Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
3.
Leukemia ; 37(2): 370-378, 2023 02.
Article in English | MEDLINE | ID: mdl-36309559

ABSTRACT

In acute myeloid leukemia (AML), p53 tumor suppressor activity can be reduced due to enhanced expression of MDM2 which promotes the degradation of p53. In TP53 wild-type malignancies, therapy with small molecule antagonists of MDM2 results in antileukemic activity. Current treatment strategies, however, have been limited by poor tolerability and incomplete clinical activity. We have developed a proteolysis-targeting chimera (PROTAC) MS3227 that targets MDM2 by recruiting the E3 ligase Von Hippel-Lindau, resulting in proteasome-dependent degradation of MDM2. In WT TP53 leukemia cell lines, MS3227 led to activation of p53 targets p21, PUMA, and MDM2 and resulted in cell-cycle arrest, apoptosis, and decreased viability. The catalytic PROTAC MS3227 led to more potent activation when compared to a stoichiometric inhibitor, in part by dampening the negative feedback mechanism in the p53 - MDM2 circuit. The effectiveness of MS3227 was also observed in primary patient specimens with selectivity towards leukemic blasts. The addition of MS3227 enhanced the activity of other anti-leukemic agents including azacytidine, cytarabine, and venetoclax. In particular, MS3227 treatment was shown to downregulate MCL-1, a known mediator of resistance to venetoclax. A PROTAC-based approach may provide a means of improving MDM2 inhibition to gain greater therapeutic potential in AML.


Subject(s)
Leukemia, Myeloid, Acute , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Apoptosis , Cell Line, Tumor
4.
Hemasphere ; 5(4): e549, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33718803

ABSTRACT

Hypomethylating agents (HMAs) in combination with venetoclax have been widely adopted as the standard of care for patients who cannot tolerate induction chemotherapy and for patients who have relapsed/refractory (R/R) acute myeloid leukemia (AML). This study retrospectively analyzed the outcomes of all patients with AML (n = 65) or myelodysplastic syndrome (n = 7) who received the combination of HMA and venetoclax at our institution. Outcomes measured included complete remission (CR) and CR with incomplete hematologic recovery (CRi) rates, duration of response (DOR), and overall survival (OS). Patient mutational profiles and transfusion requirements were also assessed. Of 26 newly diagnosed AML patients, the CR/CRi rate was 53.8%. The median DOR and OS were 6.9 months and not reached, respectively. Of 39 R/R AML patients, the CR/CRi rate was 38.5%. The median DOR and OS were both 8.1 months. Responders to HMA and venetoclax were enriched for TET2, IDH1, and IDH2 mutations, while nonresponders were associated with FLT3 and RAS mutations. Adaptive resistance was observed through various mechanisms including acquired RAS pathway mutations. Of transfusion-dependent patients, 12.2% and 15.2% achieved red blood cell (RBC) and platelet transfusion independence, respectively, while 44.8% and 35.1% of RBC and platelet transfusion independent patients, respectively, became transfusion dependent. In total 59.1% of patients developed a ≥grade 3 infection and 46.5% neutropenic fever. HMA + venetoclax can lead to impressive response rates with moderately durable remissions and survival. However, the benefits of this combination are diminished by the significant toxicities from infection, persistent cytopenias, and transfusion requirements.

5.
Hematol Oncol Clin North Am ; 35(2): 253-265, 2021 04.
Article in English | MEDLINE | ID: mdl-33641867

ABSTRACT

Myeloproliferative neoplasms, such as polycythemia vera, essential thrombocythemia, and primary myelofibrosis, are bone marrow disorders that result in the overproduction of mature clonal myeloid elements. Identification of recurrent genetic mutations has been described and aid in diagnosis and prognostic determination. Mouse models of these mutations have confirmed the biologic significance of these mutations in myeloproliferative neoplasm disease biology and provided greater insights on the pathways that are dysregulated with each mutation. The models are useful tools that have led to preclinical testing and provided data as validation for future myeloproliferative neoplasm clinical trials.


Subject(s)
Disease Models, Animal , Myeloproliferative Disorders , Neoplasms , Animals , Mice , Mutation , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Neoplasms/diagnosis , Neoplasms/genetics
7.
Cancer Discov ; 10(12): 1934-1949, 2020 12.
Article in English | MEDLINE | ID: mdl-32938585

ABSTRACT

Epigenetic allele diversity is linked to inferior prognosis in acute myeloid leukemia (AML). However, the source of epiallele heterogeneity in AML is unknown. Herein we analyzed epiallele diversity in a genetically and clinically annotated AML cohort. Notably, AML driver mutations linked to transcription factors and favorable outcome are associated with epigenetic destabilization in a defined set of susceptible loci. In contrast, AML subtypes linked to inferior prognosis manifest greater abundance and highly stochastic epiallele patterning. We report an epiallele outcome classifier supporting the link between epigenetic diversity and treatment failure. Mouse models with TET2 or IDH2 mutations show that epiallele diversity is especially strongly induced by IDH mutations, precedes transformation to AML, and is enhanced by cooperation between somatic mutations. Furthermore, epiallele complexity was partially reversed by epigenetic therapies in AML driven by TET2/IDH2, suggesting that epigenetic therapy might function in part by reducing population complexity and fitness of AMLs. SIGNIFICANCE: We show for the first time that epigenetic clonality is directly linked to specific mutations and that epigenetic allele diversity precedes and potentially contributes to malignant transformation. Furthermore, epigenetic clonality is reversible with epigenetic therapy agents.This article is highlighted in the In This Issue feature, p. 1775.


Subject(s)
Epigenesis, Genetic/genetics , Leukemia, Myeloid, Acute/genetics , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Mutation
8.
Cancer Discov ; 9(7): 872-889, 2019 07.
Article in English | MEDLINE | ID: mdl-31076479

ABSTRACT

Disruption of epigenetic regulation is a hallmark of acute myeloid leukemia (AML), but epigenetic therapy is complicated by the complexity of the epigenome. Herein, we developed a long-term primary AML ex vivo platform to determine whether targeting different epigenetic layers with 5-azacytidine and LSD1 inhibitors would yield improved efficacy. This combination was most effective in TET2 mut AML, where it extinguished leukemia stem cells and particularly induced genes with both LSD1-bound enhancers and cytosine-methylated promoters. Functional studies indicated that derepression of genes such as GATA2 contributes to drug efficacy. Mechanistically, combination therapy increased enhancer-promoter looping and chromatin-activating marks at the GATA2 locus. CRISPRi of the LSD1-bound enhancer in patient-derived TET2 mut AML was associated with dampening of therapeutic GATA2 induction. TET2 knockdown in human hematopoietic stem/progenitor cells induced loss of enhancer 5-hydroxymethylation and facilitated LSD1-mediated enhancer inactivation. Our data provide a basis for rational targeting of cooperating aberrant promoter and enhancer epigenetic marks driven by mutant epigenetic modifiers. SIGNIFICANCE: Somatic mutations of genes encoding epigenetic modifiers are a hallmark of AML and potentially disrupt many components of the epigenome. Our study targets two different epigenetic layers at promoters and enhancers that cooperate to aberrant gene silencing, downstream of the actions of a mutant epigenetic regulator.This article is highlighted in the In This Issue feature, p. 813.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Animals , Azacitidine/pharmacology , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA Methylation/drug effects , DNA-Binding Proteins/genetics , Dioxygenases , Enhancer Elements, Genetic , Epigenome , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Genes, Tumor Suppressor , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins/genetics , Random Allocation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Cancer Discov ; 9(6): 756-777, 2019 06.
Article in English | MEDLINE | ID: mdl-30862724

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) is important for reductive carboxylation in cancer cells, and the IDH1 R132H mutation plays a pathogenic role in cancers including acute myeloid leukemia (AML). However, the regulatory mechanisms modulating mutant and/or wild-type (WT) IDH1 function remain unknown. Here, we show that two groups of tyrosine kinases (TK) enhance the activation of mutant and WT IDH1 through preferential Y42 or Y391 phosphorylation. Mechanistically, Y42 phosphorylation occurs in IDH1 monomers, which promotes dimer formation with enhanced substrate (isocitrate or α-ketoglutarate) binding, whereas Y42-phosphorylated dimers show attenuated disruption to monomers. Y391 phosphorylation occurs in both monomeric and dimeric IDH1, which enhances cofactor (NADP+ or NADPH) binding. Diverse oncogenic TKs phosphorylate IDH1 WT at Y42 and activate Src to phosphorylate IDH1 at Y391, which contributes to reductive carboxylation and tumor growth, whereas FLT3 or the FLT3-ITD mutation activates JAK2 to enhance mutant IDH1 activity through phosphorylation of Y391 and Y42, respectively, in AML cells. SIGNIFICANCE: We demonstrated an intrinsic connection between oncogenic TKs and activation of WT and mutant IDH1, which involves distinct TK cascades in related cancers. In particular, these results provide an additional rationale supporting the combination of FLT3 and mutant IDH1 inhibitors as a promising clinical treatment of mutant IDH1-positive AML.See related commentary by Horton and Huntly, p. 699.This article is highlighted in the In This Issue feature, p. 681.


Subject(s)
Isocitrate Dehydrogenase/genetics , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Cell Line, Tumor , Disease Management , Humans , Isocitrate Dehydrogenase/chemistry , Janus Kinase 2/metabolism , Models, Biological , NADP/metabolism , Neoplasms/pathology , Phosphorylation , Protein Binding , Protein Multimerization , fms-Like Tyrosine Kinase 3/genetics
10.
Cancer Discov ; 8(12): 1540-1547, 2018 12.
Article in English | MEDLINE | ID: mdl-30355724

ABSTRACT

Somatic mutations in cytosolic or mitochondrial isoforms of isocitrate dehydrogenase (IDH1 or IDH2, respectively) contribute to oncogenesis via production of the metabolite 2-hydroxyglutarate (2HG). Isoform-selective IDH inhibitors suppress 2HG production and induce clinical responses in patients with IDH1- and IDH2-mutant malignancies. Despite the promising activity of IDH inhibitors, the mechanisms that mediate resistance to IDH inhibition are poorly understood. Here, we describe four clinical cases that identify mutant IDH isoform switching, either from mutant IDH1 to mutant IDH2 or vice versa, as a mechanism of acquired clinical resistance to IDH inhibition in solid and liquid tumors. SIGNIFICANCE: IDH-mutant cancers can develop resistance to isoform-selective IDH inhibition by "isoform switching" from mutant IDH1 to mutant IDH2 or vice versa, thereby restoring 2HG production by the tumor. These findings underscore a role for continued 2HG production in tumor progression and suggest therapeutic strategies to prevent or overcome resistance.This article is highlighted in the In This Issue feature, p. 1494.


Subject(s)
Drug Resistance/genetics , Isocitrate Dehydrogenase/genetics , Mutation , Acute Disease , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Aged , Enzyme Inhibitors/pharmacology , Female , Humans , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/metabolism , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/enzymology , Leukemia, Myeloid/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Male , Middle Aged , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/enzymology , Myelodysplastic Syndromes/genetics
11.
Cancer Discov ; 8(12): 1632-1653, 2018 12.
Article in English | MEDLINE | ID: mdl-30274972

