Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Blood ; 138(18): 1691-1704, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34324630

ABSTRACT

Histone H3 lysine 4 methylation (H3K4Me) is most often associated with chromatin activation, and removing H3K4 methyl groups has been shown to be coincident with gene repression. H3K4Me demethylase KDM1a/LSD1 is a therapeutic target for multiple diseases, including for the potential treatment of ß-globinopathies (sickle cell disease and ß-thalassemia), because it is a component of γ-globin repressor complexes, and LSD1 inactivation leads to robust induction of the fetal globin genes. The effects of LSD1 inhibition in definitive erythropoiesis are not well characterized, so we examined the consequences of conditional inactivation of Lsd1 in adult red blood cells using a new Gata1creERT2 bacterial artificial chromosome transgene. Erythroid-specific loss of Lsd1 activity in mice led to a block in erythroid progenitor differentiation and to the expansion of granulocyte-monocyte progenitor-like cells, converting hematopoietic differentiation potential from an erythroid fate to a myeloid fate. The analogous phenotype was also observed in human hematopoietic stem and progenitor cells, coincident with the induction of myeloid transcription factors (eg, PU.1 and CEBPα). Finally, blocking the activity of the transcription factor PU.1 or RUNX1 at the same time as LSD1 inhibition rescued myeloid lineage conversion to an erythroid phenotype. These data show that LSD1 promotes erythropoiesis by repressing myeloid cell fate in adult erythroid progenitors and that inhibition of the myeloid-differentiation pathway reverses the lineage switch induced by LSD1 inactivation.


Subject(s)
Erythroid Cells/cytology , Erythropoiesis , Histone Demethylases/metabolism , Myeloid Cells/cytology , Animals , Cell Line , Cells, Cultured , Erythroid Cells/metabolism , Gene Deletion , Histone Demethylases/genetics , Humans , Mice , Myeloid Cells/metabolism
2.
Am J Pathol ; 191(7): 1193-1208, 2021 07.
Article in English | MEDLINE | ID: mdl-33894177

ABSTRACT

Pulmonary fibrosis (PF) can arise from unknown causes, as in idiopathic PF, or as a consequence of infections, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Current treatments for PF slow, but do not stop, disease progression. We report that treatment with a runt-related transcription factor 1 (RUNX1) inhibitor (Ro24-7429), previously found to be safe, although ineffective, as a Tat inhibitor in patients with HIV, robustly ameliorates lung fibrosis and inflammation in the bleomycin-induced PF mouse model. RUNX1 inhibition blunted fundamental mechanisms downstream pathologic mediators of fibrosis and inflammation, including transforming growth factor-ß1 and tumor necrosis factor-α, in cultured lung epithelial cells, fibroblasts, and vascular endothelial cells, indicating pleiotropic effects. RUNX1 inhibition also reduced the expression of angiotensin-converting enzyme 2 and FES Upstream Region (FURIN), host proteins critical for SARS-CoV-2 infection, in mice and in vitro. A subset of human lungs with SARS-CoV-2 infection overexpress RUNX1. These data suggest that RUNX1 inhibition via repurposing of Ro24-7429 may be beneficial for PF and to battle SARS-CoV-2, by reducing expression of viral mediators and by preventing respiratory complications.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/metabolism , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Furin/metabolism , Lung/drug effects , Pulmonary Fibrosis/drug therapy , Animals , Bleomycin , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Lung/metabolism , Lung/pathology , Male , Mice , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Treatment Outcome
3.
FASEB J ; 35(2): e21155, 2021 02.
Article in English | MEDLINE | ID: mdl-33135824

