1
|
Wanet A, Bassal MA, Patel SB, Marchi F, Mariani SA, Ahmed N, Zhang H, Borchiellini M, Chen S, Zhang J, Di Ruscio A, Miyake K, Tsai M, Paranjape A, Park SY, Karasuyama H, Schroeder T, Dzierzak E, Galli SJ, Tenen DG, Welner RS. E-cadherin is regulated by GATA-2 and marks the early commitment of mouse hematopoietic progenitors to the basophil and mast cell fates. Sci Immunol 2021; 6:6/56/eaba0178. [PMID: 33547048 DOI: 10.1126/sciimmunol.aba0178] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 09/09/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022]
Abstract
E-cadherin is a calcium-dependent cell-cell adhesion molecule extensively studied for its involvement in tissue formation, epithelial cell behavior, and suppression of cancer. However, E-cadherin expression in the hematopoietic system has not been fully elucidated. Combining single-cell RNA-sequencing analyses and immunophenotyping, we revealed that progenitors expressing high levels of E-cadherin and contained within the granulocyte-monocyte progenitors (GMPs) fraction have an enriched capacity to differentiate into basophils and mast cells. We detected E-cadherin expression on committed progenitors before the expression of other reported markers of these lineages. We named such progenitors pro-BMPs (pro-basophil and mast cell progenitors). Using RNA sequencing, we observed transcriptional priming of pro-BMPs to the basophil and mast cell lineages. We also showed that GATA-2 directly regulates E-cadherin expression in the basophil and mast cell lineages, thus providing a mechanistic connection between the expression of this cell surface marker and the basophil and mast cell fate specification.
Collapse
Affiliation(s)
- Anaïs Wanet
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Mahmoud A Bassal
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Sweta B Patel
- Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Samanta A Mariani
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Nouraiz Ahmed
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Haoran Zhang
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Marta Borchiellini
- Department of Health Sciences, University of Eastern Piedmont, Novara 28100, Italy.,Department of Translational Medicine, University of Eastern Piedmont, Novara 28100, Italy
| | - Sisi Chen
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Junyan Zhang
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Annalisa Di Ruscio
- Department of Translational Medicine, University of Eastern Piedmont, Novara 28100, Italy.,Harvard Medical School Initiative for RNA Medicine, Boston, MA 02115, USA.,Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Kensuke Miyake
- Inflammation, Infection, Immunity Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Mindy Tsai
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anuya Paranjape
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shin-Young Park
- Transfusion Medicine, Boston Children's Hospital and Harvard Medical School, Harvard Medical School, Boston, MA 02115, USA
| | - Hajime Karasuyama
- Inflammation, Infection, Immunity Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
| | - Elaine Dzierzak
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Microbiology and Immunology and Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daniel G Tenen
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA. .,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Robert S Welner
- Division of Hematology/Oncology, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
2
|
Sainz de Aja J, Menchero S, Rollan I, Barral A, Tiana M, Jawaid W, Cossio I, Alvarez A, Carreño‐Tarragona G, Badia‐Careaga C, Nichols J, Göttgens B, Isern J, Manzanares M. The pluripotency factor NANOG controls primitive hematopoiesis and directly regulates Tal1. EMBO J 2019; 38:embj.201899122. [PMID: 30814124 PMCID: PMC6443201 DOI: 10.15252/embj.201899122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 01/24/2019] [Accepted: 01/25/2019] [Indexed: 02/02/2023] Open
Abstract
Progenitors of the first hematopoietic cells in the mouse arise in the early embryo from Brachyury-positive multipotent cells in the posterior-proximal region of the epiblast, but the mechanisms that specify primitive blood cells are still largely unknown. Pluripotency factors maintain uncommitted cells of the blastocyst and embryonic stem cells in the pluripotent state. However, little is known about the role played by these factors during later development, despite being expressed in the postimplantation epiblast. Using a dual transgene system for controlled expression at postimplantation stages, we found that Nanog blocks primitive hematopoiesis in the gastrulating embryo, resulting in a loss of red blood cells and downregulation of erythropoietic genes. Accordingly, Nanog-deficient embryonic stem cells are prone to erythropoietic differentiation. Moreover, Nanog expression in adults prevents the maturation of erythroid cells. By analysis of previous data for NANOG binding during stem cell differentiation and CRISPR/Cas9 genome editing, we found that Tal1 is a direct NANOG target. Our results show that Nanog regulates primitive hematopoiesis by directly repressing critical erythroid lineage specifiers.