ABSTRACT

TET2 somatic mutations occur in ∼10% of diffuse large B-cell lymphomas (DLBCL) but are of unknown significance. Herein, we show that TET2 is required for the humoral immune response and is a DLBCL tumor suppressor. TET2 loss of function disrupts transit of B cells through germinal centers (GC), causing GC hyperplasia, impaired class switch recombination, blockade of plasma cell differentiation, and a preneoplastic phenotype. TET2 loss was linked to focal loss of enhancer hydroxymethylation and transcriptional repression of genes that mediate GC exit, such as PRDM1. Notably, these enhancers and genes are also repressed in CREBBP-mutant DLBCLs. Accordingly, TET2 mutation in patients yields a CREBBP-mutant gene-expression signature, CREBBP and TET2 mutations are generally mutually exclusive, and hydroxymethylation loss caused by TET2 deficiency impairs enhancer H3K27 acetylation. Hence, TET2 plays a critical role in the GC reaction, and its loss of function results in lymphomagenesis through failure to activate genes linked to GC exit signals. SIGNIFICANCE: We show that TET2 is required for exit of the GC, B-cell differentiation, and is a tumor suppressor for mature B cells. Loss of TET2 phenocopies CREBBP somatic mutation. These results advocate for sequencing TET2 in patients with lymphoma and for the testing of epigenetic therapies to treat these tumors.See related commentary by Shingleton and Dave, p. 1515.This article is highlighted in the In This Issue feature, p. 1494.


Subject(s)
Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Germinal Center/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Plasma Cells/metabolism , Proto-Oncogene Proteins/genetics , Animals , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , DNA-Binding Proteins/metabolism , Dioxygenases , Epigenesis, Genetic/genetics , Gene Expression Profiling/methods , Germinal Center/pathology , Hematopoietic Stem Cells/metabolism , Humans , Hyperplasia , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice, Knockout , Mice, Transgenic , Mutation , Plasma Cells/pathology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Proto-Oncogene Proteins/metabolism
13.
Nature ; 559(7712): 125-129, 2018 07.
Article in English | MEDLINE | ID: mdl-29950729

ABSTRACT

Somatic mutations in the isocitrate dehydrogenase 2 gene (IDH2) contribute to the pathogenesis of acute myeloid leukaemia (AML) through the production of the oncometabolite 2-hydroxyglutarate (2HG)1-8. Enasidenib (AG-221) is an allosteric inhibitor that binds to the IDH2 dimer interface and blocks the production of 2HG by IDH2 mutants9,10. In a phase I/II clinical trial, enasidenib inhibited the production of 2HG and induced clinical responses in relapsed or refractory IDH2-mutant AML11. Here we describe two patients with IDH2-mutant AML who had a clinical response to enasidenib followed by clinical resistance, disease progression, and a recurrent increase in circulating levels of 2HG. We show that therapeutic resistance is associated with the emergence of second-site IDH2 mutations in trans, such that the resistance mutations occurred in the IDH2 allele without the neomorphic R140Q mutation. The in trans mutations occurred at glutamine 316 (Q316E) and isoleucine 319 (I319M), which are at the interface where enasidenib binds to the IDH2 dimer. The expression of either of these mutant disease alleles alone did not induce the production of 2HG; however, the expression of the Q316E or I319M mutation together with the R140Q mutation in trans allowed 2HG production that was resistant to inhibition by enasidenib. Biochemical studies predicted that resistance to allosteric IDH inhibitors could also occur via IDH dimer-interface mutations in cis, which was confirmed in a patient with acquired resistance to the IDH1 inhibitor ivosidenib (AG-120). Our observations uncover a mechanism of acquired resistance to a targeted therapy and underscore the importance of 2HG production in the pathogenesis of IDH-mutant malignancies.


