048 single-cell profiles. Our analysis infers mutation-driven perturbations in cell abundance, cellular lineage fate, cellular metabolism, and gene expression at the continuous resolution, pinpointing cell populations with transcriptional alterations associated with differentiation bias. We further develop an 11-gene scoring system (Stem11) on the basis of preleukemic transcriptional signatures that predicts AML patient outcomes. Our results demonstrate that a single-cell-resolution deep characterization of preleukemic biology has the potential to enhance our understanding of AML heterogeneity and inform more effective risk stratification strategies.

2.
Nat Genet ; 55(9): 1542-1554, 2023 09.
Article in English | MEDLINE | ID: mdl-37580596

ABSTRACT

Cellular differentiation requires extensive alterations in chromatin structure and function, which is elicited by the coordinated action of chromatin and transcription factors. By contrast with transcription factors, the roles of chromatin factors in differentiation have not been systematically characterized. Here, we combine bulk ex vivo and single-cell in vivo CRISPR screens to characterize the role of chromatin factor families in hematopoiesis. We uncover marked lineage specificities for 142 chromatin factors, revealing functional diversity among related chromatin factors (i.e. barrier-to-autointegration factor subcomplexes) as well as shared roles for unrelated repressive complexes that restrain excessive myeloid differentiation. Using epigenetic profiling, we identify functional interactions between lineage-determining transcription factors and several chromatin factors that explain their lineage dependencies. Studying chromatin factor functions in leukemia, we show that leukemia cells engage homeostatic chromatin factor functions to block differentiation, generating specific chromatin factor-transcription factor interactions that might be therapeutically targeted. Together, our work elucidates the lineage-determining properties of chromatin factors across normal and malignant hematopoiesis.


Subject(s)
Chromatin , Leukemia , Humans , Chromatin/genetics , Cell Lineage/genetics , Hematopoiesis/genetics , Cell Differentiation/genetics , Transcription Factors/genetics
3.
Nat Commun ; 14(1): 2132, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37059720

ABSTRACT

Resistance to standard and novel therapies remains the main obstacle to cure in acute myeloid leukaemia (AML) and is often driven by metabolic adaptations which are therapeutically actionable. Here we identify inhibition of mannose-6-phosphate isomerase (MPI), the first enzyme in the mannose metabolism pathway, as a sensitizer to both cytarabine and FLT3 inhibitors across multiple AML models. Mechanistically, we identify a connection between mannose metabolism and fatty acid metabolism, that is mediated via preferential activation of the ATF6 arm of the unfolded protein response (UPR). This in turn leads to cellular accumulation of polyunsaturated fatty acids, lipid peroxidation and ferroptotic cell death in AML cells. Our findings provide further support to the role of rewired metabolism in AML therapy resistance, unveil a connection between two apparently independent metabolic pathways and support further efforts to achieve eradication of therapy-resistant AML cells by sensitizing them to ferroptotic cell death.


Subject(s)
Leukemia, Myeloid, Acute , Mannose , Humans , Cell Death , Cytarabine/pharmacology , Cell Line, Tumor , Leukemia, Myeloid, Acute/metabolism , Apoptosis , fms-Like Tyrosine Kinase 3
4.
Blood ; 141(14): 1737-1754, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36577137

ABSTRACT

HOXA9 is commonly upregulated in acute myeloid leukemia (AML), in which it confers a poor prognosis. Characterizing the protein interactome of endogenous HOXA9 in human AML, we identified a chromatin complex of HOXA9 with the nuclear matrix attachment protein SAFB. SAFB perturbation phenocopied HOXA9 knockout to decrease AML proliferation, increase differentiation and apoptosis in vitro, and prolong survival in vivo. Integrated genomic, transcriptomic, and proteomic analyses further demonstrated that the HOXA9-SAFB (H9SB)-chromatin complex associates with nucleosome remodeling and histone deacetylase (NuRD) and HP1γ to repress the expression of factors associated with differentiation and apoptosis, including NOTCH1, CEBPδ, S100A8, and CDKN1A. Chemical or genetic perturbation of NuRD and HP1γ-associated catalytic activity also triggered differentiation, apoptosis, and the induction of these tumor-suppressive genes. Importantly, this mechanism is operative in other HOXA9-dependent AML genotypes. This mechanistic insight demonstrates the active HOXA9-dependent differentiation block as a potent mechanism of disease maintenance in AML that may be amenable to therapeutic intervention by targeting the H9SB interface and/or NuRD and HP1γ activity.


Subject(s)
Leukemia, Myeloid, Acute , Matrix Attachment Region Binding Proteins , Humans , Proteomics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Transcription Factors/genetics , Nuclear Matrix-Associated Proteins , Chromatin , Receptors, Estrogen/genetics , Receptors, Estrogen/therapeutic use , Matrix Attachment Region Binding Proteins/genetics
5.
Sci Adv ; 8(31): eabn4886, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35921412

ABSTRACT

Transcriptional variability facilitates stochastic cell diversification and can in turn underpin adaptation to stress or injury. We hypothesize that it may analogously facilitate progression of premalignancy to cancer. To investigate this, we initiated preleukemia in mouse cells with enhanced transcriptional variability due to conditional disruption of the histone lysine acetyltransferase gene Kat2a. By combining single-cell RNA sequencing of preleukemia with functional analysis of transformation, we show that Kat2a loss results in global variegation of cell identity and accumulation of preleukemic cells. Leukemia progression is subsequently facilitated by destabilization of ribosome biogenesis and protein synthesis, which confer a transient transformation advantage. The contribution of transcriptional variability to early cancer evolution reflects a generic role in promoting cell fate transitions, which, in the case of well-adapted malignancies, contrastingly differentiates and depletes cancer stem cells. That is, transcriptional variability confers forward momentum to cell fate systems, with differential multistage impact throughout cancer evolution.


Subject(s)
Leukemia , Preleukemia , Animals , Cell Differentiation , Leukemia/genetics , Mice , Preleukemia/genetics , Preleukemia/pathology , Protein Biosynthesis
6.
Blood ; 139(16): 2471-2482, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35134130

ABSTRACT

The accessibility of cell surface proteins makes them tractable for targeting by cancer immunotherapy, but identifying suitable targets remains challenging. Here we describe plasma membrane profiling of primary human myeloma cells to identify an unprecedented number of cell surface proteins of a primary cancer. We used a novel approach to prioritize immunotherapy targets and identified a cell surface protein not previously implicated in myeloma, semaphorin-4A (SEMA4A). Using knock-down by short-hairpin RNA and CRISPR/nuclease-dead Cas9 (dCas9), we show that expression of SEMA4A is essential for normal myeloma cell growth in vitro, indicating that myeloma cells cannot downregulate the protein to avoid detection. We further show that SEMA4A would not be identified as a myeloma therapeutic target by standard CRISPR/Cas9 knockout screens because of exon skipping. Finally, we potently and selectively targeted SEMA4A with a novel antibody-drug conjugate in vitro and in vivo.


Subject(s)
Multiple Myeloma , Semaphorins , Cell Membrane/metabolism , Humans , Immunologic Factors , Immunotherapy , Membrane Proteins , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Proteomics , Semaphorins/genetics , Semaphorins/metabolism
7.
Blood Adv ; 6(1): 165-180, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34654054

ABSTRACT

Epigenetic histone modifiers are key regulators of cell fate decisions in normal and malignant hematopoiesis. Their enzymatic activities are of particular significance as putative therapeutic targets in leukemia. In contrast, less is known about the contextual role in which those enzymatic activities are exercised and specifically how different macromolecular complexes configure the same enzymatic activity with distinct molecular and cellular consequences. We focus on KAT2A, a lysine acetyltransferase responsible for histone H3 lysine 9 acetylation, which we recently identified as a dependence in acute myeloid leukemia stem cells and that participates in 2 distinct macromolecular complexes: Ada two-A-containing (ATAC) and Spt-Ada-Gcn5-Acetyltransferase (SAGA). Through analysis of human cord blood hematopoietic stem cells and progenitors, and of myeloid leukemia cells, we identify unique respective contributions of the ATAC complex to regulation of biosynthetic activity in undifferentiated self-renewing cells and of the SAGA complex to stabilization or correct progression of cell type-specific programs with putative preservation of cell identity. Cell type and stage-specific dependencies on ATAC and SAGA-regulated programs explain multilevel KAT2A requirements in leukemia and in erythroid lineage specification and development. Importantly, they set a paradigm against which lineage specification and identity can be explored across developmental stem cell systems.


Subject(s)
Histone Acetyltransferases , Leukemia, Myeloid, Acute , Acetylation , Hematopoiesis , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism
8.
Nat Genet ; 53(10): 1443-1455, 2021 10.
Article in English | MEDLINE | ID: mdl-34556857

ABSTRACT

Altered transcription is a cardinal feature of acute myeloid leukemia (AML); however, exactly how mutations synergize to remodel the epigenetic landscape and rewire three-dimensional DNA topology is unknown. Here, we apply an integrated genomic approach to a murine allelic series that models the two most common mutations in AML: Flt3-ITD and Npm1c. We then deconvolute the contribution of each mutation to alterations of the epigenetic landscape and genome organization, and infer how mutations synergize in the induction of AML. Our studies demonstrate that Flt3-ITD signals to chromatin to alter the epigenetic environment and synergizes with mutations in Npm1c to alter gene expression and drive leukemia induction. These analyses also allow the identification of long-range cis-regulatory circuits, including a previously unknown superenhancer of Hoxa locus, as well as larger and more detailed gene-regulatory networks, driven by transcription factors including PU.1 and IRF8, whose importance we demonstrate through perturbation of network members.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA, Neoplasm/chemistry , Gene Expression Regulation, Leukemic , Histones/metabolism , Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Protein Processing, Post-Translational , Animals , Base Sequence , Disease Models, Animal , Enhancer Elements, Genetic/genetics , Gene Regulatory Networks , Genetic Loci , Humans , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Nucleophosmin , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , fms-Like Tyrosine Kinase 3/metabolism
9.
Mol Cell ; 81(19): 4059-4075.e11, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34437837

ABSTRACT

DDX3X is a ubiquitously expressed RNA helicase involved in multiple stages of RNA biogenesis. DDX3X is frequently mutated in Burkitt lymphoma, but the functional basis for this is unknown. Here, we show that loss-of-function DDX3X mutations are also enriched in MYC-translocated diffuse large B cell lymphoma and reveal functional cooperation between mutant DDX3X and MYC. DDX3X promotes the translation of mRNA encoding components of the core translational machinery, thereby driving global protein synthesis. Loss-of-function DDX3X mutations moderate MYC-driven global protein synthesis, thereby buffering MYC-induced proteotoxic stress during early lymphomagenesis. Established lymphoma cells restore full protein synthetic capacity by aberrant expression of DDX3Y, a Y chromosome homolog, the expression of which is normally restricted to the testis. These findings show that DDX3X loss of function can buffer MYC-driven proteotoxic stress and highlight the capacity of male B cell lymphomas to then compensate for this loss by ectopic DDX3Y expression.


Subject(s)
B-Lymphocytes/enzymology , DEAD-box RNA Helicases/metabolism , Lymphoma, B-Cell/enzymology , Minor Histocompatibility Antigens/metabolism , Neoplasm Proteins/biosynthesis , Proto-Oncogene Proteins c-myc/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , B-Lymphocytes/pathology , Cell Line, Tumor , Child , Child, Preschool , DEAD-box RNA Helicases/genetics , Endoplasmic Reticulum Stress , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Loss of Function Mutation , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Male , Mice, Transgenic , Middle Aged , Minor Histocompatibility Antigens/genetics , Neoplasm Proteins/genetics , Protein Biosynthesis , Proteome , Proteostasis , Proto-Oncogene Proteins c-myc/genetics , Young Adult
11.
Nat Commun ; 11(1): 1407, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32179751

ABSTRACT

Leukaemogenic mutations commonly disrupt cellular differentiation and/or enhance proliferation, thus perturbing the regulatory programs that control self-renewal and differentiation of stem and progenitor cells. Translocations involving the Mll1 (Kmt2a) gene generate powerful oncogenic fusion proteins, predominantly affecting infant and paediatric AML and ALL patients. The early stages of leukaemogenic transformation are typically inaccessible from human patients and conventional mouse models. Here, we take advantage of cells conditionally blocked at the multipotent haematopoietic progenitor stage to develop a MLL-r model capturing early cellular and molecular consequences of MLL-ENL expression based on a clear clonal relationship between parental and leukaemic cells. Through a combination of scRNA-seq, ATAC-seq and genome-scale CRISPR-Cas9 screening, we identify pathways and genes likely to drive the early phases of leukaemogenesis. Finally, we demonstrate the broad utility of using matched parental and transformed cells for small molecule inhibitor studies by validating both previously known and other potential therapeutic targets.


Subject(s)
Cell Transformation, Neoplastic , Histone-Lysine N-Methyltransferase/metabolism , Leukemia, Myeloid, Acute/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Hematopoietic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/physiopathology , Mice , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Elife ; 92020 01 27.
Article in English | MEDLINE | ID: mdl-31985402

ABSTRACT

Acute Myeloid Leukemia (AML) is an aggressive hematological malignancy with abnormal progenitor self-renewal and defective white blood cell differentiation. Its pathogenesis comprises subversion of transcriptional regulation, through mutation and by hijacking normal chromatin regulation. Kat2a is a histone acetyltransferase central to promoter activity, that we recently associated with stability of pluripotency networks, and identified as a genetic vulnerability in AML. Through combined chromatin profiling and single-cell transcriptomics of a conditional knockout mouse, we demonstrate that Kat2a contributes to leukemia propagation through preservation of leukemia stem-like cells. Kat2a loss impacts transcription factor binding and reduces transcriptional burst frequency in a subset of gene promoters, generating enhanced variability of transcript levels. Destabilization of target programs shifts leukemia cell fate out of self-renewal into differentiation. We propose that control of transcriptional variability is central to leukemia stem-like cell propagation, and establish a paradigm exploitable in different tumors and distinct stages of cancer evolution.


Less than 30% of patients with acute myeloid leukaemia ­ an aggressive cancer of the white blood cells ­ survive five years post-diagnosis. This disease disrupts the maturation of white blood cells, resulting in the accumulation of immature cells that multiply and survive but are incapable of completing their maturation process. Amongst these, a group of cancer cells known as leukemic stem cells is responsible for continually replenishing the leukaemia, thus perpetuating its growth. Cancers develop when cells in the body acquire changes or mutations to their genetic makeup. The mutations that lead to acute myeloid leukaemia often affect the activity of genes known as epigenetic regulators. These genes regulate which proteins and other molecules cells make by controlling the way in which cells 'read' their genetic instructions. The epigenetic regulator Kat2a is thought to 'tune' the frequency at which cells read their genetic instructions. This tuning mechanism decreases random fluctuations in the execution of the instructions cells receive to make proteins and other molecules. In turn, this helps to ensure that individual cells of the same type behave in a similar way, for example by keeping leukaemia cells in an immature state. Here, Domingues, Kulkarni et al. investigated whether interfering with Kat2a can make acute myeloid leukaemia less aggressive by allowing the immature white blood cells to mature. Domingues, Kulkarni et al. genetically engineered mice to remove Kat2a from blood cells on demand and then inserted a mutation that causes acute myeloid leukaemia. The experiments showed that the loss of Kat2a delayed the development of leukaemia in the mice and progressively depleted leukaemia stem cells, causing the disease to become less aggressive. The results also showed that loss of Kat2a caused more fluctuations in how the white blood cells read their genetic code, which resulted in more variability in the molecules they produced and increased the tendency of the cells to mature. These findings establish that loss of Kat2a causes leukaemia stem cells to mature and stop multiplying by untuning the frequency at which the cells read their genetic instructions. In the future, it may be possible to develop drugs that target human KAT2A to treat acute myeloid leukaemia.


Subject(s)
Histone Acetyltransferases , Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Animals , Chromatin/genetics , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Humans , Mice , Mice, Knockout , Single-Cell Analysis , Transcription, Genetic/genetics , Transcriptome/genetics
13.
Cancers (Basel) ; 11(12)2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31817495

ABSTRACT

Approximately 18% of acute myeloid leukemia (AML) cases express a fusion transcript. However, few fusions are recurrent across AML and the identification of these rare chimeras is of interest to characterize AML patients. Here, we studied the transcriptome of 8 adult AML patients with poorly described chromosomal translocation(s), with the aim of identifying novel and rare fusion transcripts. We integrated RNA-sequencing data with multiple approaches including computational analysis, Sanger sequencing, fluorescence in situ hybridization and in vitro studies to assess the oncogenic potential of the ZEB2-BCL11B chimera. We detected 7 different fusions with partner genes involving transcription factors (OAZ-MAFK, ZEB2-BCL11B), tumor suppressors (SAV1-GYPB, PUF60-TYW1, CNOT2-WT1) and rearrangements associated with the loss of NF1 (CPD-PXT1, UTP6-CRLF3). Notably, ZEB2-BCL11B rearrangements co-occurred with FLT3 mutations and were associated with a poorly differentiated or mixed phenotype leukemia. Although the fusion alone did not transform murine c-Kit+ bone marrow cells, 45.4% of 14q32 non-rearranged AML cases were also BCL11B-positive, suggesting a more general and complex mechanism of leukemogenesis associated with BCL11B expression. Overall, by combining different approaches, we described rare fusion events contributing to the complexity of AML and we linked the expression of some chimeras to genomic alterations hitting known genes in AML.

14.
Blood ; 134(24): 2195-2208, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31515253

ABSTRACT

Cohesin complex disruption alters gene expression, and cohesin mutations are common in myeloid neoplasia, suggesting a critical role in hematopoiesis. Here, we explore cohesin dynamics and regulation of hematopoietic stem cell homeostasis and differentiation. Cohesin binding increases at active regulatory elements only during erythroid differentiation. Prior binding of the repressive Ets transcription factor Etv6 predicts cohesin binding at these elements and Etv6 interacts with cohesin at chromatin. Depletion of cohesin severely impairs erythroid differentiation, particularly at Etv6-prebound loci, but augments self-renewal programs. Together with corroborative findings in acute myeloid leukemia and myelodysplastic syndrome patient samples, these data suggest cohesin-mediated alleviation of Etv6 repression is required for dynamic expression at critical erythroid genes during differentiation and how this may be perturbed in myeloid malignancies.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Leukemic , Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Biomarkers, Tumor , Cell Line, Tumor , Female , Gene Dosage , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Histones/metabolism , Humans , Male , Myeloproliferative Disorders/diagnosis , Neoplasm Grading , Protein Binding , Proto-Oncogene Proteins c-ets/metabolism , Regulatory Sequences, Nucleic Acid , Repressor Proteins/metabolism , Cohesins , ETS Translocation Variant 6 Protein
15.
J Exp Med ; 216(4): 966-981, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30890554

ABSTRACT

Epigenetic regulators, such as EZH2, are frequently mutated in cancer, and loss-of-function EZH2 mutations are common in myeloid malignancies. We have examined the importance of cellular context for Ezh2 loss during the evolution of acute myeloid leukemia (AML), where we observed stage-specific and diametrically opposite functions for Ezh2 at the early and late stages of disease. During disease maintenance, WT Ezh2 exerts an oncogenic function that may be therapeutically targeted. In contrast, Ezh2 acts as a tumor suppressor during AML induction. Transcriptional analysis explains this apparent paradox, demonstrating that loss of Ezh2 derepresses different expression programs during disease induction and maintenance. During disease induction, Ezh2 loss derepresses a subset of bivalent promoters that resolve toward gene activation, inducing a feto-oncogenic program that includes genes such as Plag1, whose overexpression phenocopies Ezh2 loss to accelerate AML induction in mouse models. Our data highlight the importance of cellular context and disease phase for the function of Ezh2 and its potential therapeutic implications.


Subject(s)
Disease Progression , Enhancer of Zeste Homolog 2 Protein/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Loss of Function Mutation , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Line, Tumor , Cohort Studies , Disease Models, Animal , Gene Frequency , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Prognosis , Survival Rate , Transduction, Genetic , Transplantation, Homologous
16.
Cancer Cell ; 34(4): 533-535, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30300577

ABSTRACT

In this issue of Cancer Cell, de Boer et al. refine a set of acute myeloid leukemia (AML)-enriched plasma membrane markers that can be used to identify, prospectively isolate, and longitudinally track leukemic subclones within individual AML patients, correlating immunophenotypic profiles with specific mutational signatures, transcription, functional behavior, and therapeutic outcomes.


Subject(s)
Leukemia, Myeloid, Acute , Cells, Cultured , Humans , Mutation , Prospective Studies
17.
Blood ; 131(15): 1639-1653, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29463564

ABSTRACT

FLT3 internal tandem duplication (FLT3ITD) mutations are common in acute myeloid leukemia (AML) associated with poor patient prognosis. Although new-generation FLT3 tyrosine kinase inhibitors (TKI) have shown promising results, the outcome of FLT3ITD AML patients remains poor and demands the identification of novel, specific, and validated therapeutic targets for this highly aggressive AML subtype. Utilizing an unbiased genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 screen, we identify GLS, the first enzyme in glutamine metabolism, as synthetically lethal with FLT3-TKI treatment. Using complementary metabolomic and gene-expression analysis, we demonstrate that glutamine metabolism, through its ability to support both mitochondrial function and cellular redox metabolism, becomes a metabolic dependency of FLT3ITD AML, specifically unmasked by FLT3-TKI treatment. We extend these findings to AML subtypes driven by other tyrosine kinase (TK) activating mutations and validate the role of GLS as a clinically actionable therapeutic target in both primary AML and in vivo models. Our work highlights the role of metabolic adaptations as a resistance mechanism to several TKI and suggests glutaminolysis as a therapeutically targetable vulnerability when combined with specific TKI in FLT3ITD and other TK activating mutation-driven leukemias.


Subject(s)
Glutamine/metabolism , Leukemia, Myeloid, Acute , Mutation , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3 , CRISPR-Cas Systems , Enzyme Activation/drug effects , Enzyme Activation/genetics , Genome-Wide Association Study , Glutamine/genetics , Humans , K562 Cells , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , THP-1 Cells , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
18.
Cancer Cell ; 33(2): 274-291.e8, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29438697

ABSTRACT

Lympho-myeloid restricted early thymic progenitors (ETPs) are postulated to be the cell of origin for ETP leukemias, a therapy-resistant leukemia associated with frequent co-occurrence of EZH2 and RUNX1 inactivating mutations, and constitutively activating signaling pathway mutations. In a mouse model, we demonstrate that Ezh2 and Runx1 inactivation targeted to early lymphoid progenitors causes a marked expansion of pre-leukemic ETPs, showing transcriptional signatures characteristic of ETP leukemia. Addition of a RAS-signaling pathway mutation (Flt3-ITD) results in an aggressive leukemia co-expressing myeloid and lymphoid genes, which can be established and propagated in vivo by the expanded ETPs. Both mouse and human ETP leukemias show sensitivity to BET inhibition in vitro and in vivo, which reverses aberrant gene expression induced by Ezh2 inactivation.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Animals , Gene Expression Regulation, Leukemic , Mice, Knockout , Myeloid Cells/metabolism , Signal Transduction/genetics , Stem Cells
19.
Nat Cell Biol ; 19(9): 1093-1104, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28825697

ABSTRACT

Loss-of-function mutations of cyclic-AMP response element binding protein, binding protein (CREBBP) are prevalent in lymphoid malignancies. However, the tumour suppressor functions of CREBBP remain unclear. We demonstrate that loss of Crebbp in murine haematopoietic stem and progenitor cells (HSPCs) leads to increased development of B-cell lymphomas. This is preceded by accumulation of hyperproliferative lymphoid progenitors with a defective DNA damage response (DDR) due to a failure to acetylate p53. We identify a premalignant lymphoma stem cell population with decreased H3K27ac, which undergoes transcriptional and genetic evolution due to the altered DDR, resulting in lymphomagenesis. Importantly, when Crebbp is lost later in lymphopoiesis, cellular abnormalities are lost and tumour generation is attenuated. We also document that CREBBP mutations may occur in HSPCs from patients with CREBBP-mutated lymphoma. These data suggest that earlier loss of Crebbp is advantageous for lymphoid transformation and inform the cellular origins and subsequent evolution of lymphoid malignancies.


Subject(s)
CREB-Binding Protein/deficiency , CREB-Binding Protein/metabolism , Cell Transformation, Neoplastic/metabolism , Lymphoid Progenitor Cells/metabolism , Lymphoma/metabolism , Neoplastic Stem Cells/metabolism , Acetylation , Animals , CREB-Binding Protein/genetics , Cell Proliferation , Cell Self Renewal , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , DNA Damage , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Histones/metabolism , Lymphangiogenesis , Lymphoid Progenitor Cells/pathology , Lymphoma/genetics , Lymphoma/pathology , Lymphopoiesis , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neoplastic Stem Cells/pathology , Phenotype , Signal Transduction , Time Factors , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
20.
Cell Rep ; 16(4): 1039-1054, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27396339

ABSTRACT

MLL-AF4+ infant B cell acute lymphoblastic leukemia is characterized by an early onset and dismal survival. It initiates before birth, and very little is known about the early stages of the disease's development. Using a conditional Mll-AF4-expressing mouse model in which fusion expression is targeted to the earliest definitive hematopoietic cells generated in the mouse embryo, we demonstrate that Mll-AF4 imparts enhanced B lymphoid potential and increases repopulation and self-renewal capacity during a putative pre-leukemic state. This occurs between embryonic days 12 and 14 and manifests itself most strongly in the lymphoid-primed multipotent progenitor population, thus pointing to a window of opportunity and a potential cell of origin. However, this state alone is insufficient to generate disease, with the mice succumbing to B cell lymphomas only after a long latency. Future analysis of the molecular details of this pre-leukemic state will shed light on additional events required for progression to acute leukemia.


Subject(s)
Cell Self Renewal/physiology , Lymphocytes/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Animals , Leukemia/metabolism , Lymphoma, B-Cell/metabolism , Mice , Mice, Transgenic , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...