ABSTRACT

Runt-related transcription factor 1 (RUNX1) acts as a mediator of aberrant retinal angiogenesis and has been implicated in the progression of proliferative diabetic retinopathy (PDR). Patients with PDR, retinopathy of prematurity (ROP), and wet age-related macular degeneration (wet AMD) have been found to have elevated levels of Tumor Necrosis Factor-alpha (TNF-α) in the eye. In fibrovascular membranes (FVMs) taken from patients with PDR RUNX1 expression was increased in the vasculature, while in human retinal microvascular endothelial cells (HRMECs), TNF-α stimulation causes increased RUNX1 expression, which can be modulated by RUNX1 inhibitors. Using TNF-α pathway inhibitors, we determined that in HRMECs, TNF-α-induced RUNX1 expression occurs via JNK activation, while NF-κB and p38/MAPK inhibition did not affect RUNX1 expression. JNK inhibitors were also effective at stopping high D-glucose-stimulated RUNX1 expression. We further linked JNK to RUNX1 through Activator Protein 1 (AP-1) and investigated the JNK-AP-1-RUNX1 regulatory feedback loop, which can be modulated by VEGF. Additionally, stimulation with TNF-α and D-glucose had an additive effect on RUNX1 expression, which was downregulated by VEGF modulation. These data suggest that the downregulation of RUNX1 in conjunction with anti-VEGF agents may be important in future treatments for the management of diseases of pathologic ocular angiogenesis.


Subject(s)
Choroidal Neovascularization/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Diabetic Retinopathy/metabolism , Endothelial Cells/metabolism , MAP Kinase Signaling System/drug effects , Retinopathy of Prematurity/metabolism , Tumor Necrosis Factor-alpha/metabolism , Wet Macular Degeneration/metabolism , Animals , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Disease Models, Animal , Endothelial Cells/drug effects , Glucose/pharmacology , Humans , Mice , Mice, Inbred C57BL , Retina/cytology , Retina/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
4.
Blood ; 134(1): 59-73, 2019 07 04.
Article in English | MEDLINE | ID: mdl-31023702

ABSTRACT

RUNX1 transcription factor regulates normal and malignant hematopoiesis. Somatic or germline mutant RUNX1 (mtRUNX1) is associated with poorer outcome in acute myeloid leukemia (AML). Knockdown or inhibition of RUNX1 induced more apoptosis of AML expressing mtRUNX1 versus wild-type RUNX1 and improved survival of mice engrafted with mtRUNX1-expressing AML. CRISPR/Cas9-mediated editing-out of RUNX1 enhancer (eR1) within its intragenic super-enhancer, or BET protein BRD4 depletion by short hairpin RNA, repressed RUNX1, inhibited cell growth, and induced cell lethality in AML cells expressing mtRUNX1. Moreover, treatment with BET protein inhibitor or degrader (BET-proteolysis targeting chimera) repressed RUNX1 and its targets, inducing apoptosis and improving survival of mice engrafted with AML expressing mtRUNX1. Library of Integrated Network-based Cellular Signatures 1000-connectivity mapping data sets queried with messenger RNA signature of RUNX1 knockdown identified novel expression-mimickers (EMs), which repressed RUNX1 and exerted in vitro and in vivo efficacy against AML cells expressing mtRUNX1. In addition, the EMs cinobufagin, anisomycin, and narciclasine induced more lethality in hematopoietic progenitor cells (HPCs) expressing germline mtRUNX1 from patients with AML compared with HPCs from patients with familial platelet disorder (FPD), or normal untransformed HPCs. These findings highlight novel therapeutic agents for AML expressing somatic or germline mtRUNX1.


Subject(s)
Antineoplastic Agents/pharmacology , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Leukemia, Myeloid, Acute/genetics , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Core Binding Factor Alpha 2 Subunit/genetics , Gene Knockdown Techniques , Germ-Line Mutation , Hematopoietic Stem Cells/drug effects , Humans , Mice
5.
Mol Pharmacol ; 98(6): 648-657, 2020 12.
Article in English | MEDLINE | ID: mdl-32978326

ABSTRACT

Protein tyrosine phosphatase (PTP) 4A3 is frequently overexpressed in human solid tumors and hematologic malignancies and is associated with tumor cell invasion, metastasis, and a poor patient prognosis. Several potent, selective, and allosteric small molecule inhibitors of PTP4A3 were recently identified. A lead compound in the series, JMS-053 (7-imino-2-phenylthieno[3,2-c]pyridine-4,6(5H,7H)-dione), has a long plasma half-life (∼ 24 hours) in mice, suggesting possible binding to serum components. We confirmed by isothermal titration calorimetry that JMS-053 binds to human serum albumin. A single JMS-053 binding site was identified by X-ray crystallography in human serum albumin at drug site 3, which is also known as subdomain IB. The binding of JMS-053 to human serum albumin, however, did not markedly alter the overall albumin structure. In the presence of serum albumin, the potency of JMS-053 as an in vitro inhibitor of PTP4A3 and human A2780 ovarian cancer cell growth was reduced. The reversible binding of JMS-053 to serum albumin may serve to increase JMS-053's plasma half-life and thus extend the delivery of the compound to tumors. SIGNIFICANCE STATEMENT: X-ray crystallography revealed that a potent, reversible, first-in-class small molecule inhibitor of the oncogenic phosphatase protein tyrosine phosphatase 4A3 binds to at least one site on human serum albumin, which is likely to extend the compound's plasma half-life and thus assist in drug delivery into tumors.


Subject(s)
Imines/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Protein Tyrosine Phosphatases/antagonists & inhibitors , Pyridines/pharmacology , Serum Albumin, Human/metabolism , Binding Sites , Calorimetry , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Enzyme Assays , Half-Life , Humans , Imines/chemistry , Imines/therapeutic use , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Protein Tyrosine Phosphatases/metabolism , Pyridines/chemistry , Pyridines/therapeutic use , Serum Albumin, Human/ultrastructure
6.
Blood ; 130(15): 1722-1733, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28790107

ABSTRACT

The gene encoding the RUNX1 transcription factor is mutated in a subset of T-cell acute lymphoblastic leukemia (T-ALL) patients, and RUNX1 mutations are associated with a poor prognosis. These mutations cluster in the DNA-binding Runt domain and are thought to represent loss-of-function mutations, indicating that RUNX1 suppresses T-cell transformation. RUNX1 has been proposed to have tumor suppressor roles in T-cell leukemia homeobox 1/3-transformed human T-ALL cell lines and NOTCH1 T-ALL mouse models. Yet, retroviral insertional mutagenesis screens identify RUNX genes as collaborating oncogenes in MYC-driven leukemia mouse models. To elucidate RUNX1 function(s) in leukemogenesis, we generated Tal1/Lmo2/Rosa26-CreERT2Runx1f/f mice and examined leukemia progression in the presence of vehicle or tamoxifen. We found that Runx1 deletion inhibits mouse leukemic growth in vivo and that RUNX silencing in human T-ALL cells triggers apoptosis. We demonstrate that a small molecule inhibitor, designed to interfere with CBFß binding to RUNX proteins, impairs the growth of human T-ALL cell lines and primary patient samples. We demonstrate that a RUNX1 deficiency alters the expression of a crucial subset of TAL1- and NOTCH1-regulated genes, including the MYB and MYC oncogenes, respectively. These studies provide genetic and pharmacologic evidence that RUNX1 has oncogenic roles and reveal RUNX1 as a novel therapeutic target in T-ALL.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Enhancer Elements, Genetic/genetics , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chromatin/metabolism , Core Binding Factor beta Subunit/metabolism , Gene Deletion , Gene Expression Regulation, Leukemic , Humans , Mice , Protein Binding , Proto-Oncogene Proteins/metabolism , Receptors, Notch/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1
7.
Gynecol Oncol ; 149(2): 350-360, 2018 05.
Article in English | MEDLINE | ID: mdl-29551565

ABSTRACT

OBJECTIVE: Ovarian cancer survival and treatment have improved minimally in the past 20years. Novel treatment strategies are needed to combat this disease. This study investigates the effects of chemical inhibition of the CBFß/RUNX protein-protein interaction on ovarian cancer cell lines. METHODS: Ovarian cancer cell lines were treated with CBFß/RUNX inhibitors, and the effects on proliferation, DNA replication, wound healing, and anchorage-independent growth were measured. RNA-Seq was performed on compound-treated cells to identify differentially expressed genes. Genes altered by compound treatment were targeted with siRNAs, and effects on DNA replication and wound healing were measured. RESULTS: Chemical inhibition of the CBFß/RUNX interaction decreases ovarian cancer cell proliferation. Inhibitor treatment leads to an S-phase cell cycle delay, as indicated by an increased percentage of cells in S-phase, and a decreased DNA replication rate. Inhibitor treatment also reduces wound healing and anchorage-independent growth. RNA-Seq on compound-treated cells revealed changes in a small number of genes related to proliferation and epithelial-to-mesenchymal transition. siRNA-mediated knockdown of INHBA and MMP1 - two genes whose expression decreases with compound treatment - slowed DNA replication and impaired wound healing. CONCLUSIONS: Chemical inhibition of the CBFß/RUNX interaction is a viable strategy for the treatment of ovarian cancer.


Subject(s)
Core Binding Factor alpha Subunits/antagonists & inhibitors , Epithelial-Mesenchymal Transition/genetics , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Small Molecule Libraries/pharmacology , Animals , Carcinoma, Ovarian Epithelial , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Core Binding Factor alpha Subunits/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Random Allocation , Xenograft Model Antitumor Assays
8.
Adv Exp Med Biol ; 962: 229-244, 2017.
Article in English | MEDLINE | ID: mdl-28299661

ABSTRACT

Acute myeloid leukemia (AML) is characterized by recurrent chromosomal rearrangements that encode for fusion proteins which drive leukemia initiation and maintenance. The inv(16) (p13q22) rearrangement is a founding mutation and the associated CBFß-SMMHC fusion protein is essential for the survival of inv(16) AML cells. This Chapter will discuss our understanding of the function of this fusion protein in disrupting hematopoietic homeostasis and creating pre-leukemic blasts, in its cooperation with other co-occurring mutations during leukemia initiation, and in leukemia maintenance. In addition, this chapter will discuss the current approaches used for the treatment of inv(16) AML and the recent development of AI-10-49, a selective targeted inhibitor of CBFß-SMMHC/RUNX1 binding, the first candidate targeted therapy for inv(16) AML.


Subject(s)
Core Binding Factor beta Subunit/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Myosin Heavy Chains/genetics , Oncogene Proteins, Fusion/genetics , Animals , Humans
9.
Mol Cell ; 31(6): 896-908, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18922471

ABSTRACT

We describe the NMR structure of DsbB, a polytopic helical membrane protein. DsbB, a bacterial cytoplasmic membrane protein, plays a key role in disulfide bond formation. It reoxidizes DsbA, the periplasmic protein disulfide oxidant, using the oxidizing power of membrane-embedded quinones. We determined the structure of an interloop disulfide bond form of DsbB, an intermediate in catalysis. Analysis of the structure and interactions with substrates DsbA and quinone reveals functionally relevant changes induced by these substrates. Analysis of the structure, dynamics measurements, and NMR chemical shifts around the interloop disulfide bond suggest how electron movement from DsbA to quinone through DsbB is regulated and facilitated. Our results demonstrate the extraordinary utility of NMR for functional characterization of polytopic integral membrane proteins and provide insights into the mechanism of DsbB catalysis.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Membrane/enzymology , Disulfides/metabolism , Escherichia coli/cytology , Escherichia coli/enzymology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Binding Sites , Catalysis , Cysteine/metabolism , Electron Spin Resonance Spectroscopy , Escherichia coli Proteins/chemistry , Lipid Bilayers/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Oxidation-Reduction , Periplasm/enzymology , Protein Disulfide-Isomerases/chemistry , Protein Interaction Mapping , Protein Structure, Secondary , Solutions , Ubiquinone
10.
Proc Natl Acad Sci U S A ; 110(33): 13374-9, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23898196

ABSTRACT

The Ets-Related Gene (ERG) belongs to the Ets family of transcription factors and is critically important for maintenance of the hematopoietic stem cell population. A chromosomal translocation observed in the majority of human prostate cancers leads to the aberrant overexpression of ERG. We have identified regions flanking the ERG Ets domain responsible for autoinhibition of DNA binding and solved crystal structures of uninhibited, autoinhibited, and DNA-bound ERG. NMR-based measurements of backbone dynamics show that uninhibited ERG undergoes substantial dynamics on the millisecond-to-microsecond timescale but autoinhibited and DNA-bound ERG do not. We propose a mechanism whereby the allosteric basis of ERG autoinhibition is mediated predominantly by the regulation of Ets-domain dynamics with only modest structural changes.


Subject(s)
DNA-Binding Proteins/chemistry , DNA/metabolism , Models, Molecular , Trans-Activators/chemistry , Allosteric Regulation/physiology , Calorimetry , Cloning, Molecular , Crystallography, X-Ray , DNA-Binding Proteins/metabolism , Humans , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Oligonucleotides/genetics , Protein Structure, Tertiary , Spectrum Analysis , Time Factors , Trans-Activators/metabolism , Transcriptional Regulator ERG
11.
Cancer Cell ; 11(6): 483-97, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17560331

ABSTRACT

AML1/ETO results from the t(8;21) associated with 12%-15% of acute myeloid leukemia. The AML1/ETO MYND domain mediates interactions with the corepressors SMRT and N-CoR and contributes to AML1/ETO's ability to repress proliferation and differentiation of primary bone marrow cells as well as to enhance their self renewal in vitro. We solved the solution structure of the MYND domain and show it to be structurally homologous to the PHD and RING finger families of proteins. We also determined the solution structure of an MYND-SMRT peptide complex. We demonstrated that a single amino acid substitution that disrupts the interaction between the MYND domain and the SMRT peptide attenuated AML1/ETO's effects on proliferation, differentiation, and gene expression.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Repressor Proteins/metabolism , Animals , Bone Marrow Cells , Cell Differentiation , Cell Line , Cell Proliferation , Gene Expression , Humans , Mice , Models, Molecular , Mutation , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1 , Protein Binding , Protein Structure, Tertiary , RUNX1 Translocation Partner 1 Protein , Repressor Proteins/genetics
12.
J Biol Chem ; 288(41): 29901-10, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-23990460

ABSTRACT

The MLL CXXC domain binds nonmethylated CpG-containing DNA and is essential for the oncogenic properties of MLL fusion proteins. To determine potential functional promiscuity of similar DNA binding domains, we replaced the MLL CXXC domain in the context of the leukemogenic MLL-AF9 fusion with CXXC domains from DNMT1, CGBP (CFP1), and MBD1, or with a methyl-CpG-binding domain (MBD) from MBD1. MLL(DNMT1 CXXC)-AF9 shows robust in vitro colony forming activity and in vivo leukemogenesis, similar to MLL-AF9. However, colony forming ability and leukemogenicity are abrogated in MLL-AF9 containing either the CGBP or MBD1 CXXC domains or the MBD1 MBD domain. Direct comparison of in vitro DNA binding affinity of the isolated CXXC or MBD domains demonstrated that MLL, DNMT1, and CGBP CXXC domains could each bind to unmethylated DNA but with differing affinity. In contrast, the isolated MBD1 CXXC and MBD1 MBD domains were unable to bind to the same DNA. However, all substituted domains still allowed targeting of the MLL fusions to the functionally important Hoxa9 locus in primary bone marrow progenitor cells. In addition to DNA binding activity, it was critical that specific CpG residues in the Hoxa9 locus were protected from methylation for leukemia development. This ultimately prevented histone 3 lysine 9 trimethylation (H3K9me3) of the locus and enabled Hoxa9 expression. These were properties shared by MLL and DNMT1 CXXC domains but not by CGBP CXXC or the other swapped fusions tested. We demonstrate that similar CXXC domains can be mechanistically distinguished by specificity of CpG nucleotides preferentially protected from DNA methylation.


Subject(s)
CpG Islands , DNA/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Cell Line , Cells, Cultured , Chromatin Immunoprecipitation , DNA/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Lysine/metabolism , Methylation , Mice , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Protein Binding , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Cancer Cell ; 9(4): 249-60, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16616331

ABSTRACT

AML1/ETO is the chimeric protein resulting from the t(8;21) in acute myeloid leukemia. The Nervy homology 2 (NHR2) domain in ETO mediates oligomerization and AML1/ETO's interactions with ETO, MTGR1, and MTG16, and with the corepressor molecules mSin3A and HDAC1 and HDAC3. We solved the NHR2 domain structure and found it to be an alpha-helical tetramer. We show that oligomerization contributes to AML1/ETO's inhibition of granulocyte differentiation, is essential for its ability to enhance the clonogenic potential of primary mouse bone marrow cells, and affects AML1/ETO's activity on several endogenous genes. Oligomerization is also required for AML1/ETO's interactions with ETO, MTGR1, and MTG16, but not with other corepressor molecules.


Subject(s)
Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/metabolism , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , Crystallography, X-Ray , Gene Expression Regulation , Humans , Hydrophobic and Hydrophilic Interactions , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Oncogene Proteins, Fusion/genetics , Protein Binding , Protein Structure, Quaternary , RUNX1 Translocation Partner 1 Protein , Sequence Alignment
14.
Sci Adv ; 10(9): eadh8493, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38416825

ABSTRACT

N-MYC (encoded by MYCN) is a critical regulator of hematopoietic stem cell function. While the role of N-MYC deregulation is well established in neuroblastoma, the importance of N-MYC deregulation in leukemogenesis remains elusive. Here, we demonstrate that N-MYC is overexpressed in acute myeloid leukemia (AML) cells with chromosome inversion inv(16) and contributes to the survival and maintenance of inv(16) leukemia. We identified a previously unknown MYCN enhancer, active in multiple AML subtypes, essential for MYCN mRNA levels and survival in inv(16) AML cells. We also identified eukaryotic translation initiation factor 4 gamma 1 (eIF4G1) as a key N-MYC target that sustains leukemic survival in inv(16) AML cells. The oncogenic role of eIF4G1 in AML has not been reported before. Our results reveal a mechanism whereby N-MYC drives a leukemic transcriptional program and provides a rationale for the therapeutic targeting of the N-MYC/eIF4G1 axis in myeloid leukemia.


Subject(s)
Leukemia, Myeloid, Acute , Humans , N-Myc Proto-Oncogene Protein , Cell Survival/genetics , Leukemia, Myeloid, Acute/genetics , Carcinogenesis , Hematopoietic Stem Cells
15.
ACS Med Chem Lett ; 13(8): 1363-1369, 2022 Aug 11.
Article in English | MEDLINE | ID: mdl-35978680

ABSTRACT

The CXXC domain is a reader of DNA methylation which preferentially binds to unmethylated CpG DNA motifs. Chromosomal translocations involving the MLL1 gene produce in-frame fusion proteins in which the N-terminal portion of the MLL1 protein harboring its CXXC domain is fused to the C-terminal portion of multiple partners. For the MLL-AF9 fusion, mutations which disrupt CXXC domain-DNA binding abrogate the ability to cause leukemia in mice. Based on this, we initiated an effort to develop small-molecule inhibitors of the MLL1 CXXC domain as a novel approach to therapy. We developed a fluorescence polarization-based assay for MLL CXXC domain-DNA binding and screened a library of Cys-reactive molecules. For the most potent hit from this screen, we have synthesized a library of analogs to explore the structure-activity relationship, defined the binding site using chemical shift perturbations in NMR spectra, and explored the selectivity of compounds across the CXXC domain family.