Subject(s)
Aminopyridines/pharmacology , Drug Resistance, Neoplasm/genetics , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , Mutant Proteins/genetics , Mutation , Protein Multimerization/genetics , Triazines/pharmacology , Alleles , Allosteric Site/drug effects , Allosteric Site/genetics , Aminopyridines/chemistry , Aminopyridines/therapeutic use , Animals , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Disease Progression , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Glutamine/genetics , Glutarates/blood , Glutarates/metabolism , HEK293 Cells , Humans , Isoleucine/genetics , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/drug therapy , Mice , Mice, Inbred C57BL , Models, Molecular , Mutant Proteins/antagonists & inhibitors , Triazines/chemistry , Triazines/therapeutic use
14.
J Clin Invest ; 128(2): 789-804, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29355841

ABSTRACT

Patients with myeloproliferative neoplasms (MPNs) frequently progress to bone marrow failure or acute myeloid leukemia (AML), and mutations in epigenetic regulators such as the metabolic enzyme isocitrate dehydrogenase (IDH) are associated with poor outcomes. Here, we showed that combined expression of Jak2V617F and mutant IDH1R132H or Idh2R140Q induces MPN progression, alters stem/progenitor cell function, and impairs differentiation in mice. Jak2V617F Idh2R140Q-mutant MPNs were sensitive to small-molecule inhibition of IDH. Combined inhibition of JAK2 and IDH2 normalized the stem and progenitor cell compartments in the murine model and reduced disease burden to a greater extent than was seen with JAK inhibition alone. In addition, combined JAK2 and IDH2 inhibitor treatment also reversed aberrant gene expression in MPN stem cells and reversed the metabolite perturbations induced by concurrent JAK2 and IDH2 mutations. Combined JAK2 and IDH2 inhibitor therapy also showed cooperative efficacy in cells from MPN patients with both JAK2mut and IDH2mut mutations. Taken together, these data suggest that combined JAK and IDH inhibition may offer a therapeutic advantage in this high-risk MPN subtype.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Isocitrate Dehydrogenase/genetics , Janus Kinase 2/genetics , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Aged , Animals , Disease Progression , Epigenesis, Genetic , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Middle Aged , Mutation , Phenotype , Stem Cells
15.
Cancer Cell ; 33(1): 44-59.e8, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29275866

ABSTRACT

Mutations in epigenetic modifiers and signaling factors often co-occur in myeloid malignancies, including TET2 and NRAS mutations. Concurrent Tet2 loss and NrasG12D expression in hematopoietic cells induced myeloid transformation, with a fully penetrant, lethal chronic myelomonocytic leukemia (CMML), which was serially transplantable. Tet2 loss and Nras mutation cooperatively led to decrease in negative regulators of mitogen-activated protein kinase (MAPK) activation, including Spry2, thereby causing synergistic activation of MAPK signaling by epigenetic silencing. Tet2/Nras double-mutant leukemia showed preferential sensitivity to MAPK kinase (MEK) inhibition in both mouse model and patient samples. These data provide insights into how epigenetic and signaling mutations cooperate in myeloid transformation and provide a rationale for mechanism-based therapy in CMML patients with these high-risk genetic lesions.


Subject(s)
DNA-Binding Proteins/genetics , GTP Phosphohydrolases/genetics , Leukemia, Myelomonocytic, Chronic/genetics , Membrane Proteins/genetics , Monomeric GTP-Binding Proteins/genetics , Mutation/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Transformation, Neoplastic/genetics , Dioxygenases , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice, Transgenic , Myeloproliferative Disorders/genetics , Protein Serine-Threonine Kinases , Signal Transduction/genetics
16.
Cancer Discov ; 7(5): 494-505, 2017 05.
Article in English | MEDLINE | ID: mdl-28193779

ABSTRACT

Genomic studies in acute myeloid leukemias (AML) have identified mutations that drive altered DNA methylation, including TET2 and IDH2 Here, we show that models of AML resulting from TET2 or IDH2 mutations combined with FLT3ITD mutations are sensitive to 5-azacytidine or to the IDH2 inhibitor AG-221, respectively. 5-azacytidine and AG-221 treatment induced an attenuation of aberrant DNA methylation and transcriptional output and resulted in a reduction in leukemic blasts consistent with antileukemic activity. These therapeutic benefits were associated with restoration of leukemic cell differentiation, and the normalization of hematopoiesis was derived from mutant cells. By contrast, combining AG-221 or 5-azacytidine with FLT3 inhibition resulted in a reduction in mutant allele burden, progressive recovery of normal hematopoiesis from non-mutant stem-progenitor cells, and reversal of dysregulated DNA methylation and transcriptional output. Together, our studies suggest combined targeting of signaling and epigenetic pathways can increase therapeutic response in AML.Significance: AMLs with mutations in TET2 or IDH2 are sensitive to epigenetic therapy through inhibition of DNA methyltransferase activity by 5-azacytidine or inhibition of mutant IDH2 through AG-221. These inhibitors induce a differentiation response and can be used to inform mechanism-based combination therapy. Cancer Discov; 7(5); 494-505. ©2017 AACR.See related commentary by Thomas and Majeti, p. 459See related article by Yen et al., p. 478This article is highlighted in the In This Issue feature, p. 443.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , DNA-Binding Proteins/genetics , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/drug therapy , Molecular Targeted Therapy/methods , Proto-Oncogene Proteins/genetics , Aminopyridines/pharmacology , Animals , Azacitidine/pharmacology , DNA Methylation/drug effects , Dioxygenases , Epigenesis, Genetic/drug effects , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Mutant Strains , Mutation , Signal Transduction/drug effects , Triazines/pharmacology , fms-Like Tyrosine Kinase 3/genetics
17.
Blood ; 129(13): 1779-1790, 2017 Mar 30.
Article in English | MEDLINE | ID: mdl-28077417

ABSTRACT

Recent studies have reported that activation-induced cytidine deaminase (AID) and ten-eleven-translocation (TET) family members regulate active DNA demethylation. Genetic alterations of TET2 occur in myeloid malignancies, and hematopoietic-specific loss of Tet2 induces aberrant hematopoietic stem cell (HSC) self-renewal/differentiation, implicating TET2 as a master regulator of normal and malignant hematopoiesis. Despite the functional link between AID and TET in epigenetic gene regulation, the role of AID loss in hematopoiesis and myeloid transformation remains to be investigated. Here, we show that Aid loss in mice leads to expansion of myeloid cells and reduced erythroid progenitors resulting in anemia, with dysregulated expression of Cebpa and Gata1, myeloid/erythroid lineage-specific transcription factors. Consistent with data in the murine context, silencing of AID in human bone marrow cells skews differentiation toward myelomonocytic lineage. However, in contrast to Tet2 loss, Aid loss does not contribute to enhanced HSC self-renewal or cooperate with Flt3-ITD to induce myeloid transformation. Genome-wide transcription and differential methylation analysis uncover the critical role of Aid as a key epigenetic regulator. These results indicate that AID and TET2 share common effects on myeloid and erythroid lineage differentiation, however, their role is nonredundant in regulating HSC self-renewal and in myeloid transformation.


Subject(s)
Cell Differentiation , Cytidine Deaminase/physiology , DNA Methylation , Hematopoietic Stem Cells/metabolism , Animals , Cell Lineage , Cell Self Renewal , Cell Transformation, Neoplastic , Cytidine Deaminase/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Dioxygenases , Erythroid Cells/cytology , Gene Silencing , Hematopoietic Stem Cells/cytology , Humans , Mice , Myeloid Cells/cytology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology
18.
Mol Cell ; 63(4): 711-719, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27477909

ABSTRACT

We present a highly sensitive and selective chemical labeling and capture approach for genome-wide profiling of 5-hydroxylmethylcytosine (5hmC) using DNA isolated from ∼1,000 cells (nano-hmC-Seal). Using this technology, we assessed 5hmC occupancy and dynamics across different stages of hematopoietic differentiation. Nano-hmC-Seal profiling of purified Tet2-mutant acute myeloid leukemia (AML) murine stem cells allowed us to identify leukemia-specific, differentially hydroxymethylated regions that harbor known and candidate disease-specific target genes with differential 5hmC peaks compared to normal stem cells. The change of 5hmC patterns in AML strongly correlates with differential gene expression, demonstrating the importance of dynamic alterations of 5hmC in regulating transcription in AML. Together, covalent 5hmC labeling offers an effective approach to study and detect DNA methylation dynamics in in vivo disease models and in limited clinical samples.


Subject(s)
5-Methylcytosine/analogs & derivatives , DNA Methylation , Epigenesis, Genetic , Gene Expression Profiling/methods , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , High-Throughput Nucleotide Sequencing/methods , Leukemia, Promyelocytic, Acute/genetics , 5-Methylcytosine/metabolism , Animals , Cells, Cultured , Computational Biology , DNA-Binding Proteins/genetics , Databases, Genetic , Dioxygenases , Gene Expression Regulation, Neoplastic , Gene Library , Genome-Wide Association Study , Leukemia, Promyelocytic, Acute/metabolism , Mice , Mutation , Nanotechnology , Proto-Oncogene Proteins/genetics , Time Factors , fms-Like Tyrosine Kinase 3/genetics
19.
Cancer Cell ; 30(2): 337-348, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27424808

ABSTRACT

Mutations in the isocitrate dehydrogenase-1 gene (IDH1) are common drivers of acute myeloid leukemia (AML) but their mechanism is not fully understood. It is thought that IDH1 mutants act by inhibiting TET2 to alter DNA methylation, but there are significant unexplained clinical differences between IDH1- and TET2-mutant diseases. We have discovered that mice expressing endogenous mutant IDH1 have reduced numbers of hematopoietic stem cells (HSCs), in contrast to Tet2 knockout (TET2-KO) mice. Mutant IDH1 downregulates the DNA damage (DD) sensor ATM by altering histone methylation, leading to impaired DNA repair, increased sensitivity to DD, and reduced HSC self-renewal, independent of TET2. ATM expression is also decreased in human IDH1-mutated AML. These findings may have implications for treatment of IDH-mutant leukemia.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/enzymology , Isocitrate Dehydrogenase/genetics , Proto-Oncogene Proteins/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA-Binding Proteins/metabolism , Dioxygenases , Down-Regulation , Hematopoietic Stem Cells/cytology , Humans , Isocitrate Dehydrogenase/metabolism , Mice , Mutation , Proto-Oncogene Proteins/metabolism
20.
J Exp Med ; 213(1): 25-34, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26666262

ABSTRACT

t(8;21) is one of the most frequent chromosomal abnormalities observed in acute myeloid leukemia (AML). However, expression of AML1-ETO is not sufficient to induce transformation in vivo. Consistent with this observation, patients with this translocation harbor additional genetic abnormalities, suggesting a requirement for cooperating mutations. To better define the genetic landscape in AML and distinguish driver from passenger mutations, we compared the mutational profiles of AML1-ETO-driven mouse models of leukemia with the mutational profiles of human AML patients. We identified TET2 and PTPN11 mutations in both mouse and human AML and then demonstrated the ability of Tet2 loss and PTPN11 D61Y to initiate leukemogenesis in concert with expression of AML1-ETO in vivo. This integrative genetic profiling approach allowed us to accurately predict cooperating events in t(8;21)(+) AML in a robust and unbiased manner, while also revealing functional convergence in mouse and human AML.


Subject(s)
Alleles , Epistasis, Genetic , Genomics/methods , Leukemia, Myeloid, Acute/genetics , Animals , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit/genetics , Disease Models, Animal , Gene Expression Regulation, Leukemic , Gene Knockout Techniques , Humans , Mice , Mutation , Oncogene Proteins, Fusion/genetics , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , RUNX1 Translocation Partner 1 Protein , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...