Collapse
Affiliation(s)
- Julio Sainz de Aja
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)MadridSpain
| | - Sergio Menchero
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)MadridSpain
| | - Isabel Rollan
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)MadridSpain
| | - Antonio Barral
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)MadridSpain
| | - Maria Tiana
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)MadridSpain
| | - Wajid Jawaid
- Wellcome‐Medical Research Council Cambridge Stem Cell InstituteCambridgeUK,Department of HaematologyCambridge Institute for Medical ResearchUniversity of CambridgeCambridgeUK
| | - Itziar Cossio
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)MadridSpain
| | - Alba Alvarez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)MadridSpain
| | - Gonzalo Carreño‐Tarragona
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)MadridSpain,Department of HaematologyHospital 12 de OctubreMadridSpain
| | | | - Jennifer Nichols
- Wellcome‐Medical Research Council Cambridge Stem Cell InstituteCambridgeUK,Department of PhysiologyDevelopment and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Berthold Göttgens
- Wellcome‐Medical Research Council Cambridge Stem Cell InstituteCambridgeUK,Department of HaematologyCambridge Institute for Medical ResearchUniversity of CambridgeCambridgeUK
| | - Joan Isern
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)MadridSpain,Department of Experimental & Health SciencesUniversity Pompeu Fabra (UPF)BarcelonaSpain
| | - Miguel Manzanares
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| |
Collapse
|
3
|
Palam LR, Mali RS, Ramdas B, Srivatsan SN, Visconte V, Tiu RV, Vanhaesebroeck B, Roers A, Gerbaulet A, Xu M, Janga SC, Takemoto CM, Paczesny S, Kapur R. Loss of epigenetic regulator TET2 and oncogenic KIT regulate myeloid cell transformation via PI3K pathway. JCI Insight 2018; 3:94679. [PMID: 29467326 DOI: 10.1172/jci.insight.94679] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 01/18/2018] [Indexed: 01/08/2023] Open
Abstract
Mutations in KIT and TET2 are associated with myeloid malignancies. We show that loss of TET2-induced PI3K activation and -increased proliferation is rescued by targeting the p110α/δ subunits of PI3K. RNA-Seq revealed a hyperactive c-Myc signature in Tet2-/- cells, which is normalized by inhibiting PI3K signaling. Loss of TET2 impairs the maturation of myeloid lineage-derived mast cells by dysregulating the expression of Mitf and Cebpa, which is restored by low-dose ascorbic acid and 5-azacytidine. Utilizing a mouse model in which the loss of TET2 precedes the expression of oncogenic Kit, similar to the human disease, results in the development of a non-mast cell lineage neoplasm (AHNMD), which is responsive to PI3K inhibition. Thus, therapeutic approaches involving hypomethylating agents, ascorbic acid, and isoform-specific PI3K inhibitors are likely to be useful for treating patients with TET2 and KIT mutations.
Collapse
Affiliation(s)
- Lakshmi Reddy Palam
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Raghuveer Singh Mali
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Baskar Ramdas
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ramon V Tiu
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Axel Roers
- Institute for Immunology, Dresden, Germany
| | | | - Mingjiang Xu
- Sylvester Comprehensive Cancer Center, Department of Biochemistry & Molecular Biology, University of Miami School of Medicine, Miami, Florida, USA
| | - Sarath Chandra Janga
- School of Informatics and Computing, Indiana University & Purdue University, Indianapolis, Indiana, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Clifford M Takemoto
- Department of Pediatrics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sophie Paczesny
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Reuben Kapur
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Biochemistry and Molecular Biology and.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
4
|
SCL/TAL1: a multifaceted regulator from blood development to disease. Blood 2017; 129:2051-2060. [DOI: 10.1182/blood-2016-12-754051] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/17/2017] [Indexed: 12/12/2022] Open
Abstract
Abstract
SCL/TAL1 (stem cell leukemia/T-cell acute lymphoblastic leukemia [T-ALL] 1) is an essential transcription factor in normal and malignant hematopoiesis. It is required for specification of the blood program during development, adult hematopoietic stem cell survival and quiescence, and terminal maturation of select blood lineages. Following ectopic expression, SCL contributes to oncogenesis in T-ALL. Remarkably, SCL’s activities are all mediated through nucleation of a core quaternary protein complex (SCL:E-protein:LMO1/2 [LIM domain only 1 or 2]:LDB1 [LIM domain-binding protein 1]) and dynamic recruitment of conserved combinatorial associations of additional regulators in a lineage- and stage-specific context. The finely tuned control of SCL’s regulatory functions (lineage priming, activation, and repression of gene expression programs) provides insight into fundamental developmental and transcriptional mechanisms, and highlights mechanistic parallels between normal and oncogenic processes. Importantly, recent discoveries are paving the way to the development of innovative therapeutic opportunities in SCL+ T-ALL.
Collapse
|
5
|
Wang LD, Rao TN, Rowe RG, Nguyen PT, Sullivan JL, Pearson DS, Doulatov S, Wu L, Lindsley RC, Zhu H, DeAngelo DJ, Daley GQ, Wagers AJ. The role of Lin28b in myeloid and mast cell differentiation and mast cell malignancy. Leukemia 2015; 29:1320-30. [PMID: 25655194 PMCID: PMC4456252 DOI: 10.1038/leu.2015.19] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 12/23/2014] [Accepted: 12/31/2014] [Indexed: 02/06/2023]
Abstract
Mast cells (MCs) are critical components of the innate immune system and important for host defense, allergy, autoimmunity, tissue regeneration and tumor progression. Dysregulated MC development leads to systemic mastocytosis (SM), a clinically variable but often devastating family of hematologic disorders. Here we report that induced expression of Lin28, a heterochronic gene and pluripotency factor implicated in driving a fetal hematopoietic program, caused MC accumulation in adult mice in target organs such as the skin and peritoneal cavity. In vitro assays revealed a skewing of myeloid commitment in LIN28B-expressing hematopoietic progenitors, with increased levels of LIN28B in common myeloid and basophil-MC progenitors altering gene expression patterns to favor cell fate choices that enhanced MC specification. In addition, LIN28B-induced MCs appeared phenotypically and functionally immature, and in vitro assays suggested a slowing of MC terminal differentiation in the context of LIN28B upregulation. Finally, interrogation of human MC leukemia samples revealed upregulation of LIN28B in abnormal MCs from patients with SM. This work identifies Lin28 as a novel regulator of innate immune function and a new protein of interest in MC disease.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Animals
- Blotting, Western
- Bone Marrow Transplantation
- Cell Differentiation
- Cells, Cultured
- DNA-Binding Proteins/physiology
- Female
- Flow Cytometry
- Hematopoiesis/physiology
- Humans
- Leukemia, Mast-Cell/metabolism
- Leukemia, Mast-Cell/pathology
- Leukemia, Mast-Cell/therapy
- Male
- Mast Cells/cytology
- Mast Cells/metabolism
- Mastocytosis, Systemic/metabolism
- Mastocytosis, Systemic/pathology
- Mastocytosis, Systemic/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Middle Aged
- Myeloid Cells/cytology
- Myeloid Cells/metabolism
- RNA, Messenger/genetics
- RNA-Binding Proteins/metabolism
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Leo D. Wang
- Joslin Diabetes Center, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Dana-Farber/Boston Children’s Center for Cancer and Blood Disorders, Boston, MA, USA
- Department of Medicine, Boston Children’s Hospital, Boston, MA, USA
| | - Tata Nageswara Rao
- Joslin Diabetes Center, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - R. Grant Rowe
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Dana-Farber/Boston Children’s Center for Cancer and Blood Disorders, Boston, MA, USA
- Department of Medicine, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease Research, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Phi T. Nguyen
- Joslin Diabetes Center, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Jessica L. Sullivan
- Joslin Diabetes Center, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Daniel S. Pearson
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Dana-Farber/Boston Children’s Center for Cancer and Blood Disorders, Boston, MA, USA
- Department of Medicine, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease Research, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Medical Scientist Training Program, Harvard Medical School, Boston, MA, USA
| | - Sergei Doulatov
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Dana-Farber/Boston Children’s Center for Cancer and Blood Disorders, Boston, MA, USA
- Department of Medicine, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease Research, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Linwei Wu
- Children’s Research Institute, Department of Pediatrics and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Organ Transplant Center, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - R. Coleman Lindsley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Hematology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Hao Zhu
- Children’s Research Institute, Department of Pediatrics and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel J. DeAngelo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - George Q. Daley
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Dana-Farber/Boston Children’s Center for Cancer and Blood Disorders, Boston, MA, USA
- Department of Medicine, Boston Children’s Hospital, Boston, MA, USA
- Manton Center for Orphan Disease Research, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Division of Hematology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Amy J. Wagers
- Joslin Diabetes Center, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| |
Collapse
|
6
|
GATA2 is critical for the maintenance of cellular identity in differentiated mast cells derived from mouse bone marrow. Blood 2015; 125:3306-15. [PMID: 25855601 DOI: 10.1182/blood-2014-11-612465] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/31/2015] [Indexed: 12/13/2022] Open
Abstract
GATA2 plays a crucial role for the mast cell fate decision. We herein demonstrate that GATA2 is also required for the maintenance of the cellular identity in committed mast cells derived from mouse bone marrow (BMMCs). The deletion of the GATA2 DNA binding domain (GATA2ΔCF) in BMMCs resulted in a loss of the mast cell phenotype and an increase in the number of CD11b- and/or Ly6G/C-positive cells. These cells showed the ability to differentiate into macrophage- and neutrophil-like cells but not into eosinophils. Although the mRNA levels of basophil-specific genes were elevated, CD49b, a representative basophil marker, never appeared on these cells. GATA2 ablation led to a significant upregulation of C/EBPα, and forced expression of C/EBPα in wild-type BMMCs phenocopied the GATA2ΔCF cells. Interestingly, simultaneous deletion of the Gata2 and Cebpa genes in BMMCs restored the aberrant increases of CD11b and Ly6G/C while retaining the reduced c-Kit expression. Chromatin immunoprecipitation assays indicated that GATA2 directly binds to the +37-kb region of the Cebpa gene and thereby inhibits the RUNX1 and PU.1 binding to the neighboring region. Upregulation of C/EBPα following the loss of GATA2 was not observed in cultured mast cells derived from peritoneal fluid, whereas the repression of c-Kit and other mast cell-specific genes were observed in these cells. Collectively, these results indicate that GATA2 maintains cellular identity by preventing Cebpa gene activation in a subpopulation of mast cells, whereas it plays a fundamental role as a positive regulator of mast cell-specific genes throughout development of this cell lineage.
Collapse
|
7
|
Hwang S, Lee SJ, Park SH, Chitteti BR, Srour EF, Cooper S, Hangoc G, Broxmeyer HE, Kwon BS. Nonmarrow hematopoiesis occurs in a hyaluronic-acid-rich node and duct system in mice. Stem Cells Dev 2014; 23:2661-71. [PMID: 24914588 DOI: 10.1089/scd.2014.0075] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
A hyaluronic-acid-rich node and duct system (HAR-NDS) was found on the surface of internal organs of mice, and inside their blood and lymph vessels. The nodes (HAR-Ns) were filled with immune cells of the innate system and were especially enriched with mast cells and histiocytes. They also contained hematopoietic progenitor cells (HPCs), such as granulocyte-macrophage, erythroid, multipotential progenitors, and mast cell progenitors (MCPs). MCPs were the most abundant among the HPCs in HAR-Ns. Their frequency was fivefold higher than that of the MCPs in bone marrow. In addition, the system contained pluripotent stem cells (PSCs) capable of producing CD45(-)Flk1(+) hemangioblast-like cells, which subsequently generated various types of HPCs and differentiated blood cells. Although HAR-Ns did not appear to harbor enough number of cells capable of long-term reconstitution or short-term radioprotection of lethally irradiated recipients, bone marrow cells were able to engraft in the HAR-NDS and reconstitute hematopoietic potentials of the system. PSCs and HPCs were consistently found in intravenous, intralymphatic, and intestinal HAR-ND. We infer that PSCs and HPCs reside in the HAR-ND and that this novel system may serve as an alternative means to traffic immature and mature blood cells throughout the body.
Collapse
Affiliation(s)
- Sunhee Hwang
- 1 Cancer Biology and Immunotherapy, National Cancer Center , Goyang, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Calero-Nieto FJ, Ng FS, Wilson NK, Hannah R, Moignard V, Leal-Cervantes AI, Jimenez-Madrid I, Diamanti E, Wernisch L, Göttgens B. Key regulators control distinct transcriptional programmes in blood progenitor and mast cells. EMBO J 2014; 33:1212-26. [PMID: 24760698 PMCID: PMC4168288 DOI: 10.1002/embj.201386825] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 02/27/2014] [Accepted: 03/20/2014] [Indexed: 12/21/2022] Open
Abstract
Despite major advances in the generation of genome-wide binding maps, the mechanisms by which transcription factors (TFs) regulate cell type identity have remained largely obscure. Through comparative analysis of 10 key haematopoietic TFs in both mast cells and blood progenitors, we demonstrate that the largely cell type-specific binding profiles are not opportunistic, but instead contribute to cell type-specific transcriptional control, because (i) mathematical modelling of differential binding of shared TFs can explain differential gene expression, (ii) consensus binding sites are important for cell type-specific binding and (iii) knock-down of blood stem cell regulators in mast cells reveals mast cell-specific genes as direct targets. Finally, we show that the known mast cell regulators Mitf and c-fos likely contribute to the global reorganisation of TF binding profiles. Taken together therefore, our study elucidates how key regulatory TFs contribute to transcriptional programmes in several distinct mammalian cell types.
Collapse
Affiliation(s)
- Fernando J Calero-Nieto
- Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, Cambridge University, Cambridge, UK
| | - Felicia S Ng
- Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, Cambridge University, Cambridge, UK
| | - Nicola K Wilson
- Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, Cambridge University, Cambridge, UK
| | - Rebecca Hannah
- Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, Cambridge University, Cambridge, UK
| | - Victoria Moignard
- Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, Cambridge University, Cambridge, UK
| | - Ana I Leal-Cervantes
- Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, Cambridge University, Cambridge, UK
| | - Isabel Jimenez-Madrid
- Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, Cambridge University, Cambridge, UK
| | - Evangelia Diamanti
- Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, Cambridge University, Cambridge, UK
| | - Lorenz Wernisch
- MRC Biostatistics Unit, Institute of Public Health, Cambridge, UK
| | - Berthold Göttgens
- Department of Haematology, Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, Cambridge University, Cambridge, UK
| |
Collapse
|
9
|
Transcription factor GATA1 is dispensable for mast cell differentiation in adult mice. Mol Cell Biol 2014; 34:1812-26. [PMID: 24615013 DOI: 10.1128/mcb.01524-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Although previous studies have shown that GATA1 is required for mast cell differentiation, the effects of the complete ablation of GATA1 in mast cells have not been examined. Using conditional Gata1 knockout mice (Gata1(-/y)), we demonstrate here that the complete ablation of GATA1 has a minimal effect on the number and distribution of peripheral tissue mast cells in adult mice. The Gata1(-/y) bone marrow cells were capable of differentiating into mast cells ex vivo. Microarray analyses showed that the repression of GATA1 in bone marrow mast cells (BMMCs) has a small impact on the mast cell-specific gene expression in most cases. Interestingly, however, the expression levels of mast cell tryptases in the mouse chromosome 17A3.3 were uniformly reduced in the GATA1 knockdown cells, and GATA1 was found to bind to a 500-bp region at the 5' end of this locus. Revealing a sharp contrast to that observed in the Gata1-null BMMCs, GATA2 deficiency resulted in a significant loss of the c-Kit(+) FcεRIα(+) mast cell fraction and a reduced expression of several mast cell-specific genes. Collectively, GATA2 plays a more important role than GATA1 in the regulation of most mast cell-specific genes, while GATA1 might play specific roles in mast cell functions.
Collapse
|
10
|
Love PE, Warzecha C, Li L. Ldb1 complexes: the new master regulators of erythroid gene transcription. Trends Genet 2013; 30:1-9. [PMID: 24290192 DOI: 10.1016/j.tig.2013.10.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 10/17/2013] [Accepted: 10/18/2013] [Indexed: 10/26/2022]
Abstract
Elucidation of the genetic pathways that control red blood cell development has been a central goal of erythropoiesis research over the past decade. Notably, data from several recent studies have provided new insights into the regulation of erythroid gene transcription. Transcription profiling demonstrates that erythropoiesis is mainly controlled by a small group of lineage-restricted transcription factors [Gata binding protein 1 (Gata1), T cell acute lymphocytic leukemia 1 protein (Tal1), and Erythroid Kruppel-like factor (EKLF; henceforth referred to as Klf1)]. Binding-site mapping using ChIP-Seq indicates that most DNA-bound Gata1 and Tal1 proteins are contained within higher order complexes (Ldb1 complexes) that include the nuclear adapters Ldb1 and Lmo2. Ldb1 complexes regulate Klf1, and Ldb1 complex-binding sites frequently colocalize with Klf1 at erythroid genes and cis-regulatory elements, indicating strong functional synergy between Gata1, Tal1, and Klf1. Together with new data demonstrating that Ldb1 can mediate long-range promoter-enhancer interactions, these findings provide a foundation for the first comprehensive models of the global regulation of erythroid gene transcription.
Collapse
Affiliation(s)
- Paul E Love
- Eunice Kennedy Shriver, National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Claude Warzecha
- Eunice Kennedy Shriver, National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - LiQi Li
- Eunice Kennedy Shriver, National Institute of Child Health & Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
11
|
Solek CM, Oliveri P, Loza-Coll M, Schrankel CS, Ho EC, Wang G, Rast JP. An ancient role for Gata-1/2/3 and Scl transcription factor homologs in the development of immunocytes. Dev Biol 2013; 382:280-92. [DOI: 10.1016/j.ydbio.2013.06.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 06/07/2013] [Accepted: 06/12/2013] [Indexed: 12/30/2022]
|
12
|
Impaired in vitro erythropoiesis following deletion of the Scl (Tal1) +40 enhancer is largely compensated for in vivo despite a significant reduction in expression. Mol Cell Biol 2013; 33:1254-66. [PMID: 23319051 DOI: 10.1128/mcb.01525-12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The Scl (Tal1) gene encodes a helix-loop-helix transcription factor essential for hematopoietic stem cell and erythroid development. The Scl +40 enhancer is situated downstream of Map17, the 3' flanking gene of Scl, and is active in transgenic mice during primitive and definitive erythropoiesis. To analyze the in vivo function of the Scl +40 enhancer within the Scl/Map17 transcriptional domain, we deleted this element in the germ line. Scl(Δ40/Δ40) mice were viable with reduced numbers of erythroid CFU in both bone marrow and spleen yet displayed a normal response to stress hematopoiesis. Analysis of Scl(Δ40/Δ40) embryonic stem (ES) cells revealed impaired erythroid differentiation, which was accompanied by a failure to upregulate Scl when erythropoiesis was initiated. Map17 expression was also reduced in hematopoietic tissues and differentiating ES cells, and the Scl +40 element was able to enhance activity of the Map17 promoter. However, only Scl but not Map17 could rescue the Scl(Δ40/Δ40) ES phenotype. Together, these data demonstrate that the Scl +40 enhancer is an erythroid cell-specific enhancer that regulates the expression of both Scl and Map17. Moreover, deletion of the +40 enhancer causes a novel erythroid phenotype, which can be rescued by ectopic expression of Scl but not Map17.
Collapse
|
13
|
E2A transcription factors limit expression of Gata3 to facilitate T lymphocyte lineage commitment. Blood 2013; 121:1534-42. [PMID: 23297135 DOI: 10.1182/blood-2012-08-449447] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The E2A transcription factors promote the development of thymus-seeding cells, but it remains unknown whether these proteins play a role in T lymphocyte lineage specification or commitment. Here, we showed that E2A proteins were required to promote T-lymphocyte commitment from DN2 thymocytes and to extinguish their potential for alternative fates. E2A proteins functioned in DN2 cells to limit expression of Gata3, which encodes an essential T-lymphocyte transcription factor whose ectopic expression can arrest T-cell differentiation. Genetic, or small interfering RNA-mediated, reduction of Gata3 rescued T-cell differentiation in the absence of E2A and restricted the development of alternative lineages by limiting the expanded self-renewal potential in E2A−/− DN2 cells. Our data support a novel paradigm in lymphocyte lineage commitment in which the E2A proteins are necessary to limit the expression of an essential lineage specification and commitment factor to restrain self-renewal and to prevent an arrest in differentiation.
Collapse
|
14
|
Chlon TM, Crispino JD. Combinatorial regulation of tissue specification by GATA and FOG factors. Development 2012; 139:3905-16. [PMID: 23048181 DOI: 10.1242/dev.080440] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The development of complex organisms requires the formation of diverse cell types from common stem and progenitor cells. GATA family transcriptional regulators and their dedicated co-factors, termed Friend of GATA (FOG) proteins, control cell fate and differentiation in multiple tissue types from Drosophila to man. FOGs can both facilitate and antagonize GATA factor transcriptional regulation depending on the factor, cell, and even the specific gene target. In this review, we highlight recent studies that have elucidated mechanisms by which FOGs regulate GATA factor function and discuss how these factors use these diverse modes of gene regulation to control cell lineage specification throughout metazoans.
Collapse
Affiliation(s)
- Timothy M Chlon
- Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | | |
Collapse
|
15
|
Regulation of GATA factor expression is distinct between erythroid and mast cell lineages. Mol Cell Biol 2012; 32:4742-55. [PMID: 22988301 DOI: 10.1128/mcb.00718-12] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The zinc finger transcription factors GATA1 and GATA2 participate in mast cell development. Although the expression of these factors is regulated in a cell lineage-specific and differentiation stage-specific manner, their regulation during mast cell development has not been clarified. Here, we show that the GATA2 mRNA level was significantly increased while GATA1 was maintained at low levels during the differentiation of mast cells derived from mouse bone marrow (BMMCs). Unlike in erythroid cells, forced expression or small interfering RNA (siRNA)-mediated knockdown of GATA1 rarely affected GATA2 expression, and vice versa, in mast cells, indicating the absence of cross-regulation between Gata1 and Gata2 genes. Chromatin immunoprecipitation assays revealed that both GATA factors bound to most of the conserved GATA sites of Gata1 and Gata2 loci in BMMCs. However, the GATA1 hematopoietic enhancer (G1HE) of the Gata1 gene, which is essential for GATA1 expression in erythroid and megakaryocytic lineages, was bound only weakly by both GATA factors in BMMCs. Furthermore, transgenic-mouse reporter assays revealed that the G1HE is not essential for reporter expression in BMMCs and peritoneal mast cells. Collectively, these results demonstrate that the expression of GATA factors in mast cells is regulated in a manner quite distinct from that in erythroid cells.
Collapse
|
16
|
Spensberger D, Kotsopoulou E, Ferreira R, Broccardo C, Scott LM, Fourouclas N, Ottersbach K, Green AR, Göttgens B. Deletion of the Scl +19 enhancer increases the blood stem cell compartment without affecting the formation of mature blood lineages. Exp Hematol 2012; 40:588-598.e1. [PMID: 22401818 PMCID: PMC3387379 DOI: 10.1016/j.exphem.2012.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 02/11/2012] [Accepted: 02/15/2012] [Indexed: 11/29/2022]
Abstract
The stem cell leukemia (Scl)/Tal1 gene is essential for normal blood and endothelial development, and is expressed in hematopoietic stem cells (HSCs), progenitors, erythroid, megakaryocytic, and mast cells. The Scl +19 enhancer is active in HSCs and progenitor cells, megakaryocytes, and mast cells, but not mature erythroid cells. Here we demonstrate that in vivo deletion of the Scl +19 enhancer (Scl(Δ19/Δ19)) results in viable mice with normal Scl expression in mature hematopoietic lineages. By contrast, Scl expression is reduced in the stem/progenitor compartment and flow cytometry analysis revealed that the HSC and megakaryocyte-erythroid progenitor populations are enlarged in Scl(Δ19/Δ19) mice. The increase in HSC numbers contributed to enhanced expansion in bone marrow transplantation assays, but did not affect multilineage repopulation or stress responses. These results affirm that the Scl +19 enhancer plays a key role in the development of hematopoietic stem/progenitor cells, but is not necessary for mature hematopoietic lineages. Moreover, active histone marks across the Scl locus were significantly reduced in Scl(Δ19/Δ19) fetal liver cells without major changes in steady-state messenger RNA levels, suggesting post-transcriptional compensation for loss of a regulatory element, a result that might be widely relevant given the frequent observation of mild phenotypes after deletion of regulatory elements.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Anthony R. Green
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
17
|
Curtis DJ, Salmon JM, Pimanda JE. Concise Review: Blood Relatives: Formation and regulation of hematopoietic stem cells by the basic helix-loop-helix transcription factors stem cell leukemia and lymphoblastic leukemia-derived sequence 1. Stem Cells 2012; 30:1053-8. [DOI: 10.1002/stem.1093] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
18
|
Ishijima Y, Ohmori S, Uenishi A, Ohneda K. GATA transcription factors are involved in IgE-dependent mast cell degranulation by enhancing the expression of phospholipase C-γ1. Genes Cells 2012; 17:285-301. [DOI: 10.1111/j.1365-2443.2012.01588.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
19
|
The role of the GATA2 transcription factor in normal and malignant hematopoiesis. Crit Rev Oncol Hematol 2011; 82:1-17. [PMID: 21605981 DOI: 10.1016/j.critrevonc.2011.04.007] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 03/18/2011] [Accepted: 04/21/2011] [Indexed: 11/23/2022] Open
Abstract
Hematopoiesis involves an elaborate regulatory network of transcription factors that coordinates the expression of multiple downstream genes, and maintains homeostasis within the hematopoietic system through the accurate orchestration of cellular proliferation, differentiation and survival. As a result, defects in the expression levels or the activity of these transcription factors are intimately linked to hematopoietic disorders, including leukemia. The GATA family of nuclear regulatory proteins serves as a prototype for the action of lineage-restricted transcription factors. GATA1 and GATA2 are expressed principally in hematopoietic lineages, and have essential roles in the development of multiple hematopoietic cells, including erythrocytes and megakaryocytes. Moreover, GATA2 is crucial for the proliferation and maintenance of hematopoietic stem cells and multipotential progenitors. In this review, we summarize the current knowledge regarding the biological properties and functions of the GATA2 transcription factor in normal and malignant hematopoiesis.
Collapse
|
20
|
FOG1 requires NuRD to promote hematopoiesis and maintain lineage fidelity within the megakaryocytic-erythroid compartment. Blood 2010; 115:2156-66. [PMID: 20065294 DOI: 10.1182/blood-2009-10-251280] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nuclear factors regulate the development of complex tissues by promoting the formation of one cell lineage over another. The cofactor FOG1 interacts with transcription factors GATA1 and GATA2 to control erythroid and megakaryocyte (MK) differentiation. In contrast, FOG1 antagonizes the ability of GATA factors to promote mast cell (MC) development. Normal FOG1 function in late-stage erythroid cells and MK requires interaction with the chromatin remodeling complex NuRD. Here, we report that mice in which the FOG1/NuRD interaction is disrupted (Fog(ki/ki)) produce MK-erythroid progenitors that give rise to significantly fewer and less mature MK and erythroid colonies in vitro while retaining multilineage capacity, capable of generating MCs and other myeloid lineage cells. Gene expression profiling of Fog(ki/ki) MK-erythroid progenitors revealed inappropriate expression of several MC-specific genes. Strikingly, aberrant MC gene expression persisted in mature Fog(ki/ki) MK and erythroid progeny. Using a GATA1-dependent committed erythroid cell line, select MC genes were found to be occupied by NuRD, suggesting a direct mechanism of repression. Together, these observations suggest that a simple heritable silencing mechanism is insufficient to permanently repress MC genes. Instead, the continuous presence of GATA1, FOG1, and NuRD is required to maintain lineage fidelity throughout MK-erythroid ontogeny.
Collapse
|
21
|
Souroullas GP, Salmon JM, Sablitzky F, Curtis DJ, Goodell MA. Adult hematopoietic stem and progenitor cells require either Lyl1 or Scl for survival. Cell Stem Cell 2009; 4:180-6. [PMID: 19200805 DOI: 10.1016/j.stem.2009.01.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Revised: 12/15/2008] [Accepted: 01/09/2009] [Indexed: 12/01/2022]
Abstract
Scl and Lyl1 encode two related basic-helix-loop-helix transcription factors implicated in T cell acute lymphoblastic leukemia. Previous studies showed that Scl is essential for embryonic and adult erythropoiesis, while Lyl1 is important for B cell development. Single-knockout mice have not revealed an essential function for Scl or Lyl1 in adult hematopoietic stem cells (HSCs). To determine if maintenance of HSCs in single-knockout mice is due to functional redundancy, we generated Lyl1;Scl-conditional double-knockout mice. Here, we report a striking genetic interaction between the two genes, with a clear dose dependence for the presence of Scl or Lyl1 alleles for HSC function. Bone marrow repopulation assays and analyses demonstrated rapid loss of hematopoietic progenitors due to apoptosis. The function of HSCs could be rescued by a single allele of Lyl1 but not Scl. These results show that expression of at least one of these factors is essential for maintenance of adult HSC function.
Collapse
Affiliation(s)
- George P Souroullas
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
22
|
SCL and associated proteins distinguish active from repressive GATA transcription factor complexes. Blood 2008; 113:2191-201. [PMID: 19011221 DOI: 10.1182/blood-2008-07-169417] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
GATA-1 controls hematopoietic development by activating and repressing gene transcription, yet the in vivo mechanisms that specify these opposite activities are unknown. By examining the composition of GATA-1-associated protein complexes in a conditional erythroid rescue system as well as through the use of tiling arrays we detected the SCL/TAL1, LMO2, Ldb1, E2A complex at all positively acting GATA-1-bound elements examined. Similarly, the SCL complex is present at all activating GATA elements in megakaryocytes and mast cells. In striking contrast, at sites where GATA-1 functions as a repressor, the SCL complex is depleted. A DNA-binding defective form of SCL maintains association with a subset of active GATA elements indicating that GATA-1 is a key determinant for SCL recruitment. Knockdown of LMO2 selectively impairs activation but not repression by GATA-1. ETO-2, an SCL-associated protein with the potential for transcription repression, is also absent from GATA-1-repressed genes but, unlike SCL, fails to accumulate at GATA-1-activated genes. Together, these studies identify the SCL complex as a critical and consistent determinant of positive GATA-1 activity in multiple GATA-1-regulated hematopoietic cell lineages.
Collapse
|
23
|
Abstract
Dissecting the molecular mechanisms used by developmental regulators is essential to understand tissue specification/differentiation. SCL/TAL-1 is a basic helix-loop-helix transcription factor absolutely critical for hematopoietic stem/progenitor cell specification and lineage maturation. Using in vitro and forced expression experimental systems, we previously suggested that SCL might have DNA-binding-independent functions. Here, to assess the requirements for SCL DNA-binding activity in vivo, we examined hematopoietic development in mice carrying a germline DNA-binding mutation. Remarkably, in contrast to complete absence of hematopoiesis and early lethality in scl-null embryos, specification of hematopoietic cells occurred in homozygous mutant embryos, indicating that direct DNA binding is dispensable for this process. Lethality was forestalled to later in development, although some mice survived to adulthood. Anemia was documented throughout development and in adulthood. Cellular and molecular studies showed requirements for SCL direct DNA binding in red cell maturation and indicated that scl expression is positively autoregulated in terminally differentiating erythroid cells. Thus, different mechanisms of SCL's action predominate depending on the developmental/cellular context: indirect DNA binding activities and/or sequestration of other nuclear regulators are sufficient in specification processes, whereas direct DNA binding functions with transcriptional autoregulation are critically required in terminal maturation processes.
Collapse
|
24
|
Mast cell transcriptional networks. Blood Cells Mol Dis 2008; 41:82-90. [PMID: 18406636 DOI: 10.1016/j.bcmd.2008.02.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 02/06/2008] [Indexed: 11/20/2022]
Abstract
Unregulated activation of mast cells can contribute to the pathogenesis of inflammatory and allergic diseases, including asthma, rheumatoid arthritis, inflammatory bowel disease, and multiple sclerosis. Absence of mast cells in animal models can lead to impairment in the innate immune response to parasites and bacterial infections. Aberrant clonal accumulation and proliferation of mast cells can result in a variety of diseases ranging from benign cutaneous mastocytosis to systemic mastocytosis or mast cell leukemia. Understanding mast cell differentiation provides important insights into mechanisms of lineage selection during hematopoiesis and can provide targets for new drug development to treat mast cell disorders. In this review, we discuss controversies related to development, sites of origin, and the transcriptional program of mast cells.
Collapse
|
25
|
Metcalf D, Majewski I, Mifsud S, Di Rago L, Alexander WS. Clonogenic mast cell progenitors and their excess numbers in chimeric BALB/c mice with inactivated GATA-1. Proc Natl Acad Sci U S A 2007; 104:18642-7. [PMID: 18000035 PMCID: PMC2141830 DOI: 10.1073/pnas.0709625104] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Indexed: 11/18/2022] Open
Abstract
In agar cultures of marrow cells from adult female BALB/c chimeric GATA-1(Plt13/+) mice, a high frequency of unusual dispersed colonies was noted. Analysis showed that these were colonies of mast cells and that mast cell colony-forming cells (progenitors) could be detected in clonal cultures of adult marrow, neonatal marrow, or fetal liver if the combined stimulus of stem cell factor and interleukin-3 was used. Mast cell progenitors were in active cell cycle and showed an extensive capacity for self-generation. Mast cell colonies both from control GATA-1(+/+) mice and GATA-1(Plt13/+) mice could generate growth factor-dependent cloned cell lines that grew for >18 months. Surprisingly, the majority of the excessive numbers of mast cell progenitors in chimeric GATA-1(Plt13/+) mice were transcribing the inactive Plt13 allele of GATA-1, suggesting that GATA-1 normally acts to restrict the emergence of committed mast cell progenitors. In sharp contrast, all eosinophil progenitors in these mice were transcribing the normal GATA-1 allele. No excess tissue mast cells were observed in GATA-1(Plt13/+) mice, suggesting that the excess mast cell progenitors in these mice might be generating mast cells with a defective in vivo proliferative or tissue homing capacity.
Collapse
Affiliation(s)
- Donald Metcalf
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville 3050, Victoria, Australia.
| | | | | | | | | |
Collapse
|