16.
Sci Adv ; 8(7): eabl4618, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35171684

ABSTRACT

EP300, a transcription coactivator important in proliferation and differentiation, is frequently mutated in diverse cancer types, including small cell lung cancer (SCLC). While these mutations are thought to result in loss of EP300 function, the impact on tumorigenesis remains largely unknown. Here, we demonstrate that EP300 mutants lacking acetyltransferase domain accelerate tumor development in mouse models of SCLC. However, unexpectedly, complete Ep300 knockout suppresses SCLC development and proliferation. Dissection of EP300 domains identified kinase inducible domain-interacting (KIX) domain, specifically its interaction with transcription factors including MYB, as the determinant of protumorigenic activity. Ala627 in EP300 KIX results in a higher protein-binding affinity than Asp647 at the equivalent position in CREBBP KIX, underlying the selectivity of KIX-binding partners for EP300. Blockade of KIX-mediated interactions inhibits SCLC development in mice and cell growth. This study unravels domain-specific roles for EP300 in SCLC and unique vulnerability of the EP300 KIX domain for therapeutic intervention.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Animals , E1A-Associated p300 Protein , Lung Neoplasms/genetics , Mice , Protein Binding , Small Cell Lung Carcinoma/genetics , Transcription Factors/metabolism
17.
Nat Commun ; 13(1): 7124, 2022 11 21.
Article in English | MEDLINE | ID: mdl-36411286

ABSTRACT

The ETV6-RUNX1 onco-fusion arises in utero, initiating a clinically silent pre-leukemic state associated with the development of pediatric B-acute lymphoblastic leukemia (B-ALL). We characterize the ETV6-RUNX1 regulome by integrating chromatin immunoprecipitation- and RNA-sequencing and show that ETV6-RUNX1 functions primarily through competition for RUNX1 binding sites and transcriptional repression. In pre-leukemia, this results in ETV6-RUNX1 antagonization of cell cycle regulation by RUNX1 as evidenced by mass cytometry analysis of B-lineage cells derived from ETV6-RUNX1 knock-in human pluripotent stem cells. In frank leukemia, knockdown of RUNX1 or its co-factor CBFß results in cell death suggesting sustained requirement for RUNX1 activity which is recapitulated by chemical perturbation using an allosteric CBFß-inhibitor. Strikingly, we show that RUNX1 addiction extends to other genetic subtypes of pediatric B-ALL and also adult disease. Importantly, inhibition of RUNX1 activity spares normal hematopoiesis. Our results suggest that chemical intervention in the RUNX1 program may provide a therapeutic opportunity in ALL.


Subject(s)
Core Binding Factor Alpha 2 Subunit , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adult , Child , Humans , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , B-Lymphocytes , Gene Fusion
18.
Blood ; 113(15): 3558-67, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19204326

ABSTRACT

AML1-ETO is the chimeric protein product of the t(8;21) in acute myeloid leukemia. The ETO portion of the fusion protein includes the eTAFH domain, which is homologous to several TATA binding protein-associated factors (TAFs) and interacts with E proteins (E2A and HEB). It has been proposed that AML1-ETO-mediated silencing of E protein function might be important for t(8;21) leukemogenesis. Here, we determined the solution structure of a complex between the AML1-ETO eTAFH domain and an interacting peptide from HEB. On the basis of the structure, key residues in AML1-ETO for HEB association were mutated. These mutations do not impair the ability of AML1-ETO to enhance the clonogenic capacity of primary mouse bone marrow cells and do not eliminate its ability to repress proliferation or granulocyte differentiation. Therefore, the eTAFH-E protein interaction appears to contribute relatively little to the activity of AML1-ETO.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , DNA-Binding Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , COS Cells , Cell Differentiation/immunology , Cell Division/immunology , Cells, Cultured , Chlorocebus aethiops , Core Binding Factor Alpha 2 Subunit/chemistry , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Granulocytes/cytology , Granulocytes/physiology , Leukemia, Myeloid, Acute/genetics , Male , Mice , Mice, Inbred C57BL , Nuclear Magnetic Resonance, Biomolecular , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , STAT6 Transcription Factor/metabolism , Structure-Activity Relationship , Transcription Factors/chemistry , Transcription Factors/genetics
19.
Blood ; 113(13): 3070-9, 2009 Mar 26.
Article in English | MEDLINE | ID: mdl-19179469

