Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Nat Genet ; 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39294495

ABSTRACT

Transcription factor (TF) DNA-binding dynamics govern cell fate and identity. However, our ability to pharmacologically control TF localization is limited. Here we leverage chemically driven binding site restriction leading to robust and DNA-sequence-specific redistribution of PU.1, a pioneer TF pertinent to many hematopoietic malignancies. Through an innovative technique, 'CLICK-on-CUT&Tag', we characterize the hierarchy of de novo PU.1 motifs, predicting occupancy in the PU.1 cistrome under binding site restriction. Temporal and single-molecule studies of binding site restriction uncover the pioneering dynamics of native PU.1 and identify the paradoxical activation of an alternate target gene set driven by PU.1 localization to second-tier binding sites. These transcriptional changes were corroborated by genetic blockade and site-specific reporter assays. Binding site restriction and subsequent PU.1 network rewiring causes primary human leukemia cells to differentiate. In summary, pharmacologically induced TF redistribution can be harnessed to govern TF localization, actuate alternate gene networks and direct cell fate.

2.
Cell Stem Cell ; 31(3): 378-397.e12, 2024 03 07.
Article in English | MEDLINE | ID: mdl-38402617

ABSTRACT

Mechanisms governing the maintenance of blood-producing hematopoietic stem and multipotent progenitor cells (HSPCs) are incompletely understood, particularly those regulating fate, ensuring long-term maintenance, and preventing aging-associated stem cell dysfunction. We uncovered a role for transitory free cytoplasmic iron as a rheostat for adult stem cell fate control. We found that HSPCs harbor comparatively small amounts of free iron and show the activation of a conserved molecular response to limited iron-particularly during mitosis. To study the functional and molecular consequences of iron restriction, we developed models allowing for transient iron bioavailability limitation and combined single-molecule RNA quantification, metabolomics, and single-cell transcriptomic analyses with functional studies. Our data reveal that the activation of the limited iron response triggers coordinated metabolic and epigenetic events, establishing stemness-conferring gene regulation. Notably, we find that aging-associated cytoplasmic iron loading reversibly attenuates iron-dependent cell fate control, explicating intervention strategies for dysfunctional aged stem cells.


Subject(s)
Hematopoiesis , Iron , Hematopoiesis/genetics , Iron/metabolism , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Gene Expression Regulation , Cell Differentiation
3.
J Exp Med ; 219(11)2022 11 07.
Article in English | MEDLINE | ID: mdl-36066492

ABSTRACT

Cell surfaces display a wide array of molecules that confer identity. While flow cytometry and cluster of differentiation (CD) markers have revolutionized cell characterization and purification, functionally heterogeneous cellular subtypes remain unresolvable by the CD marker system alone. Using hematopoietic lineages as a paradigm, we leverage the extraordinary molecular diversity of heparan sulfate (HS) glycans to establish cellular "glycotypes" by utilizing a panel of anti-HS single-chain variable fragment antibodies (scFvs). Prospective sorting with anti-HS scFvs identifies functionally distinct glycotypes within heterogeneous pools of mouse and human hematopoietic progenitor cells and enables further stratification of immunophenotypically pure megakaryocyte-erythrocyte progenitors. This stratification correlates with expression of a heptad of HS-related genes that is reflective of the HS epitope recognized by specific anti-HS scFvs. While we show that HS glycotyping provides an orthogonal set of tools for resolution of hematopoietic lineages, we anticipate broad utility of this approach in defining and isolating novel, viable cell types across diverse tissues and species.


Subject(s)
Hematopoiesis , Single-Chain Antibodies , Flow Cytometry , Hematopoiesis/genetics , Hematopoietic Stem Cells , Heparitin Sulfate , Humans , Prospective Studies
4.
Mol Cell ; 81(17): 3445-3446, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34478652

ABSTRACT

In this issue of Molecular Cell, Cao et al. (2021) report that AML cells are specifically addicted to an IRF8-MEF2D gene expression network. Furthermore, they identify a chromatin reader, ZMYND8, as the upstream regulator of the IRF8-MEF2D program whose activity is critical for AML cell survival.


Subject(s)
Leukemia, Myeloid, Acute , Tumor Suppressor Proteins , Chromatin , Humans , Interferon Regulatory Factors/genetics , Leukemia, Myeloid, Acute/genetics , Tumor Suppressor Proteins/genetics
5.
Semin Hematol ; 58(1): 15-26, 2021 01.
Article in English | MEDLINE | ID: mdl-33509439

ABSTRACT

Erythroid differentiation program is comprised of lineage commitment, erythroid progenitor proliferation, and termination differentiation. Each stage of the differentiation program is heavily influenced by epigenetic modifiers that alter the epigenome in a dynamic fashion influenced by cytokines/humeral factors and are amicable to target by drugs. The epigenetic modifiers can be classified as DNA modifiers (DNMT, TET), mRNA modifiers (RNA methylases and demethylases) and histone protein modifiers (methyltransferases, acetyltransferases, demethylases, and deacetylases). Here we describe mechanisms by which these epigenetic modifiers influence and guide erythroid-lineage differentiation during normal and malignant erythropoiesis and also benign diseases that arise from their altered structure or function.


Subject(s)
Epigenesis, Genetic , Histones , Epigenomics , Erythropoiesis/genetics , Histones/genetics , Histones/metabolism , Humans
6.
J Exp Med ; 218(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33427877

ABSTRACT

In this issue of JEM, Louka et al. (https://doi.org/10.1084/jem.20180853) report that leukemia stem cells lie within the phenotypic hematopoietic stem cell and progenitor cell compartments in juvenile myelomonocytic leukemia (JMML). Furthermore, they identify several candidate biomarker/therapeutic targets, such as CD96 and SLC2A1, that are of translational significance in JMML.


Subject(s)
Leukemia, Myelomonocytic, Juvenile , Hematopoietic Stem Cells , Humans
7.
Clin Cancer Res ; 25(18): 5638-5649, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31308061

ABSTRACT

PURPOSE: Myelodysplastic syndromes (MDS) with deletion of chromosome 7q/7 [-7/(del)7q MDS] is associated with worse outcomes and needs novel insights into pathogenesis. Reduced expression of signaling protein dedicator of cytokinesis 4 (DOCK4) in patients with -7/(del)7q MDS leads to a block in hematopoietic stem cell (HSC) differentiation. Identification of targetable signaling networks downstream of DOCK4 will provide means to restore hematopoietic differentiation in MDS.Experimental Design: We utilized phosphoproteomics approaches to identify signaling proteins perturbed as a result of reduced expression of DOCK4 in human HSCs and tested their functional significance in primary model systems. RESULTS: We demonstrate that reduced levels of DOCK4 lead to increased global tyrosine phosphorylation of proteins in primary human HSCs. LYN kinase and phosphatases INPP5D (SHIP1) and PTPN6 (SHP1) displayed greatest levels of tyrosine phosphorylation when DOCK4 expression levels were reduced using DOCK4-specific siRNA. Our data also found that increased phosphorylation of SHIP1 and SHP1 phosphatases were due to LYN kinase targeting these phosphatases as substrates. Increased migration and impediment of HSC differentiation were consequences of these signaling alterations. Pharmacologic inhibition of SHP1 reversed these functional aberrations in HSCs expressing low DOCK4 levels. In addition, differentiation block seen in DOCK4 haplo-insufficient [-7/(del)7q] MDS was rescued by inhibition of SHP1 phosphatase. CONCLUSIONS: LYN kinase and phosphatases SHP1 and SHIP1 are perturbed when DOCK4 expression levels are low. Inhibition of SHP1 promotes erythroid differentiation in healthy HSCs and in -7/(del)7q MDS samples with low DOCK4 expression. Inhibitors of LYN, SHP1 and SHIP1 also abrogated increased migratory properties in HSCs expressing reduced levels of DOCK4.


Subject(s)
Antineoplastic Agents/pharmacology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Loss of Function Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Signal Transduction/drug effects , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Myelodysplastic Syndromes/pathology , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Phosphorylation , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism
8.
Cancer Discov ; 9(6): 778-795, 2019 06.
Article in English | MEDLINE | ID: mdl-30944118

ABSTRACT

Even though the Ten-eleven translocation (TET) enzymes catalyze the generation of 5-hydroxymethylcytosines required for lineage commitment and subsequent differentiation of stem cells into erythroid cells, the mechanisms that link extracellular signals to TET activation and DNA hydroxymethylation are unknown. We demonstrate that hematopoietic cytokines phosphorylate TET2, leading to its activation in erythroid progenitors. Specifically, cytokine receptor-associated JAK2 phosphorylates TET2 at tyrosines 1939 and 1964. Phosphorylated TET2 interacts with the erythroid transcription factor KLF1, and this interaction with TET2 is increased upon exposure to erythropoietin. The activating JAK2V617F mutation seen in myeloproliferative disease patient samples and in mouse models is associated with increased TET activity and cytosine hydroxymethylation as well as genome-wide loss of cytosine methylation. These epigenetic and functional changes are also associated with increased expression of several oncogenic transcripts. Thus, we demonstrate that JAK2-mediated TET2 phosphorylation provides a mechanistic link between extracellular signals and epigenetic changes during hematopoiesis. SIGNIFICANCE: Identification of TET2 phosphorylation and activation by cytokine-stimulated JAK2 links extracellular signals to chromatin remodeling during hematopoietic differentiation. This provides potential avenues to regulate TET2 function in the context of myeloproliferative disorders and myelodysplastic syndromes associated with the JAK2V617F-activating mutation.This article is highlighted in the In This Issue feature, p. 681.


Subject(s)
Cytokines/metabolism , DNA-Binding Proteins/genetics , Hematopoiesis/genetics , Janus Kinase 2/metabolism , Proto-Oncogene Proteins/genetics , Transcriptional Activation , Biomarkers , Dioxygenases , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Phosphorylation
9.
J Clin Invest ; 127(4): 1316-1320, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28240607

ABSTRACT

Primary myelofibrosis (PMF) is a clonal hematologic malignancy characterized by BM fibrosis, extramedullary hematopoiesis, circulating CD34+ cells, splenomegaly, and a propensity to evolve to acute myeloid leukemia. Moreover, the spleen and BM of patients harbor atypical, clustered megakaryocytes, which contribute to the disease by secreting profibrotic cytokines. Here, we have revealed that megakaryocytes in PMF show impaired maturation that is associated with reduced GATA1 protein. In investigating the cause of GATA1 downregulation, our gene-expression study revealed the presence of the RPS14-deficient gene signature, which is associated with defective ribosomal protein function and linked to the erythroid lineage in 5q deletion myelodysplastic syndrome. Surprisingly, reduced GATA1 expression and impaired differentiation were limited to megakaryocytes, consistent with a proproliferative effect of a GATA1 deficiency on this lineage. Importantly, expression of GATA1 effectively rescued maturation of PMF megakaryocytes. Together, these results suggest that ribosomal deficiency contributes to impaired megakaryopoiesis in myeloproliferative neoplasms.


Subject(s)
Down-Regulation , GATA1 Transcription Factor/biosynthesis , Megakaryocytes/metabolism , Primary Myelofibrosis/metabolism , Thrombopoiesis , Animals , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Chromosomes, Human, Pair 5/metabolism , GATA1 Transcription Factor/genetics , Humans , Megakaryocytes/pathology , Mice , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Ribosomal Proteins/biosynthesis , Ribosomal Proteins/genetics
10.
Proc Natl Acad Sci U S A ; 112(46): E6359-68, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26578796

ABSTRACT

Anemia is the predominant clinical manifestation of myelodysplastic syndromes (MDS). Loss or deletion of chromosome 7 is commonly seen in MDS and leads to a poor prognosis. However, the identity of functionally relevant, dysplasia-causing, genes on 7q remains unclear. Dedicator of cytokinesis 4 (DOCK4) is a GTPase exchange factor, and its gene maps to the commonly deleted 7q region. We demonstrate that DOCK4 is underexpressed in MDS bone marrow samples and that the reduced expression is associated with decreased overall survival in patients. We show that depletion of DOCK4 levels leads to erythroid cells with dysplastic morphology both in vivo and in vitro. We established a novel single-cell assay to quantify disrupted F-actin filament network in erythroblasts and demonstrate that reduced expression of DOCK4 leads to disruption of the actin filaments, resulting in erythroid dysplasia that phenocopies the red blood cell (RBC) defects seen in samples from MDS patients. Reexpression of DOCK4 in -7q MDS patient erythroblasts resulted in significant erythropoietic improvements. Mechanisms underlying F-actin disruption revealed that DOCK4 knockdown reduces ras-related C3 botulinum toxin substrate 1 (RAC1) GTPase activation, leading to increased phosphorylation of the actin-stabilizing protein ADDUCIN in MDS samples. These data identify DOCK4 as a putative 7q gene whose reduced expression can lead to erythroid dysplasia.


Subject(s)
Erythroblasts/metabolism , GTPase-Activating Proteins/biosynthesis , Gene Expression Regulation , Myelodysplastic Syndromes/metabolism , Actins/genetics , Actins/metabolism , Animals , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Erythroblasts/pathology , Female , GTPase-Activating Proteins/genetics , Humans , Male , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
11.
Cell Rep ; 6(1): 231-244, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24373966

ABSTRACT

Hematopoietic stem cell differentiation involves the silencing of self-renewal genes and induction of a specific transcriptional program. Identification of multiple covalent cytosine modifications raises the question of how these derivatized bases influence stem cell commitment. Using a replicative primary human hematopoietic stem/progenitor cell differentiation system, we demonstrate dynamic changes of 5-hydroxymethylcytosine (5-hmC) during stem cell commitment and differentiation to the erythroid lineage. Genomic loci that maintain or gain 5-hmC density throughout erythroid differentiation contain binding sites for erythroid transcription factors and several factors not previously recognized as erythroid-specific factors. The functional importance of 5-hmC was demonstrated by impaired erythroid differentiation, with augmentation of myeloid potential, and disrupted 5-hmC patterning in leukemia patient-derived CD34+ stem/early progenitor cells with TET methylcytosine dioxygenase 2 (TET2) mutations. Thus, chemical conjugation and affinity purification of 5-hmC-enriched sequences followed by sequencing serve as resources for deciphering functional implications for gene expression during stem cell commitment and differentiation along a particular lineage.


Subject(s)
DNA Methylation , Erythroid Cells/metabolism , Erythropoiesis/genetics , Regulatory Sequences, Nucleic Acid , 5-Methylcytosine/analogs & derivatives , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cells, Cultured , Cytosine/analogs & derivatives , Cytosine/analysis , Dioxygenases/genetics , Dioxygenases/metabolism , Erythroid Cells/cytology , Erythroid Cells/drug effects , Histones/metabolism , Humans , Mutation , Transcription Factors/metabolism
12.
J Biol Chem ; 288(13): 8805-14, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23306203

ABSTRACT

Differentiation of hematopoietic stem cells to red cells requires coordinated expression of numerous erythroid genes and is characterized by nuclear condensation and extrusion during terminal development. To understand the regulatory mechanisms governing these widespread phenotypic changes, we conducted a high resolution methylomic and transcriptomic analysis of six major stages of human erythroid differentiation. We observed widespread epigenetic differences between early and late stages of erythropoiesis with progressive loss of methylation being the dominant change during differentiation. Gene bodies, intergenic regions, and CpG shores were preferentially demethylated during erythropoiesis. Epigenetic changes at transcription factor binding sites correlated significantly with changes in gene expression and were enriched for binding motifs for SCL, MYB, GATA, and other factors not previously implicated in erythropoiesis. Demethylation at gene promoters was associated with increased expression of genes, whereas epigenetic changes at gene bodies correlated inversely with gene expression. Important gene networks encoding erythrocyte membrane proteins, surface receptors, and heme synthesis proteins were found to be regulated by DNA methylation. Furthermore, integrative analysis enabled us to identify novel, potential regulatory areas of the genome as evident by epigenetic changes in a predicted PU.1 binding site in intron 1 of the GATA1 gene. This intronic site was found to be conserved across species and was validated to be a novel PU.1 binding site by quantitative ChIP in erythroid cells. Altogether, our study provides a comprehensive analysis of methylomic and transcriptomic changes during erythroid differentiation and demonstrates that human terminal erythropoiesis is surprisingly associated with hypomethylation of the genome.


Subject(s)
Erythropoiesis/physiology , Gene Expression Profiling , Gene Expression Regulation , Antigens, CD34/biosynthesis , Binding Sites , Cell Differentiation , CpG Islands , DNA Methylation , Epigenesis, Genetic , Epigenomics , Erythrocytes/cytology , Flow Cytometry/methods , Genome, Human , Genomics , Humans , Introns , Methylation , Oligonucleotide Array Sequence Analysis , Stem Cells/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL