Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Clin Invest ; 132(19)2022 10 03.
Article in English | MEDLINE | ID: mdl-35925681

ABSTRACT

Infantile (fetal and neonatal) megakaryocytes (Mks) have a distinct phenotype consisting of hyperproliferation, limited morphogenesis, and low platelet production capacity. These properties contribute to clinical problems that include thrombocytopenia in neonates, delayed platelet engraftment in recipients of cord blood stem cell transplants, and inefficient ex vivo platelet production from pluripotent stem cell-derived Mks. The infantile phenotype results from deficiency of the actin-regulated coactivator, MKL1, which programs cytoskeletal changes driving morphogenesis. As a strategy to complement this molecular defect, we screened pathways with the potential to affect MKL1 function and found that DYRK1A inhibition dramatically enhanced Mk morphogenesis in vitro and in vivo. Dyrk1 inhibitors rescued enlargement, polyploidization, and thrombopoiesis in human neonatal Mks. Mks derived from induced pluripotent stem cells responded in a similar manner. Progenitors undergoing Dyrk1 inhibition demonstrated filamentous actin assembly, MKL1 nuclear translocation, and modulation of MKL1 target genes. Loss-of-function studies confirmed MKL1 involvement in this morphogenetic pathway. Expression of Ablim2, a stabilizer of filamentous actin, increased with Dyrk1 inhibition, and Ablim2 knockdown abrogated the actin, MKL1, and morphogenetic responses to Dyrk1 inhibition. These results delineate a pharmacologically tractable morphogenetic pathway whose manipulation may alleviate clinical problems associated with the limited thrombopoietic capacity of infantile Mks.


Subject(s)
Megakaryocytes , Thrombocytopenia , Actins/metabolism , Blood Platelets/metabolism , Humans , Infant, Newborn , Megakaryocytes/metabolism , Phenotype , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Thrombocytopenia/genetics , Thrombopoiesis/genetics , Dyrk Kinases
2.
Blood Cells Mol Dis ; 97: 102688, 2022 11.
Article in English | MEDLINE | ID: mdl-35717902

ABSTRACT

Erythropoiesis is a tightly regulated process. It is stimulated by decreased oxygen in circulation, which leads to the secretion of the hormone erythropoietin (Epo) by the kidneys. An additional layer of control involves the coordinated sensing and use of nutrients. Much cellular machinery contributes to sensing and responding to nutrient status in cells, and one key participant is the kinase LKB1. The current study examines the role of LKB1 in erythropoiesis using a murine in vivo and ex vivo conditional knockout system. In vivo analysis showed erythroid loss of LKB1 to be associated with a robust increase in serum Epo and mild reticulocytosis. Despite these abnormalities, no evidence of anemia or hemolysis was found. Further characterization using an ex vivo progenitor culture assay demonstrated accelerated erythroid maturation in the LKB1-deficient cells. Based on pharmacologic evidence, this phenotype appeared to result from impaired AMP-activated protein kinase (AMPK) signaling downstream of LKB1. These findings reveal a role for LKB1 in fine-tuning Epo-driven erythropoiesis in association with maturational control.


Subject(s)
AMP-Activated Protein Kinases , Erythroid Precursor Cells , Erythropoiesis , Erythropoietin , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , Erythropoiesis/physiology , Erythropoietin/genetics , Erythropoietin/metabolism , Humans , Liver/metabolism , Mice , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism
3.
Nat Commun ; 12(1): 1645, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712594

ABSTRACT

Anemias of chronic disease and inflammation (ACDI) result from restricted iron delivery to erythroid progenitors. The current studies reveal an organellar response in erythroid iron restriction consisting of disassembly of the microtubule cytoskeleton and associated Golgi disruption. Isocitrate supplementation, known to abrogate the erythroid iron restriction response, induces reassembly of microtubules and Golgi in iron deprived progenitors. Ferritin, based on proteomic profiles, regulation by iron and isocitrate, and putative interaction with microtubules, is assessed as a candidate mediator. Knockdown of ferritin heavy chain (FTH1) in iron replete progenitors induces microtubule collapse and erythropoietic blockade; conversely, enforced ferritin expression rescues erythroid differentiation under conditions of iron restriction. Fumarate, a known ferritin inducer, synergizes with isocitrate in reversing molecular and cellular defects of iron restriction and in oral remediation of murine anemia. These findings identify a cytoskeletal component of erythroid iron restriction and demonstrate potential for its therapeutic targeting in ACDI.


Subject(s)
Anemia/metabolism , Anemia/therapy , Cytoskeleton/metabolism , Iron/metabolism , Microtubules/metabolism , Animals , Cell Proliferation , Disease Models, Animal , Erythroid Cells/metabolism , Erythropoiesis/physiology , Female , Ferritins/metabolism , Isocitrates , Male , Mice , Mice, Inbred C57BL , Oxidoreductases/metabolism , Proteomics
4.
Haematologica ; 105(4): 905-913, 2020 04.
Article in English | MEDLINE | ID: mdl-31171641

ABSTRACT

Healthy bone marrow progenitors yield a co-ordinated balance of hematopoietic lineages. This balance shifts with aging toward enhanced granulopoiesis with diminished erythropoiesis and lymphopoiesis, changes which likely contribute to the development of bone marrow disorders in the elderly. In this study, RUNX3 was identified as a hematopoietic stem and progenitor cell factor whose levels decline with aging in humans and mice. This decline is exaggerated in hematopoietic stem and progenitor cells from subjects diagnosed with unexplained anemia of the elderly. Hematopoietic stem cells from elderly unexplained anemia patients had diminished erythroid but unaffected granulocytic colony forming potential. Knockdown studies revealed human hematopoietic stem and progenitor cells to be strongly influenced by RUNX3 levels, with modest deficiencies abrogating erythroid differentiation at multiple steps while retaining capacity for granulopoiesis. Transcriptome profiling indicated control by RUNX3 of key erythroid transcription factors, including KLF1 and GATA1 These findings thus implicate RUNX3 as a participant in hematopoietic stem and progenitor cell aging, and a key determinant of erythroid-myeloid lineage balance.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Aged , Aging , Animals , Cell Differentiation , Core Binding Factor Alpha 3 Subunit/genetics , Erythropoiesis , Humans , Mice
5.
Exp Hematol ; 61: 1-9, 2018 05.
Article in English | MEDLINE | ID: mdl-29501467

ABSTRACT

Fetal megakaryocytes (Mks) differ from adult Mks in key parameters that affect their capacity for platelet production. However, despite being smaller, more proliferative, and less polyploid, fetal Mks generally mature in the same manner as adult Mks. The phenotypic features unique to fetal Mks predispose patients to several disease conditions, including infantile thrombocytopenia, infantile megakaryoblastic leukemias, and poor platelet recovery after umbilical cord blood stem cell transplantations. Ontogenic Mk differences also affect new strategies being developed to address global shortages of platelet transfusion units. These donor-independent, ex vivo production platforms are hampered by the limited proliferative capacity of adult-type Mks and the inferior platelet production by fetal-type Mks. Understanding the molecular programs that distinguish fetal versus adult megakaryopoiesis will help in improving approaches to these clinical problems. This review summarizes the phenotypic differences between fetal and adult Mks, the disease states associated with fetal megakaryopoiesis, and recent advances in the understanding of mechanisms that determine ontogenic Mk transitions.


Subject(s)
Megakaryocytes/cytology , Fetal Blood/cytology , Humans , Megakaryocytes/pathology , Models, Biological , Morphogenesis/physiology , Phenotype , Thrombocytopenia/pathology
6.
J Clin Invest ; 127(6): 2365-2377, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28481226

ABSTRACT

Hematopoietic transitions that accompany fetal development, such as erythroid globin chain switching, play important roles in normal physiology and disease development. In the megakaryocyte lineage, human fetal progenitors do not execute the adult morphogenesis program of enlargement, polyploidization, and proplatelet formation. Although these defects decline with gestational stage, they remain sufficiently severe at birth to predispose newborns to thrombocytopenia. These defects may also contribute to inferior platelet recovery after cord blood stem cell transplantation and may underlie inefficient platelet production by megakaryocytes derived from pluripotent stem cells. In this study, comparison of neonatal versus adult human progenitors has identified a blockade in the specialized positive transcription elongation factor b (P-TEFb) activation mechanism that is known to drive adult megakaryocyte morphogenesis. This blockade resulted from neonatal-specific expression of an oncofetal RNA-binding protein, IGF2BP3, which prevented the destabilization of the nuclear RNA 7SK, a process normally associated with adult megakaryocytic P-TEFb activation. Knockdown of IGF2BP3 sufficed to confer both phenotypic and molecular features of adult-type cells on neonatal megakaryocytes. Pharmacologic inhibition of IGF2BP3 expression via bromodomain and extraterminal domain (BET) inhibition also elicited adult features in neonatal megakaryocytes. These results identify IGF2BP3 as a human ontogenic master switch that restricts megakaryocyte development by modulating a lineage-specific P-TEFb activation mechanism, revealing potential strategies toward enhancing platelet production.


Subject(s)
Megakaryocytes/physiology , RNA-Binding Proteins/physiology , Animals , Cell Proliferation , Female , Gene Expression , Gene Expression Regulation, Developmental , HEK293 Cells , Hematopoiesis , Hematopoietic Stem Cells/physiology , Humans , Infant, Newborn , K562 Cells , Mice, Inbred C57BL , Transcriptional Activation
9.
Dev Cell ; 27(6): 607-20, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24369834

ABSTRACT

Megakaryocyte morphogenesis employs a "hypertrophy-like" developmental program that is dependent on P-TEFb kinase activation and cytoskeletal remodeling. P-TEFb activation classically occurs by a feedback-regulated process of signal-induced, reversible release of active Cdk9-cyclin T modules from large, inactive 7SK small nuclear ribonucleoprotein particle (snRNP) complexes. Here, we have identified an alternative pathway of irreversible P-TEFb activation in megakaryopoiesis that is mediated by dissolution of the 7SK snRNP complex. In this pathway, calpain 2 cleavage of the core 7SK snRNP component MePCE promoted P-TEFb release and consequent upregulation of a cohort of cytoskeleton remodeling factors, including α-actinin-1. In a subset of human megakaryocytic leukemias, the transcription factor GATA1 undergoes truncating mutation (GATA1s). Here, we linked the GATA1s mutation to defects in megakaryocytic upregulation of calpain 2 and of P-TEFb-dependent cytoskeletal remodeling factors. Restoring calpain 2 expression in GATA1s mutant megakaryocytes rescued normal development, implicating this morphogenetic pathway as a target in human leukemogenesis.


Subject(s)
Calpain/physiology , Cell Transformation, Neoplastic/pathology , GATA1 Transcription Factor/genetics , Leukemia/pathology , Megakaryocytes/pathology , Mutation/genetics , Positive Transcriptional Elongation Factor B/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Actinin/genetics , Actinin/metabolism , Animals , Blotting, Western , Cell Differentiation , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Flow Cytometry , GATA1 Transcription Factor/metabolism , Humans , Immunoprecipitation , Leukemia/metabolism , Megakaryocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Morphogenesis , Positive Transcriptional Elongation Factor B/genetics , Protein Binding , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoproteins, Small Nuclear/genetics , Transcription, Genetic
10.
J Biol Chem ; 287(23): 19207-15, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22514271

ABSTRACT

Signaling via the intracellular second messenger cyclic AMP (cAMP) has long been implicated in the repression of megakaryocytic differentiation. However, the mechanisms by which cAMP signaling impairs megakaryopoiesis have never been elucidated. In a human CD34(+) cell culture model, we show that the adenylyl cyclase agonist forskolin inhibits megakaryocytic differentiation in a protein kinase A-dependent manner. Using this system to screen for downstream effectors, we identified the transcription factor E2A as a key target in a novel repressive signaling pathway. Specifically, forskolin acting through protein kinase A-induced E2A down-regulation and enforced expression of E2A overrode the inhibitory effects of forskolin on megakaryopoiesis. The dependence of megakaryopoiesis on critical thresholds of E2A expression was confirmed in vivo in haploinsufficient mice and ex vivo using shRNA knockdown in human progenitors. Using a variety of approaches, we further identified p21 (encoded by CDKN1A) as a functionally important megakaryopoietic regulator residing downstream of E2A. These results thus implicate the E2A-CDKN1A transcriptional axis in the control of megakaryopoiesis and reveal the lineage-selective inhibition of this axis as a likely mechanistic basis for the inhibitory effects of cAMP signaling.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cyclic AMP/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Megakaryocytes/metabolism , Second Messenger Systems/physiology , Thrombopoiesis/physiology , Transcription, Genetic/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/physiology , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , HEK293 Cells , Humans , Megakaryocytes/cytology , Mice , Mice, Mutant Strains
11.
Nat Chem Biol ; 5(4): 236-43, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19172146

ABSTRACT

It has been proposed that inhibitors of an oncogene's effects on multipotent hematopoietic progenitor cell differentiation may change the properties of the leukemic stem cells and complement the clinical use of cytotoxic drugs. Using zebrafish, we developed a robust in vivo hematopoietic differentiation assay that reflects the activity of the oncogene AML1-ETO. Screening for modifiers of AML1-ETO-mediated hematopoietic dysregulation uncovered unexpected roles of COX-2- and beta-catenin-dependent pathways in AML1-ETO function. This approach may open doors for developing therapeutics targeting oncogene function within leukemic stem cells.


Subject(s)
Oncogene Proteins/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation , Dinoprostone , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , K562 Cells , Nitrobenzenes , Oncogene Proteins/genetics , Small Molecule Libraries , Sulfonamides , Transcription Factors , Zebrafish , beta Catenin
12.
Blood ; 112(13): 4884-94, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18780834

ABSTRACT

The transcription factor GATA-1 participates in programming the differentiation of multiple hematopoietic lineages. In megakaryopoiesis, loss of GATA-1 function produces complex developmental abnormalities and underlies the pathogenesis of megakaryocytic leukemia in Down syndrome. Its distinct functions in megakaryocyte and erythroid maturation remain incompletely understood. In this study, we identified functional and physical interaction of GATA-1 with components of the positive transcriptional elongation factor P-TEFb, a complex containing cyclin T1 and the cyclin-dependent kinase 9 (Cdk9). Megakaryocytic induction was associated with dynamic changes in endogenous P-TEFb composition, including recruitment of GATA-1 and dissociation of HEXIM1, a Cdk9 inhibitor. shRNA knockdowns and pharmacologic inhibition both confirmed contribution of Cdk9 activity to megakaryocytic differentiation. In mice with megakaryocytic GATA-1 deficiency, Cdk9 inhibition produced a fulminant but reversible megakaryoblastic disorder reminiscent of the transient myeloproliferative disorder of Down syndrome. P-TEFb has previously been implicated in promoting elongation of paused RNA polymerase II and in programming hypertrophic differentiation of cardiomyocytes. Our results offer evidence for P-TEFb cross-talk with GATA-1 in megakaryocytic differentiation, a program with parallels to cardiomyocyte hypertrophy.


Subject(s)
Cell Differentiation , Cyclin-Dependent Kinase 9/physiology , GATA1 Transcription Factor/metabolism , Megakaryocytes/cytology , Positive Transcriptional Elongation Factor B/metabolism , Receptor Cross-Talk , Animals , Cells, Cultured , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Down Syndrome , GATA1 Transcription Factor/genetics , Humans , Mice , Mice, Knockout , Myeloproliferative Disorders
13.
Crit Rev Eukaryot Gene Expr ; 17(4): 271-80, 2007.
Article in English | MEDLINE | ID: mdl-17725493

ABSTRACT

Runt-related transcription factor 1 (RUNX1) and GATA-1 are both transcription factors known to play essential roles in hematopoiesis. Genetic alterations of each are associated with abnormal platelet development, as well as predisposition to leukemia. In addition, in vitro and animal studies indicate that both factors are involved in megakaryopoiesis. We and others have previously shown that RUNX1 and GATA-1 physically interact and cooperate in the activation of megakaryocytic promoters such as alpha IIb integrin and glycoprotein Ibalpha. Moreover, transcriptional cooperation of RUNX1 with GATA-1 is conserved back to Drosophila in which RUNX1 and GATA-1 homologs cooperate in crystal cell development. In this article, we will review the molecular and functional significance of the transcriptional cross talk between RUNX1 and GATA-1. In particular, we will elaborate on recent data which suggest that GATA-1 targets RUNX1 for modification, in particular phosphorylation by cyclin-dependent kinases. Furthermore, targeting of RUNX1 by GATA-1 for phosphorylation may convert RUNX1 from a repressor to an activator. This is a potential mechanism of transcriptional cooperation and may be an essential step in megakaryocytic differentiation.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , GATA1 Transcription Factor/physiology , Gene Expression Regulation/physiology , Transcription, Genetic/physiology , Animals , GATA1 Transcription Factor/genetics , Humans , Megakaryocytes/cytology , Mice , Mutation , Phosphorylation
14.
Cancer Lett ; 251(2): 179-86, 2007 Jun 28.
Article in English | MEDLINE | ID: mdl-17125917

ABSTRACT

The leukemic fusion protein AML1-ETO occurs frequently in human acute myeloid leukemia (AML) and has received much attention over the past decade. An initial model for its pathogenetic effects emphasized the conversion of a hematopoietic transcriptional activator, RUNX1 (or AML1), into a leukemogenic repressor which blocked myeloid differentiation at the level of target gene regulation. This view has been absorbed into a larger picture of AML1-ETO pathogenesis, encompassing dysregulation of hematopoietic stem cell homeostasis at several mechanistic levels. Recent reports have highlighted a multifaceted capacity of AML1-ETO directly to inhibit key hematopoietic transcription factors that function as tumor suppressors at several nodal points during hematopoietic differentiation. A new model is presented in which AML1-ETO coordinates expansion of the stem cell compartment with diminished lineage commitment and with genome instability.


Subject(s)
Bone Marrow/physiology , Core Binding Factor Alpha 2 Subunit/metabolism , Hematopoiesis , Leukemia, Myeloid/metabolism , Oncogene Proteins, Fusion/metabolism , Acute Disease , Animals , Cell Differentiation , Hematopoietic Stem Cells/physiology , Humans , Models, Biological , Mutation , RUNX1 Translocation Partner 1 Protein , Tumor Suppressor Proteins/drug effects
15.
Cancer Res ; 66(6): 2990-6, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16540647

ABSTRACT

Human acute myeloid leukemias with the t(8;21) translocation express the AML1-ETO fusion protein in the hematopoietic stem cell compartment and show impairment in erythroid differentiation. This clinical finding is reproduced in multiple murine and cell culture model systems in which AML1-ETO specifically interferes with erythroid maturation. Using purified normal human early hematopoietic progenitor cells, we find that AML1-ETO impedes the earliest discernable steps of erythroid lineage commitment. Correspondingly, GATA-1, a central transcriptional regulator of erythroid differentiation, undergoes repression by AML1-ETO in a nonconventional histone deacetylase-independent manner. In particular, GATA-1 acetylation by its transcriptional coactivator, p300/CBP, a critical regulatory step in programming erythroid development, is efficiently blocked by AML1-ETO. Fusion of a heterologous E1A coactivator recruitment module to GATA-1 overrides the inhibitory effects of AML1-ETO on GATA-1 acetylation and transactivation. Furthermore, the E1A-GATA-1 fusion, but not wild-type GATA-1, rescues erythroid lineage commitment in primary human progenitors expressing AML1-ETO. These results ascribe a novel repressive mechanism to AML1-ETO, blockade of GATA-1 acetylation, which correlates with its inhibitory effects on primary erythroid lineage commitment.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Erythroid Precursor Cells/physiology , GATA1 Transcription Factor/metabolism , Oncogene Proteins, Fusion/physiology , Acetylation , Antigens, CD34/biosynthesis , Antigens, CD34/immunology , CD36 Antigens/biosynthesis , CD36 Antigens/immunology , Cell Differentiation/physiology , Cell Line , Cell Lineage , Core Binding Factor Alpha 2 Subunit/biosynthesis , Core Binding Factor Alpha 2 Subunit/genetics , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/immunology , Erythroid Precursor Cells/metabolism , Humans , K562 Cells , Oncogene Proteins, Fusion/biosynthesis , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein , Transcriptional Activation , Transfection , Zinc Fingers/physiology , p300-CBP Transcription Factors/metabolism
16.
Crit Rev Eukaryot Gene Expr ; 15(3): 207-16, 2005.
Article in English | MEDLINE | ID: mdl-16390317

ABSTRACT

The chromosomal translocation t(8;21), generating the AML1-ETO fusion protein, is frequently associated with French-American-British (FAB) type M2 acute myeloid leukemia (AML). t(8;21) fuses the runt domain from the hematopoietic transcription factor RUNX1 with almost the entire transcriptional repressor ETO. AML1-ETO inhibits normal definitive hematopoiesis and blocks erythroid differentiation. Several mechanistic models for the role of AML1-ETO in leukemia development have emerged over the last decade. Most of these models have emphasized the capacity of the fusion protein to redirect repressive cofactors, such as histone deacetylases (HDACs) and DNA methyltransferases (DNMTs), to RUNX target genes, thereby reversing the hematopoietic transcriptional program activated by wild-type RUNX1a phenomenon referred to collectively in this review as the "classical" corepressor model. Because erythropoiesis occurs in a RUNX-independent manner, this dominant-negative "classical" model cannot explain the prominent repression of red-cell development by AML1-ETO. This review will consider the clinical and mechanistic significance of erythroid inhibition by AML1-ETO. Additional models to account for this mysterious oncogenic function are proposed.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Erythroid Cells/cytology , Oncogene Proteins, Fusion/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Chromosomes, Human, Pair 21 , Chromosomes, Human, Pair 8 , Core Binding Factor Alpha 2 Subunit/genetics , Erythropoiesis/genetics , GATA1 Transcription Factor/physiology , Histone Deacetylases/genetics , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/genetics , RUNX1 Translocation Partner 1 Protein , Signal Transduction/genetics , Translocation, Genetic
17.
Mol Cell Biol ; 24(17): 7779-94, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15314183

ABSTRACT

Although Jun upregulation and activation have been established as critical to oncogenesis, the relevant downstream pathways remain incompletely characterized. In this study, we found that c-Jun blocks erythroid differentiation in primary human hematopoietic progenitors and, correspondingly, that Jun factors block transcriptional activation by GATA-1, the central regulator of erythroid differentiation. Mutagenesis of c-Jun suggested that its repression of GATA-1 occurs through a transcriptional mechanism involving activation of downstream genes. We identified the hairy-enhancer-of-split-related factor HERP2 as a novel gene upregulated by c-Jun. HERP2 showed physical interaction with GATA-1 and repressed GATA-1 transcriptional activation. Furthermore, transduction of HERP2 into primary human hematopoietic progenitors inhibited erythroid differentiation. These results thus define a novel regulatory pathway linking the transcription factors c-Jun, HERP2, and GATA-1. Furthermore, these results establish a connection between the Notch signaling pathway, of which the HERP factors are a critical component, and the GATA family, which participates in programming of cellular differentiation.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Erythropoiesis/physiology , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Antigens, CD34 , Basic Helix-Loop-Helix Transcription Factors , Cell Cycle Proteins/genetics , Cell Differentiation , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Helix-Loop-Helix Motifs , Hematopoietic Stem Cells/cytology , Humans , K562 Cells , Proto-Oncogene Proteins c-jun/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Transcription, Genetic
18.
Blood ; 101(11): 4333-41, 2003 Jun 01.
Article in English | MEDLINE | ID: mdl-12576332

ABSTRACT

Megakaryocytic and erythroid lineages derive from a common bipotential progenitor and share many transcription factors, most prominently factors of the GATA zinc-finger family. Little is known about transcription factors unique to the megakaryocytic lineage that might program divergence from the erythroid pathway. To identify such factors, we used the K562 system in which megakaryocyte lineage commitment is dependent on sustained extracellular regulatory kinase (ERK) activation and is inhibited by stromal cell contact. During megakaryocytic induction in this system, the myeloid transcription factor RUNX1 underwent up-regulation, dependent on ERK signaling and inhibitable by stromal cell contact. Immunostaining of healthy human bone marrow confirmed a strong expression of RUNX1 and its cofactor, core-binding factor beta (CBFbeta), in megakaryocytes and a minimal expression in erythroblasts. In primary human hematopoietic progenitor cultures, RUNX1 and CBFbeta up-regulation preceded megakaryocytic differentiation, and down-regulation of these factors preceded erythroid differentiation. Functional studies showed cooperation among RUNX1, CBFbeta, and GATA-1 in the activation of a megakaryocytic promoter. By contrast, the RUNX1-ETO leukemic fusion protein potently repressed GATA-1-mediated transactivation. These functional interactions correlated with physical interactions observed between GATA-1 and RUNX1 factors. Enforced RUNX1 expression in K562 cells enhanced the induction of the megakaryocytic integrin proteins alphaIIb and alpha2. These results suggest that RUNX1 may participate in the programming of megakaryocytic lineage commitment through functional and physical interactions with GATA transcription factors. By contrast, RUNX1-ETO inhibition of GATA function may constitute a potential mechanism for the blockade of erythroid and megakaryocytic differentiation seen in leukemias with t(8;21).


Subject(s)
DNA-Binding Proteins/physiology , Megakaryocytes/cytology , Proto-Oncogene Proteins , Transcription Factors/physiology , Cell Differentiation/physiology , Cell Lineage/physiology , Core Binding Factor Alpha 2 Subunit , DNA-Binding Proteins/biosynthesis , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Gene Expression Regulation , Humans , Integrin alpha2/analysis , K562 Cells , Leukemia/etiology , Megakaryocytes/chemistry , Oncogene Proteins, Fusion/physiology , Platelet Membrane Glycoprotein IIb/analysis , Transcription Factor AP-2 , Transcription Factors/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...