ABSTRACT

AML1-ETO and TEL-AML1 are chimeric proteins resulting from the t(8;21)(q22;q22) in acute myeloid leukemia, and the t(12;21)(p13;q22) in pre-B-cell leukemia, respectively. The Runt domain of AML1 in both proteins mediates DNA binding and heterodimerization with the core binding factor beta (CBFbeta) subunit. To determine whether CBFbeta is required for AML1-ETO and TEL-AML1 activity, we introduced amino acid substitutions into the Runt domain that disrupt heterodimerization with CBFbeta but not DNA binding. We show that CBFbeta contributes to AML1-ETO's inhibition of granulocyte differentiation, is essential for its ability to enhance the clonogenic potential of primary mouse bone marrow cells, and is indispensable for its cooperativity with the activated receptor tyrosine kinase TEL-PDGFbetaR in generating acute myeloid leukemia in mice. Similarly, CBFbeta is essential for TEL-AML1's ability to promote self-renewal of B cell precursors in vitro. These studies validate the Runt domain/CBFbeta interaction as a therapeutic target in core binding factor leukemias.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Core Binding Factor beta Subunit/physiology , Oncogene Proteins, Fusion/physiology , Animals , Cell Differentiation/genetics , Cell Proliferation , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor beta Subunit/chemistry , Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/metabolism , Granulocytes/metabolism , Granulocytes/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Models, Biological , Models, Molecular , Mutation/physiology , NIH 3T3 Cells , Oncogene Proteins, Fusion/genetics , Protein Binding , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , RUNX1 Translocation Partner 1 Protein , Transfection
20.
Proc Natl Acad Sci U S A ; 105(21): 7517-22, 2008 May 27.
Article in English | MEDLINE | ID: mdl-18483194

ABSTRACT

Homeobox (HOX) genes play a definitive role in determination of cell fate during embryogenesis and hematopoiesis. MLL-related leukemia is coincident with increased expression of a subset of HOX genes, including HOXA9. MLL functions to maintain, rather than initiate, expression of its target genes. However, the mechanism of MLL maintenance of target gene expression is not understood. Here, we demonstrate that Mll binds to specific clusters of CpG residues within the Hoxa9 locus and regulates expression of multiple transcripts. The presence of Mll at these clusters provides protection from DNA methylation. shRNA knock-down of Mll reverses the methylation protection status at the previously protected CpG clusters; methylation at these CpG residues is similar to that observed in Mll null cells. Furthermore, reconstituting MLL expression in Mll null cells can reverse DNA methylation of the same CpG residues, demonstrating a dominant effect of MLL in protecting this specific region from DNA methylation. Intriguingly, an oncogenic MLL-AF4 fusion can also reverse DNA methylation, but only for a subset of these CpGs. This method of transcriptional regulation suggests a mechanism that explains the role of Mll in transcriptional maintenance, but it may extend to other CpG DNA binding proteins. Protection from methylation may be an important mechanism of epigenetic inheritance by regulating the function of both de novo and maintenance DNA methyltransferases.


Subject(s)
CpG Islands , DNA Methylation , Gene Expression Regulation , Homeodomain Proteins/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , Cell Line , Humans , Mice , Myeloid-Lymphoid Leukemia Protein/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL