1
|
Fagnan A, Aid Z, Baille M, Drakul A, Robert E, Lopez CK, Thirant C, Lecluse Y, Rivière J, Ignacimouttou C, Salmoiraghi S, Anguita E, Naimo A, Marzac C, Pflumio F, Malinge S, Wichmann C, Huang Y, Lobry C, Chaumeil J, Soler E, Bourquin J, Nerlov C, Bernard OA, Schwaller J, Mercher T. The ETO2 transcriptional cofactor maintains acute leukemia by driving a MYB/EP300-dependent stemness program. Hemasphere 2024; 8:e90. [PMID: 38903535 PMCID: PMC11187848 DOI: 10.1002/hem3.90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/13/2024] [Accepted: 05/01/2024] [Indexed: 06/22/2024] Open
Abstract
Transcriptional cofactors of the ETO family are recurrent fusion partners in acute leukemia. We characterized the ETO2 regulome by integrating transcriptomic and chromatin binding analyses in human erythroleukemia xenografts and controlled ETO2 depletion models. We demonstrate that beyond its well-established repressive activity, ETO2 directly activates transcription of MYB, among other genes. The ETO2-activated signature is associated with a poorer prognosis in erythroleukemia but also in other acute myeloid and lymphoid leukemia subtypes. Mechanistically, ETO2 colocalizes with EP300 and MYB at enhancers supporting the existence of an ETO2/MYB feedforward transcription activation loop (e.g., on MYB itself). Both small-molecule and PROTAC-mediated inhibition of EP300 acetyltransferases strongly reduced ETO2 protein, chromatin binding, and ETO2-activated transcripts. Taken together, our data show that ETO2 positively enforces a leukemia maintenance program that is mediated in part by the MYB transcription factor and that relies on acetyltransferase cofactors to stabilize ETO2 scaffolding activity.
Collapse
Affiliation(s)
- Alexandre Fagnan
- Gustave Roussy, INSERM U1170Université Paris‐SaclayVillejuifFrance
- Equipe Labellisée Ligue Contre le CancerParisFrance
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Zakia Aid
- Gustave Roussy, INSERM U1170Université Paris‐SaclayVillejuifFrance
- Equipe Labellisée Ligue Contre le CancerParisFrance
| | - Marie Baille
- Gustave Roussy, INSERM U1170Université Paris‐SaclayVillejuifFrance
- Equipe Labellisée Ligue Contre le CancerParisFrance
| | - Aneta Drakul
- Division of Oncology and Children's Research CentreUniversity Children's Hospital ZurichZurichSwitzerland
| | - Elie Robert
- Gustave Roussy, INSERM U1170Université Paris‐SaclayVillejuifFrance
- Equipe Labellisée Ligue Contre le CancerParisFrance
| | - Cécile K. Lopez
- Gustave Roussy, INSERM U1170Université Paris‐SaclayVillejuifFrance
- Equipe Labellisée Ligue Contre le CancerParisFrance
| | - Cécile Thirant
- Gustave Roussy, INSERM U1170Université Paris‐SaclayVillejuifFrance
- Equipe Labellisée Ligue Contre le CancerParisFrance
| | - Yann Lecluse
- Gustave Roussy, Plateforme Imagerie et Cytométrie, Université Paris‐Saclay, UMS AMMICA, INSERM US23, CNRS UMS 3655VillejuifFrance
| | - Julie Rivière
- Gustave Roussy, INSERM U1170Université Paris‐SaclayVillejuifFrance
- Equipe Labellisée Ligue Contre le CancerParisFrance
| | - Cathy Ignacimouttou
- Gustave Roussy, INSERM U1170Université Paris‐SaclayVillejuifFrance
- Equipe Labellisée Ligue Contre le CancerParisFrance
| | - Silvia Salmoiraghi
- Department of Oncology and HematologyAzienda Socio Sanitaria Territoriale Papa Giovanni XXIII, FROM Research Foundation, Papa Giovanni XXIII HospitalBergamoItaly
| | - Eduardo Anguita
- Hematology Department, Hospital Clínico San Carlos (HCSC), IML, IdISSC, Department of MedicineUniversidad Complutense de Madrid (UCM)MadridSpain
| | - Audrey Naimo
- Gustave Roussy, Genomic PlatformUniversité Paris‐Saclay, UMS AMMICA, INSERM US23, CNRS UMS 3655VillejuifFrance
| | - Christophe Marzac
- Department of HematologyLeukemia Interception Program, Personalized Cancer Prevention Center, Gustave RoussyVillejuifFrance
| | - Françoise Pflumio
- Equipe Labellisée Ligue Contre le CancerParisFrance
- Unité de Recherche (UMR)‐E008 Stabilité Génétique, Cellules Souches et Radiations, Team Niche and Cancer in Hematopoiesis, Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA)Université de Paris‐Université Paris‐SaclayFontenay‐aux‐RosesFrance
- OPALE Carnot Institute, The Organization for Partnerships in LeukemiaParisFrance
| | - Sébastien Malinge
- Gustave Roussy, INSERM U1170Université Paris‐SaclayVillejuifFrance
- Telethon Kids Institute, Perth Children's HospitalNedlandsAustralia
| | - Christian Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and HaemostasisLudwig‐Maximilians‐University of MunichMunichGermany
| | - Yun Huang
- Division of Oncology and Children's Research CentreUniversity Children's Hospital ZurichZurichSwitzerland
| | - Camille Lobry
- Gustave Roussy, INSERM U1170Université Paris‐SaclayVillejuifFrance
- INSERM U944, CNRS UMR7212Institut de Recherche Saint Louis and Université de ParisParisFrance
| | - Julie Chaumeil
- Université de Paris, Institut Cochin, INSERM, CNRSParisFrance
| | - Eric Soler
- IGMM, University of Montpellier, CNRS, Montpellier, France & Université de Paris, Laboratory of Excellence GR‐ExParisFrance
| | - Jean‐Pierre Bourquin
- Division of Oncology and Children's Research CentreUniversity Children's Hospital ZurichZurichSwitzerland
| | - Claus Nerlov
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | | | - Juerg Schwaller
- Department of BiomedicineUniversity Children's Hospital Beider Basel (UKBB), University of BaselBaselSwitzerland
| | - Thomas Mercher
- Gustave Roussy, INSERM U1170Université Paris‐SaclayVillejuifFrance
- Equipe Labellisée Ligue Contre le CancerParisFrance
- OPALE Carnot Institute, The Organization for Partnerships in LeukemiaParisFrance
| |
Collapse
|
2
|
Takasaki K, Chou ST. GATA1 in Normal and Pathologic Megakaryopoiesis and Platelet Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:261-287. [PMID: 39017848 DOI: 10.1007/978-3-031-62731-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
GATA1 is a highly conserved hematopoietic transcription factor (TF), essential for normal erythropoiesis and megakaryopoiesis, that encodes a full-length, predominant isoform and an amino (N) terminus-truncated isoform GATA1s. It is consistently expressed throughout megakaryocyte development and interacts with its target genes either independently or in association with binding partners such as FOG1 (friend of GATA1). While the N-terminus and zinc finger have classically been demonstrated to be necessary for the normal regulation of platelet-specific genes, murine models, cell-line studies, and human case reports indicate that the carboxy-terminal activation domain and zinc finger also play key roles in precisely controlling megakaryocyte growth, proliferation, and maturation. Murine models have shown that disruptions to GATA1 increase the proliferation of immature megakaryocytes with abnormal architecture and impaired terminal differentiation into platelets. In humans, germline GATA1 mutations result in variable cytopenias, including macrothrombocytopenia with abnormal platelet aggregation and excessive bleeding tendencies, while acquired GATA1s mutations in individuals with trisomy 21 (T21) result in transient abnormal myelopoiesis (TAM) and myeloid leukemia of Down syndrome (ML-DS) arising from a megakaryocyte-erythroid progenitor (MEP). Taken together, GATA1 plays a key role in regulating megakaryocyte differentiation, maturation, and proliferative capacity. As sequencing and proteomic technologies expand, additional GATA1 mutations and regulatory mechanisms contributing to human diseases of megakaryocytes and platelets are likely to be revealed.
Collapse
Affiliation(s)
- Kaoru Takasaki
- Department of Pediatrics, Division of Hematology, University of Pennsylvania Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stella T Chou
- Department of Pediatrics, Division of Hematology, University of Pennsylvania Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Division of Transfusion Medicine, University of Pennsylvania Perelman School of Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Fechner J, Lausen J. Transcription Factor TAL1 in Erythropoiesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:243-258. [PMID: 39017847 DOI: 10.1007/978-3-031-62731-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Lineage-specific transcription factors (TFs) regulate differentiation of hematopoietic stem cells (HSCs). They are decisive for the establishment and maintenance of lineage-specific gene expression programs during hematopoiesis. For this they create a regulatory network between TFs, epigenetic cofactors, and microRNAs. They activate cell-type specific genes and repress competing gene expression programs. Disturbance of this process leads to impaired lineage fidelity and diseases of the blood system. The TF T-cell acute leukemia 1 (TAL1) is central for erythroid differentiation and contributes to the formation of distinct gene regulatory complexes in progenitor cells and erythroid cells. A TAL1/E47 heterodimer binds to DNA with the TFs GATA-binding factor 1 and 2 (GATA1/2), the cofactors LIM domain only 1 and 2 (LMO1/2), and LIM domain-binding protein 1 (LDB1) to form a core TAL1 complex. Furthermore, cell-type-dependent interactions of TAL1 with other TFs such as with runt-related transcription factor 1 (RUNX1) and Kruppel-like factor 1 (KLF1) are established. Moreover, TAL1 activity is regulated by the formation of TAL1 isoforms, posttranslational modifications (PTMs), and microRNAs. Here, we describe the function of TAL1 in normal hematopoiesis with a focus on erythropoiesis.
Collapse
Affiliation(s)
- Johannes Fechner
- Department of Eukaryotic Genetics, Institute of Biomedical Genetics, University of Stuttgart, Stuttgart, Germany
| | - Jörn Lausen
- Department of Eukaryotic Genetics, Institute of Biomedical Genetics, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
4
|
Huang BJ, Shannon K. NFIA-ETO2, TP53, and erythroid leukemogenesis. Blood 2023; 141:2168-2170. [PMID: 37140954 DOI: 10.1182/blood.2023019856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
|
5
|
Liu F, Hu C, Ding J, Fu C, Wang S, Li T. GATA-1 Promotes Erythroid Differentiation Through the Upregulation of miR-451a and miR-210-3p Expressions in CD34 + Cells in High-Altitude Polycythemia. High Alt Med Biol 2023; 24:59-67. [PMID: 36749159 DOI: 10.1089/ham.2022.0095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Liu, Fang, Caiyan Hu, Jin Ding, Chengbing Fu, Shuqiong Wang, and Tiantian Li. GATA-1 promotes erythroid differentiation through the upregulation of miR-451a and miR-210-3p expression in CD34+ cells in high-altitude polycythemia. High Alt Med Biol. 24:59-67, 2023. Background: The clinical manifestations of high-altitude polycythemia (HAPC) include excessive accumulation of erythrocytes, and its pathogenesis is not yet clear. Methods: Peripheral blood was collected from 10 HAPC patients (HAPC group) and normal individuals (control group) each. CD34+ cells were sorted using immunomagnetic beads and differentiated into erythroid cells for 7, 11, and 15 days. Changes in GATA-binding protein 1 (GATA-1), miR-451a, and miR-210-3p expression and their possible regulatory relationships were investigated. Results: Under hypoxia, GATA-1 expression on day 15 was about 2.4 times that on day 7 in the control group and about 1.3 times that on day 7 in the HAPC group, which was significantly lower compared with the control group. miR-451a and miR-210-3p expressions in the HAPC group were 2.6 and 1.8 times that in the control group, respectively, and were significantly increased. When GATA-1 was inhibited, miR-451a and miR-210-3p expressions were significantly decreased by 0.43 and 0.39 times, respectively, compared with those in the control group. Conclusions: Hypoxia stimulated the upregulation of GATA-1 level and accelerated the change of expression, which promoted miR-451a and miR-210-3p expressions and shortened the time taken by cells to enter end-stage differentiation, so as to enhance erythroid differentiation, which may be a pathogenetic mechanism underlying HAPC polycytosis.
Collapse
Affiliation(s)
- Fang Liu
- Department of Biochemistry, Qing Hai University Medical College, Xi Ning, China
| | - Caiyan Hu
- Baoding First Central Hospital, Bao Ding, China
| | - Jin Ding
- Xi' an Central Blood Bank, Xi' an, China
| | - Chengbing Fu
- Department of Biochemistry, Qing Hai University Medical College, Xi Ning, China
| | - Shuqiong Wang
- Department of Biochemistry, Qing Hai University Medical College, Xi Ning, China
| | - Tiantian Li
- Department of Biochemistry, Qing Hai University Medical College, Xi Ning, China
| |
Collapse
|
6
|
McCabe CF, Goodrich JM, Bakulski KM, Domino SE, Jones TR, Colacino J, Dolinoy DC, Padmanabhan V. Probing prenatal bisphenol exposures and tissue-specific DNA methylation responses in cord blood, cord tissue, and placenta. Reprod Toxicol 2023; 115:74-84. [PMID: 36473650 PMCID: PMC9851062 DOI: 10.1016/j.reprotox.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
The early-gestational fetal epigenome establishes the landscape for fetal development and is susceptible to disruption via environmental stressors including chemical exposures. Research has explored how cell- and tissue-type-specific epigenomic signatures contribute to human disease, but how the epigenome in each tissue comparatively responds to environmental exposures is largely unknown. This pilot study compared DNA methylation in four previously identified genes across matched cord blood (CB), cord tissue (CT), and placental (PL) samples from 28 mother-infant pairs in tthe Michigan Mother Infant Pairs study; evaluated association between prenatal exposure to bisphenols (BPA, BPF, and BPS) and DNA methylation (DNAm) by tissue type; compared epigenome-wide DNAm of CB and PL; and explored associations between prenatal bisphenol exposures and epigenome-wide DNAm in PL. Bisphenol concentrations were quantified in first-trimester maternal urine. DNAm was assessed at four genes via pyrosequencing in three tissues; epigenome-wide DNAm analysis via Infinium MethylationEPIC array was completed on CB and PL. Candidate gene analysis revealed tissue-specific differences across all genes. In adjusted linear regression, BPA and BPF were associated with DNAm across candidate genes in PL but not CB and CT. Epigenome-wide comparison of matched CB and PL DNAm revealed tissue-specific differences at most CpG sites and modest associations between maternal first-trimester bisphenol exposures and PL but not CB DNAm. These data endorse inclusion of a variety of tissues in prenatal exposure studies. Overlapping and divergent responses in CB, CT, and PL demonstrate their utility in combination to capture a fuller picture of the epigenetic effect of developmental exposures.
Collapse
Affiliation(s)
- Carolyn F McCabe
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jaclyn M Goodrich
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Kelly M Bakulski
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Steven E Domino
- Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Tamara R Jones
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Justin Colacino
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Dana C Dolinoy
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA; Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA; Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
7
|
Exploring the mechanistic link between SF3B1 mutation and ring sideroblast formation in myelodysplastic syndrome. Sci Rep 2022; 12:14562. [PMID: 36028755 PMCID: PMC9418223 DOI: 10.1038/s41598-022-18921-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Acquired sideroblastic anemia, characterized by bone marrow ring sideroblasts (RS), is predominantly associated with myelodysplastic syndrome (MDS). Although somatic mutations in splicing factor 3b subunit 1 (SF3B1), which is involved in the RNA splicing machinery, are frequently found in MDS-RS, the detailed mechanism contributing to RS formation is unknown. To explore the mechanism, we established human umbilical cord blood-derived erythroid progenitor-2 (HUDEP-2) cells stably expressing SF3B1K700E. SF3B1K700E expressing cells showed higher proportion of RS than the control cells along with erythroid differentiation, indicating the direct contribution of mutant SF3B1 expression in erythroblasts to RS formation. In SF3B1K700E expressing cells, ABCB7 and ALAS2, known causative genes for congenital sideroblastic anemia, were downregulated. Additionally, mis-splicing of ABCB7 was observed in SF3B1K700E expressing cells. ABCB7-knockdown HUDEP-2 cells revealed an increased frequency of RS formation along with erythroid differentiation, demonstrating the direct molecular link between ABCB7 defects and RS formation. ALAS2 protein levels were obviously decreased in ABCB7-knockdown cells, indicating decreased ALAS2 translation owing to impaired Fe–S cluster export by ABCB7 defects. Finally, RNA-seq analysis of MDS clinical samples demonstrated decreased expression of ABCB7 by the SF3B1 mutation. Our findings contribute to the elucidation of the complex mechanisms of RS formation in MDS-RS.
Collapse
|
8
|
Determination of the dynamic cellular transcriptional profiles during kidney development from birth to maturity in rats by single-cell RNA sequencing. Cell Death Discov 2021; 7:162. [PMID: 34226524 PMCID: PMC8257621 DOI: 10.1038/s41420-021-00542-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/27/2021] [Accepted: 06/03/2021] [Indexed: 01/02/2023] Open
Abstract
Recent single-cell RNA sequencing (scRNA-seq) analyses have offered much insight into the gene expression profiles in early-stage kidney development. However, comprehensive gene expression profiles from mid- and late-stage kidney development are lacking. In the present study, by using the scRNA-seq technique, we analyzed 54,704 rat kidney cells from just after birth to adulthood (six time points: postnatal days 0, 2, 5, 10, 20, and 56) including the mid and late stages of kidney development. Twenty-five original clusters and 13 different cell types were identified during these stages. Gene expression in these 13 cell types was mapped, and single cell atlas of the rat kidney from birth to maturity ( http://youngbearlab.com ) was built to enable users to search for a gene of interest and to evaluate its expression in different cells. The variation trend of six major types of kidney cells-intercalated cells of the collecting duct (CD-ICs), principal cells of the collecting duct (CD-PCs), cells of the distal convoluted tubules (DCTs), cells of the loop of Henle (LOH), podocytes (PDs), and cells of the proximal tubules (PTs)-during six postnatal time points was demonstrated. The trajectory of rat kidney development and the order of induction of the six major types of kidney cells from just after birth to maturity were determined. In addition, features of the dynamically changing genes as well as transcription factors during postnatal rat kidney development were identified. The present study provides a resource for achieving a deep understanding of the molecular basis of and regulatory events in the mid and late stages of kidney development.
Collapse
|
9
|
Jakobczyk H, Debaize L, Soubise B, Avner S, Rouger-Gaudichon J, Commet S, Jiang Y, Sérandour AA, Rio AG, Carroll JS, Wichmann C, Lie-A-Ling M, Lacaud G, Corcos L, Salbert G, Galibert MD, Gandemer V, Troadec MB. Reduction of RUNX1 transcription factor activity by a CBFA2T3-mimicking peptide: application to B cell precursor acute lymphoblastic leukemia. J Hematol Oncol 2021; 14:47. [PMID: 33743795 PMCID: PMC7981807 DOI: 10.1186/s13045-021-01051-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/24/2021] [Indexed: 12/27/2022] Open
Abstract
Background B Cell Precursor Acute Lymphoblastic Leukemia (BCP-ALL) is the most common pediatric cancer. Identifying key players involved in proliferation of BCP-ALL cells is crucial to propose new therapeutic targets. Runt Related Transcription Factor 1 (RUNX1) and Core-Binding Factor Runt Domain Alpha Subunit 2 Translocated To 3 (CBFA2T3, ETO2, MTG16) are master regulators of hematopoiesis and are implicated in leukemia. Methods We worked with BCP-ALL mononuclear bone marrow patients’ cells and BCP-ALL cell lines, and performed Chromatin Immunoprecipitations followed by Sequencing (ChIP-Seq), co-immunoprecipitations (co-IP), proximity ligation assays (PLA), luciferase reporter assays and mouse xenograft models. Results We demonstrated that CBFA2T3 transcript levels correlate with RUNX1 expression in the pediatric t(12;21) ETV6-RUNX1 BCP-ALL. By ChIP-Seq in BCP-ALL patients’ cells and cell lines, we found that RUNX1 is recruited on its promoter and on an enhancer of CBFA2T3 located − 2 kb upstream CBFA2T3 promoter and that, subsequently, the transcription factor RUNX1 drives both RUNX1 and CBFA2T3 expression. We demonstrated that, mechanistically, RUNX1 and CBFA2T3 can be part of the same complex allowing CBFA2T3 to strongly potentiate the activity of the transcription factor RUNX1. Finally, we characterized a CBFA2T3-mimicking peptide that inhibits the interaction between RUNX1 and CBFA2T3, abrogating the activity of this transcription complex and reducing BCP-ALL lymphoblast proliferation. Conclusions Altogether, our findings reveal a novel and important activation loop between the transcription regulator CBFA2T3 and the transcription factor RUNX1 that promotes BCP-ALL proliferation, supporting the development of an innovative therapeutic approach based on the NHR2 subdomain of CBFA2T3 protein. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-021-01051-z.
Collapse
Affiliation(s)
- Hélène Jakobczyk
- Univ Rennes 1, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, 35000, Rennes, France
| | - Lydie Debaize
- Univ Rennes 1, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, 35000, Rennes, France
| | - Benoit Soubise
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 29200, Brest, France
| | - Stéphane Avner
- Univ Rennes 1, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, 35000, Rennes, France
| | - Jérémie Rouger-Gaudichon
- Univ Rennes 1, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, 35000, Rennes, France.,Département d'onco-hematologie pediatrique, Centre Hospitalier Universitaire de Caen Normandie, Caen, France
| | - Séverine Commet
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 29200, Brest, France.,CHRU Brest, Service de génétique, laboratoire de génétique chromosomique, 22 avenue Camille Desmoulins, 29238, Brest Cedex 3, France
| | - Yan Jiang
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 29200, Brest, France.,Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | | | - Anne-Gaëlle Rio
- Univ Rennes 1, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, 35000, Rennes, France
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Christian Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and Haemostasis, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Michael Lie-A-Ling
- Cancer Research UK Manchester Institute, University of Manchester, Aderley Park, Macclesfield, SK10 4TG, UK
| | - Georges Lacaud
- Cancer Research UK Manchester Institute, University of Manchester, Aderley Park, Macclesfield, SK10 4TG, UK
| | - Laurent Corcos
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 29200, Brest, France
| | - Gilles Salbert
- Univ Rennes 1, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, 35000, Rennes, France
| | - Marie-Dominique Galibert
- Univ Rennes 1, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, 35000, Rennes, France.,Service de Génétique et Génomique Moléculaire, Centre Hospitalier Universitaire de Rennes (CHU-Rennes), 35033, Rennes, France
| | - Virginie Gandemer
- Univ Rennes 1, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, 35000, Rennes, France.,Department of Pediatric Hemato-Oncology, Centre Hospitalier Universitaire de Rennes (CHU-Rennes), 35203, Rennes, France
| | - Marie-Bérengère Troadec
- Univ Rennes 1, CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, 35000, Rennes, France. .,Univ Brest, Inserm, EFS, UMR 1078, GGB, 29200, Brest, France. .,CHRU Brest, Service de génétique, laboratoire de génétique chromosomique, 22 avenue Camille Desmoulins, 29238, Brest Cedex 3, France.
| |
Collapse
|
10
|
Singbrant S, Mattebo A, Sigvardsson M, Strid T, Flygare J. Prospective isolation of radiation induced erythroid stress progenitors reveals unique transcriptomic and epigenetic signatures enabling increased erythroid output. Haematologica 2020; 105:2561-2571. [PMID: 33131245 PMCID: PMC7604643 DOI: 10.3324/haematol.2019.234542] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 01/02/2020] [Indexed: 11/09/2022] Open
Abstract
Massive expansion of erythroid progenitor cells is essential for surviving anemic stress. Research towards understanding this critical process, referred to as stress-erythropoiesis, has been hampered due to lack of specific marker-combinations enabling analysis of the distinct stress-progenitor cells capable of providing radioprotection and enhanced red blood cell production. Here we present a method for precise identification and in vivo validation of progenitor cells contributing to both steady-state and stress-erythropoiesis, enabling for the first time in-depth molecular characterization of these cells. Differential expression of surface markers CD150, CD9 and Sca1 defines a hierarchy of splenic stress-progenitors during irradiation-induced stress recovery in mice, and provides high-purity isolation of the functional stress-BFU-Es with a 100-fold improved enrichment compared to state-of-the-art. By transplanting purified stress-progenitors expressing the fluorescent protein Kusabira Orange, we determined their kinetics in vivo and demonstrated that CD150+CD9+Sca1- stress-BFU-Es provide a massive but transient radioprotective erythroid wave, followed by multi-lineage reconstitution from CD150+CD9+Sca1+ multi-potent stem/progenitor cells. Whole genome transcriptional analysis revealed that stress-BFU-Es express gene signatures more associated with erythropoiesis and proliferation compared to steady-state BFU-Es, and are BMP-responsive. Evaluation of chromatin accessibility through ATAC sequencing reveals enhanced and differential accessibility to binding sites of the chromatin-looping transcription factor CTCF in stress-BFU-Es compared to steady-state BFU-Es. Our findings offer molecular insight to the unique capacity of stress-BFU-Es to rapidly form erythroid cells in response to anemia and constitute an important step towards identifying novel erythropoiesis stimulating agents.
Collapse
Affiliation(s)
- Sofie Singbrant
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Alexander Mattebo
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Mikael Sigvardsson
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Tobias Strid
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Johan Flygare
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| |
Collapse
|
11
|
Jetten AM. Emerging Roles of GLI-Similar Krüppel-like Zinc Finger Transcription Factors in Leukemia and Other Cancers. Trends Cancer 2019; 5:547-557. [PMID: 31474360 DOI: 10.1016/j.trecan.2019.07.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 01/22/2023]
Abstract
GLI-similar 1-3 (GLIS1-3), a subfamily of Krüppel-like zinc finger transcription factors, function as key regulators of several biological processes important to oncogenesis, including control of cell proliferation, differentiation, self-renewal, and epithelial-mesenchymal transition. This review provides a short overview of the critical roles genetic changes in GLIS1-3 play in the development of several malignancies. This includes intrachromosomal translocations involving GLIS2 and ETO2/CBFA2T3 in the development of pediatric non-Down's syndrome (DS), acute megakaryoblastic leukemia (AMKL), a malignancy with poor prognosis, and an association of interchromosomal translocations between GLIS3, GLIS1, and PAX8, and between GLIS3 and CLPTM1L with hyalinizing trabecular tumors (HTTs) and fibrolamellar hepatocellular carcinoma (FHCC), respectively. Targeting upstream signaling pathways that regulate GLIS signaling may offer new therapeutic strategies in the management of cancer.
Collapse
Affiliation(s)
- Anton M Jetten
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 Alexander Drive, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
12
|
Emerging Roles of MTG16 in Cell-Fate Control of Hematopoietic Stem Cells and Cancer. Stem Cells Int 2017; 2017:6301385. [PMID: 29358956 PMCID: PMC5735743 DOI: 10.1155/2017/6301385] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 10/12/2017] [Accepted: 10/23/2017] [Indexed: 12/13/2022] Open
Abstract
MTG16 (myeloid translocation gene on chromosome 16) and its related proteins, MTG8 and MTGR1, define a small family of transcriptional corepressors. These corepressors share highly conserved domain structures yet have distinct biological functions and tissue specificity. In vivo studies have shown that, of the three MTG corepressors, MTG16 is uniquely important for the regulation of hematopoietic stem/progenitor cell (HSPC) proliferation and differentiation. Apart from this physiological function, MTG16 is also involved in carcinomas and leukemias, acting as the genetic target of loss of heterozygosity (LOH) aberrations in breast cancer and recurrent translocations in leukemia. The frequent involvement of MTG16 in these disease etiologies implies an important developmental role for this transcriptional corepressor. Furthermore, mounting evidence suggests that MTG16 indirectly alters the disease course of several leukemias via its regulatory interactions with a variety of pathologic fusion proteins. For example, a recent study has shown that MTG16 can repress not only wild-type E2A-mediated transcription, but also leukemia fusion protein E2A-Pbx1-mediated transcription, suggesting that MTG16 may serve as a potential therapeutic target in acute lymphoblastic leukemia expressing the E2A-Pbx1 fusion protein. Given that leukemia stem cells share similar regulatory pathways with normal HSPCs, studies to further understand how MTG16 regulates cell proliferation and differentiation could lead to novel therapeutic approaches for leukemia treatment.
Collapse
|
13
|
Lopez CK, Malinge S, Gaudry M, Bernard OA, Mercher T. Pediatric Acute Megakaryoblastic Leukemia: Multitasking Fusion Proteins and Oncogenic Cooperations. Trends Cancer 2017; 3:631-642. [PMID: 28867167 DOI: 10.1016/j.trecan.2017.07.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/10/2017] [Accepted: 07/17/2017] [Indexed: 02/06/2023]
Abstract
Pediatric leukemia presents specific clinical and genetic features from adult leukemia but the underpinning mechanisms of transformation are still unclear. Acute megakaryoblastic leukemia (AMKL) is the malignant accumulation of progenitors of the megakaryocyte lineage that normally produce blood platelets. AMKL is diagnosed de novo, in patients showing a poor prognosis, or in Down syndrome (DS) patients with a better prognosis. Recent data show that de novo AMKL is primarily associated with chromosomal alterations leading to the expression of fusions between transcriptional regulators. This review highlights the most recurrent genetic events found in de novo pediatric AMKL patients and, based on recent functional analyses, proposes a mechanism of leukemogenesis common to de novo and DS-AMKL.
Collapse
MESH Headings
- Age Factors
- Animals
- Carcinogenesis/genetics
- Carcinogenesis/metabolism
- Cell Differentiation/genetics
- Cell Lineage/genetics
- Child
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Megakaryoblastic, Acute/drug therapy
- Leukemia, Megakaryoblastic, Acute/etiology
- Leukemia, Megakaryoblastic, Acute/metabolism
- Leukemia, Megakaryoblastic, Acute/pathology
- Megakaryocytes/metabolism
- Megakaryocytes/pathology
- Molecular Targeted Therapy
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Cécile K Lopez
- INSERM U1170, Equipe Labellisée Ligue Contre le Cancer, 94800 Villejuif, France; Gustave Roussy, 94800 Villejuif, France; Université Paris-Sud, 91405 Orsay, France
| | - Sébastien Malinge
- INSERM U1170, Equipe Labellisée Ligue Contre le Cancer, 94800 Villejuif, France; Gustave Roussy, 94800 Villejuif, France; Université Paris Diderot, 75013 Paris, France
| | - Muriel Gaudry
- INSERM U1170, Equipe Labellisée Ligue Contre le Cancer, 94800 Villejuif, France; Gustave Roussy, 94800 Villejuif, France; Université Paris-Sud, 91405 Orsay, France
| | - Olivier A Bernard
- INSERM U1170, Equipe Labellisée Ligue Contre le Cancer, 94800 Villejuif, France; Gustave Roussy, 94800 Villejuif, France; Université Paris-Sud, 91405 Orsay, France
| | - Thomas Mercher
- INSERM U1170, Equipe Labellisée Ligue Contre le Cancer, 94800 Villejuif, France; Gustave Roussy, 94800 Villejuif, France; Université Paris-Sud, 91405 Orsay, France; Université Paris Diderot, 75013 Paris, France.
| |
Collapse
|
14
|
Jin S, Su H, Tran NT, Song J, Lu SS, Li Y, Huang S, Abdel-Wahab O, Liu Y, Zhao X. Splicing factor SF3B1K700E mutant dysregulates erythroid differentiation via aberrant alternative splicing of transcription factor TAL1. PLoS One 2017; 12:e0175523. [PMID: 28545085 PMCID: PMC5436638 DOI: 10.1371/journal.pone.0175523] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 03/27/2017] [Indexed: 11/18/2022] Open
Abstract
More than 60% of myeloid dysplasia syndrome (MDS) contains mutations in genes encoding for splicing factors such as SF3B1, U2AF, SRSF2 and ZRSR2. Mutations in SF3B1 are associated with 80% cases of refractory anemia with ring sideroblast (RARS), a subtype of MDS. SF3B1K700E is the most frequently mutated site among mutations on SF3B1. Yet the molecular mechanisms on how mutations of splicing factors lead to defective erythropoiesis are not clear. SF3B1K700E mutant binds to an RNA binding protein, RBM15, stronger than the wild type SF3B1 protein in co-immunoprecipitation assays. In addition, K700E mutant alters the RNA splicing of transcription factors TAL1 and GATA1. Via alternative RNA splicing, a novel short TAL1 transcript variant (TAL1s) is generated. Enhanced interaction between SF3B1 and RBM15 promotes the production of full-length TAL1 (TAL1fl) mRNA, while reduction of RBM15 protein level via PRMT1-mediated degradation pathway changes TAL1s/TAL1fl ratio in favor of TAL1s. TAL1s contains the helix-loop-helix DNA binding domain but not the N terminal region upstream of the DNA binding domain. The TAL1s protein loses its interaction with ETO2, which represses early erythropoiesis. In this vein, overexpression of TAL1s stimulates the transcription of β-hemoglobin in human leukemia K562 cells and promotes erythroid differentiation of human cord blood CD34+ cells cultured in erythropoietin-containing medium. Therefore, mutations of SF3B1 may block erythropoiesis via dysregulation of alternative RNA splicing of transcription factor TAL1, and targeting PRMT1 may alleviate the anemic symptoms in MDS patients.
Collapse
Affiliation(s)
- Shuiling Jin
- Department of Internal Medicine, Henan Cancer Hospital & Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Hairui Su
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ngoc-Tung Tran
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jing Song
- Department of Internal Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sydney S. Lu
- Human Oncology & Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Ying Li
- Macau Institute for Applied Research in Medicine and Health, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau. China
| | - Suming Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Omar Abdel-Wahab
- Human Oncology & Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Yanyan Liu
- Department of Internal Medicine, Henan Cancer Hospital & Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinyang Zhao
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
15
|
Fujiwara T, Sasaki K, Saito K, Hatta S, Ichikawa S, Kobayashi M, Okitsu Y, Fukuhara N, Onishi Y, Harigae H. Forced FOG1 expression in erythroleukemia cells: Induction of erythroid genes and repression of myelo-lymphoid transcription factor PU.1. Biochem Biophys Res Commun 2017; 485:380-387. [PMID: 28216155 DOI: 10.1016/j.bbrc.2017.02.068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/13/2017] [Indexed: 11/27/2022]
Abstract
The transcription factor GATA-1-interacting protein Friend of GATA-1 (FOG1) is essential for proper transcriptional activation and repression of GATA-1 target genes; yet, the mechanisms by which FOG1 exerts its activating and repressing functions remain unknown. Forced FOG1 expression in human K562 erythroleukemia cells induced the expression of erythroid genes (SLC4A1, globins) but repressed that of GATA-2 and PU.1. A quantitative chromatin immunoprecipitation (ChIP) analysis demonstrated increased GATA-1 chromatin occupancy at both FOG1-activated as well as FOG1-repressed gene loci. However, while TAL1 chromatin occupancy was significantly increased at FOG1-activated gene loci, it was significantly decreased at FOG1-repressed gene loci. When FOG1 was overexpressed in TAL1-knocked down K562 cells, FOG1-mediated activation of HBA, HBG, and SLC4A1 was significantly compromised by TAL1 knockdown, suggesting that FOG1 may require TAL1 to activate GATA-1 target genes. Promoter analysis and quantitative ChIP analysis demonstrated that FOG1-mediated transcriptional repression of PU.1 would be mediated through a GATA-binding element located at its promoter, accompanied by significantly decreased H3 acetylation at lysine 4 and 9 (K4 and K9) as well as H3K4 trimethylation. Our results provide important mechanistic insight into the role of FOG1 in the regulation of GATA-1-regulated genes and suggest that FOG1 has an important role in inducing cells to differentiate toward the erythroid lineage rather than the myelo-lymphoid one by repressing the expression of PU.1.
Collapse
Affiliation(s)
- Tohru Fujiwara
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan.
| | - Katsuyuki Sasaki
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan; Department of Laboratory, Tohoku University Hospital, Sendai, 1-1 Seiryo-cho, Aoba-ku, Sendai 980-8574, Japan
| | - Kei Saito
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan
| | - Shunsuke Hatta
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan
| | - Satoshi Ichikawa
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan
| | - Masahiro Kobayashi
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan
| | - Yoko Okitsu
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan
| | - Noriko Fukuhara
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan
| | - Yasushi Onishi
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan
| | - Hideo Harigae
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan
| |
Collapse
|
16
|
Inokura K, Fujiwara T, Saito K, Iino T, Hatta S, Okitsu Y, Fukuhara N, Onishi Y, Ishizawa K, Shimoda K, Harigae H. Impact of TET2 deficiency on iron metabolism in erythroblasts. Exp Hematol 2017; 49:56-67.e5. [PMID: 28167288 DOI: 10.1016/j.exphem.2017.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 01/13/2017] [Accepted: 01/21/2017] [Indexed: 01/14/2023]
Abstract
Sideroblastic anemia is characterized by the presence of ring sideroblasts (RSs), which are caused by iron accumulation in the mitochondria of erythroblasts and are present in both the acquired and congenital forms of the disease. However, the mechanism leading to RS formation remains elusive. Acquired sideroblastic anemia is usually observed in myelodysplastic syndrome (MDS). Because a subset of MDS harbors a somatic mutation of TET2, it may be involved in iron metabolism and/or heme biosynthesis in erythroblasts. Tet2 knockdown (Tet2trap) induced exhibited mild normocytic anemia and elevated serum ferritin levels in 4-month-old mice. Although typical RSs were not observed, increased mitochondrial ferritin (FTMT) amounts were observed in the erythroblasts of Tet2-knockdown mice. Quantitative real-time polymerase chain reaction demonstrated significant dysregulation of genes involved in iron and heme metabolism, including Hmox1, Fech, Abcb7, and Sf3b1 downregulation. After the identification of a cytosine-guanine island in the promoters of Fech, Abcb7, and Sf3b1, we evaluated DNA methylation status and found significantly higher methylation levels at the CpG sites in the erythroblasts of Tet2-knockdown mice. Furthermore, Tet2 knockdown in erythroblasts resulted in decreased heme concentration and accumulation of FTMT. Therefore, TET2 plays a role in the iron and heme metabolism in erythroblasts.
Collapse
Affiliation(s)
- Kyoko Inokura
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Tohru Fujiwara
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan; Department of Molecular Hematology/Oncology, Tohoku University Graduate School, Sendai, Japan
| | - Kei Saito
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Tatsuya Iino
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Shunsuke Hatta
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Yoko Okitsu
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Noriko Fukuhara
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Yasushi Onishi
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Kenichi Ishizawa
- Department of Hematology and Cell Therapy, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Kazuya Shimoda
- Division of Gastroenterology and Hematology, Department of Internal Medicine, Faculty of Medicine, Miyazaki University, Miyazaki, Japan
| | - Hideo Harigae
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan; Department of Molecular Hematology/Oncology, Tohoku University Graduate School, Sendai, Japan.
| |
Collapse
|
17
|
Sakurai K, Fujiwara T, Hasegawa S, Okitsu Y, Fukuhara N, Onishi Y, Yamada-Fujiwara M, Ichinohasama R, Harigae H. Inhibition of human primary megakaryocyte differentiation by anagrelide: a gene expression profiling analysis. Int J Hematol 2016; 104:190-9. [DOI: 10.1007/s12185-016-2006-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 04/05/2016] [Accepted: 04/05/2016] [Indexed: 11/29/2022]
|
18
|
Identification of a novel putative mitochondrial protein FAM210B associated with erythroid differentiation. Int J Hematol 2016; 103:387-95. [PMID: 26968549 DOI: 10.1007/s12185-016-1968-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/01/2016] [Accepted: 03/01/2016] [Indexed: 10/22/2022]
Abstract
The transcription factor GATA-1 plays an essential role in erythroid differentiation. To identify novel GATA-1 target genes, we analyzed a merged ChIP-seq and expression profiling dataset. We identified FAM210B as a putative novel GATA-1 target gene. Study results demonstrated that GATA-1 directly regulates FAM210B expression, presumably by binding to an intronic enhancer region. Both human and murine FAM210B are abundantly expressed in the later stages of erythroblast development. Moreover, the deduced amino acid sequence predicted that FAM210B is a membrane protein, and Western blot analysis demonstrated its mitochondrial localization. Loss-of-function analysis in erythroid cells suggested that FAM210B may be involved in erythroid differentiation. The identification and characterization of FAM210B provides new insights in the study of erythropoiesis and hereditary anemias.
Collapse
|
19
|
Kim M, Tan YS, Cheng WC, Kingsbury TJ, Heimfeld S, Civin CI. MIR144 and MIR451 regulate human erythropoiesis via RAB14. Br J Haematol 2014; 168:583-97. [PMID: 25312678 DOI: 10.1111/bjh.13164] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 08/15/2014] [Indexed: 12/11/2022]
Abstract
Expression levels of MIR144 and MIR451 increase during erythropoiesis, a pattern that is conserved from zebrafish to humans. As these two miRs are expressed from the same polycistronic transcript, we manipulated MIR144 and MIR451 in human erythroid cells individually and together to investigate their effects on human erythropoiesis. Inhibition of endogenous human MIR451 resulted in decreased numbers of erythroid (CD71(hi) CD235a(hi) CD34(-) ) cells, consistent with prior studies in zebrafish and mice. In addition, inhibition of MIR144 impaired human erythroid differentiation, unlike in zebrafish and mouse studies where the functional effect of MIR144 on erythropoiesis was minimal. In this study, we found RAB14 is a direct target of both MIR144 and MIR451. As MIR144 and MIR451 expression increased during human erythropoiesis, RAB14 protein expression decreased. Enforced RAB14 expression phenocopied the effect of MIR144 and/or MIR451 depletion, whereas shRNA-mediated RAB14 knockdown protected cells from MIR144 and/or MIR451 depletion-mediated erythropoietic inhibition. RAB14 knockdown increased the frequency and number of erythroid cells, increased β-haemoglobin expression, and decreased CBFA2T3 expression during human erythropoiesis. In summary, we utilized MIR144 and MIR451 to identify RAB14 as a novel physiological inhibitor of human erythropoiesis.
Collapse
Affiliation(s)
- MinJung Kim
- Departments of Physiology and Pediatrics, Center for Stem Cell Biology & Regenerative Medicine, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | |
Collapse
|
20
|
Fujiwara T, Saitoh H, Inoue A, Kobayashi M, Okitsu Y, Katsuoka Y, Fukuhara N, Onishi Y, Ishizawa K, Ichinohasama R, Harigae H. 3-Deazaneplanocin A (DZNep), an inhibitor of S-adenosylmethionine-dependent methyltransferase, promotes erythroid differentiation. J Biol Chem 2014; 289:8121-34. [PMID: 24492606 PMCID: PMC3961643 DOI: 10.1074/jbc.m114.548651] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
EZH2, a core component of polycomb repressive complex 2 (PRC2), plays a role in transcriptional repression through histone H3 Lys-27 trimethylation and is involved in various biological processes, including hematopoiesis. It is well known that 3-deazaneplanocin A (DZNep), an inhibitor of S-adenosylmethionine-dependent methyltransferase that targets the degradation of EZH2, preferentially induces apoptosis in various hematological malignancies, suggesting that EZH2 may be a new target for epigenetic treatment. Because PRC2 participates in epigenetic silencing of a subset of GATA-1 target genes during erythroid differentiation, inhibition of EZH2 may influence erythropoiesis. To explore this possibility, we evaluated the impact of DZNep on erythropoiesis. DZNep treatment significantly induced erythroid differentiation of K562 cells, as assessed by benzidine staining and quantitative RT-PCR analysis for representative erythroid-related genes, including globins. When we evaluated the effects of DZNep in human primary erythroblasts derived from cord blood CD34-positive cells, the treatment significantly induced erythroid-related genes, as observed in K562 cells, suggesting that DZNep induces erythroid differentiation. Unexpectedly, siRNA-mediated EZH2 knockdown had no significant effect on the expression of erythroid-related genes. Transcriptional profiling of DZNep-treated K562 cells revealed marked up-regulation of SLC4A1 and EPB42, previously reported as representative targets of the transcriptional corepressor ETO2. In addition, DZNep treatment reduced the protein level of ETO2. These data suggest that erythroid differentiation by DZNep may not be directly related to EZH2 inhibition but may be partly associated with reduced protein level of hematopoietic corepressor ETO2. These data provide a better understanding of the mechanism of action of DZNep, which may be exploited for therapeutic applications for hematological diseases, including anemia.
Collapse
|
21
|
Expression profiling of ETO2-regulated miRNAs in erythroid cells: Possible influence on miRNA abundance. FEBS Open Bio 2013; 3:428-32. [PMID: 24251106 PMCID: PMC3821025 DOI: 10.1016/j.fob.2013.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 10/05/2013] [Accepted: 10/07/2013] [Indexed: 12/26/2022] Open
Abstract
ETO2 is a component of a protein complex containing master regulators of hematopoiesis, including GATA-1 and SCL/TAL1, and also has RNA binding properties. Although ETO2 has been reported to repress GATA-1 target genes through histone deacetylation of the target gene loci in erythroid cells, little is known about the contribution of ETO2 to microRNA (miRNA) regulation. Here, we conducted miRNA profiling in ETO2-overexpressing and ETO2-silenced K562 cells. The analysis suggests that ETO2 positively regulates the abundance of mature miRNAs, including miR-21, miR-29b and let-7e. Our data suggest a novel mode of ETO2-mediated target gene repression via effects on miRNA expression. miRNA profiling was conducted in ETO2-overexpressing and ETO2-silenced K562 cells. ETO2 positively regulates the abundance of miRNA. ETO2 positively regulates the expression of miR-21, miR-29b and let-7e.
Collapse
Key Words
- CBF1, core-binding factor 1
- ETO2
- ETO2 (CBFA2T3), core-binding factor, runt domain, alpha subunit 2, translocated to, 3
- Erythropoiesis
- IL-3, interleukin 3
- IMDM, Iscove’s Modified Dulbecco’s Media
- LMO2, LIM domain only 2
- RPMI, Roswell Park Memorial Institute
- SCF, stem cell factor
- cDNA, complementary DNA
- miRNA
- siRNA, small interfering RNA
Collapse
|
22
|
Fujiwara T, Ikeda T, Nagasaka Y, Okitsu Y, Katsuoka Y, Fukuhara N, Onishi Y, Ishizawa K, Ichinohasama R, Tomosugi N, Harigae H. A low-molecular-weight compound K7174 represses hepcidin: possible therapeutic strategy against anemia of chronic disease. PLoS One 2013; 8:e75568. [PMID: 24086573 PMCID: PMC3785497 DOI: 10.1371/journal.pone.0075568] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 08/14/2013] [Indexed: 12/21/2022] Open
Abstract
Hepcidin is the principal iron regulatory hormone, controlling the systemic absorption and remobilization of iron from intracellular stores. The expression of the hepcidin gene, HAMP, is increased in patients with anemia of chronic disease. Previously, the synthetic compound K7174 was identified through chemical screening as a novel inhibitor of the adhesion of monocytes to cytokine-stimulated endothelial cells. K7174 also ameliorated anemia induced by inflammatory cytokines in mice, which suggests a possible involvement of hepcidin regulation. The present study was performed to assess the impact of K7174 on hepcidin expression in a human hematoma cell line and in mice in vivo. We first demonstrated that K7174 treatment in HepG2 cells significantly decreased HAMP expression. Then, we conducted microarray analysis to determine the molecular mechanism by which K7174 inhibits HAMP expression. Transcriptional profiling confirmed the downregulation of HAMP. Surprisingly, we found that K7174 strongly induced GDF15, known as a negative regulator of HAMP expression. Western blotting analysis as well as ELISA confirmed the induction of GDF15 by K7174 treatment. Furthermore, K7174-mediated HAMP suppression was rescued by the silencing of GDF15 expression. Interestingly, we found that K7174 also upregulates CEBPB. Promoter analysis and chromatin immunoprecipitation analysis revealed that CEBPB could contribute to K7174-mediated transcriptional activation of GDF15. Subsequently, we also examined whether K7174 inhibits hepcidin expression in mice. Quantitative RT-PCR analysis with liver samples from K7174-treated mice demonstrated significant upregulation of Gdf15 and downregulation of Hamp expression, as compared to control mice. Furthermore, serum hepcidin concentration was also significantly decreased in K7174-treated mice. In conclusion, K7174 inhibits hepcidin expression partly by inducing GDF15. K-7174 may be a potential therapeutic option to treat anemia of chronic disease.
Collapse
Affiliation(s)
- Tohru Fujiwara
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
- Molecular Hematology/Oncology, Tohoku University Graduate School, Sendai, Japan
| | - Takashi Ikeda
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Yuki Nagasaka
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Yoko Okitsu
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Yuna Katsuoka
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Noriko Fukuhara
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Yasushi Onishi
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
| | - Kenichi Ishizawa
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
- Clinical Research, Innovation and Education Center, Tohoku University Hospital, Sendai, Japan
| | - Ryo Ichinohasama
- Hematopathology, Tohoku University Graduate School, Sendai, Japan
| | - Naohisa Tomosugi
- Aging Research Unit, Division of Advanced Medicine, Medical Research Institute, Kanazawa Medical College, Kanazawa, Japan
| | - Hideo Harigae
- Department of Hematology and Rheumatology, Tohoku University Graduate School, Sendai, Japan
- Molecular Hematology/Oncology, Tohoku University Graduate School, Sendai, Japan
| |
Collapse
|
23
|
Inoue A, Fujiwara T, Okitsu Y, Katsuoka Y, Fukuhara N, Onishi Y, Ishizawa K, Harigae H. Elucidation of the role of LMO2 in human erythroid cells. Exp Hematol 2013; 41:1062-76.e1. [PMID: 24041784 DOI: 10.1016/j.exphem.2013.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 08/15/2013] [Accepted: 09/03/2013] [Indexed: 01/08/2023]
Abstract
LIM-only protein 2 (LMO2) is a non-DNA-binding component of a protein complex containing master regulators of hematopoiesis, including GATA-1, SCL/TAL1, and LDB1. However, the role of LMO2 in human erythroid differentiation is unclear. LMO2 knockdown in hemin-treated K562 cells reduced the benzidine-positive cell ratio, suggesting that LMO2 retards hemin-mediated K562 cell differentiation. Microarray analysis using K562 cells after siRNA-mediated LMO2 knockdown indicated that 177 and 78 genes were upregulated and downregulated (>1.5-fold), respectively. The downregulated gene ensemble contained prototypical erythroid genes (HBB, SLC4A1). Whereas LMO2 knockdown did not affect GATA-1 or SCL/TAL1 expression, it resulted in significantly reduced chromatin occupancy of GATA-1, SCL/TAL1, and LDB1 at the β-globin locus control region and SLC4A1 locus in both K562 cells and human induced pluripotent stem cell-derived erythroid cells. Introduction of GATA-1 mutations, shown to impair direct interaction with LMO2, significantly diminished chromatin occupancy. On the other hand, knockdown of either SCL/TAL1 or LDB1 also resulted in significantly reduced chromatin occupancy of GATA-1 at endogenous loci, suggesting that impaired assembly of these components also affects GATA-1 chromatin occupancy. In an ex vivo model of erythroid differentiation from CD34(+) cells, LMO2 protein level peaked on day 5 and decreased at later stages of differentiation. The LMO2 expression pattern was similar to those of GATA-1 and SCL/TAL1. Furthermore, shRNA-mediated LMO2 knockdown in primary erythroblasts suggested that LMO2 regulates HBB, HBA, and SLC4A1 expression. LMO2 contributes to GATA-1 target gene expression by affecting assembly of the GATA-SCL/TAL1 complex components at endogenous loci.
Collapse
Affiliation(s)
- Ai Inoue
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|