1
|
Bacha R, Pedersen S, Ismail R, Alwisi N, Al-Mansoori L. GATA3: Orchestrating cellular fate through differentiation and proliferation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119893. [PMID: 39725219 DOI: 10.1016/j.bbamcr.2024.119893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/07/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Cell proliferation and differentiation are two fundamental biological processes that occur in biological systems, tightly regulated by various factors such as transcription factors (TFs). Zinc finger proteins are TFs responsible for maintaining the biological balance via coordinating development and functionality within the living cells. GATA binding protein 3 (GATA3), one of the zinc finger proteins, plays an essential role in driving differentiation and proliferation-related processes, thereby contributing to the regulation of the dynamism and productivity of living cells. By elucidating the complex interactions governed by GATA3, this underscores its significance in maintaining cellular homeostasis. Thus, the current review delves into the molecular pathways influenced by GATA3, highlighting its involvement in multiple developmental processes of various tissues and body sites, particularly in the hematopoietic system (T-cell differentiation), neural tissue differentiation, adipose tissue, as well as epithelial cell maturation.
Collapse
Affiliation(s)
- Rim Bacha
- College of Medicine, QU Health, Qatar University, Doha, P.O. Box 2713, Qatar; College of Health Science, QU Health, Qatar University, Doha, P.O. Box 2713, Qatar; Biomedical Research Center, Qatar University, Doha, P.O. Box 2713, Qatar
| | - Shona Pedersen
- College of Medicine, QU Health, Qatar University, Doha, P.O. Box 2713, Qatar
| | - Rana Ismail
- College of Medicine, QU Health, Qatar University, Doha, P.O. Box 2713, Qatar
| | - Nouran Alwisi
- College of Medicine, QU Health, Qatar University, Doha, P.O. Box 2713, Qatar
| | - Layla Al-Mansoori
- Biomedical Research Center, Qatar University, Doha, P.O. Box 2713, Qatar.
| |
Collapse
|
2
|
Young C, Russell JR, Van De Lagemaat LN, Lawson H, Mapperley C, Kranc KR, Christophorou MA. Intrinsic function of the peptidylarginine deiminase PADI4 is dispensable for normal haematopoiesis. Biol Open 2022; 11:bio059143. [PMID: 35603697 PMCID: PMC9212077 DOI: 10.1242/bio.059143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/16/2022] [Indexed: 11/20/2022] Open
Abstract
Peptidylarginine deiminases (PADIs) are strongly associated with the development of autoimmunity, neurodegeneration and cancer but their physiological roles are ill-defined. The nuclear deiminase PADI4 regulates pluripotency in the mammalian pre-implantation embryo but its function in tissue development is unknown. PADI4 is primarily expressed in the bone marrow, as part of a self-renewal-associated gene signature. It has been shown to regulate the proliferation of multipotent haematopoietic progenitors and proposed to impact on the differentiation of haematopoietic stem cells (HSCs), suggesting that it controls haematopoietic development or regeneration. Using conditional in vivo models of steady state and acute Padi4 ablation, we examined the role of PADI4 in the development and function of the haematopoietic system. We found that PADI4 loss does not significantly affect HSC self-renewal or differentiation potential upon injury or serial transplantation, nor does it lead to HSC exhaustion or premature ageing. Thus PADI4 is dispensable for cell-autonomous HSC maintenance, differentiation and haematopoietic regeneration. This work represents the first study of PADI4 in tissue development and indicates that pharmacological PADI4 inhibition may be tolerated without adverse effects.
Collapse
Affiliation(s)
- Christine Young
- MRC Human Genetics Unit, The Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
| | - John R. Russell
- MRC Human Genetics Unit, The Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Louie N. Van De Lagemaat
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M6BQ, United Kingdom
| | - Hannah Lawson
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M6BQ, United Kingdom
| | - Christopher Mapperley
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M6BQ, United Kingdom
| | - Kamil R. Kranc
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Laboratory of Haematopoietic Stem Cell & Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London EC1M6BQ, United Kingdom
| | - Maria A. Christophorou
- MRC Human Genetics Unit, The Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, United Kingdom
- Epiegetics, Babraham Institute, Cambridge CB22 3AT, United Kingdom
| |
Collapse
|
3
|
Kucinski I, Wilson NK, Hannah R, Kinston SJ, Cauchy P, Lenaerts A, Grosschedl R, Göttgens B. Interactions between lineage-associated transcription factors govern haematopoietic progenitor states. EMBO J 2020; 39:e104983. [PMID: 33103827 PMCID: PMC7737608 DOI: 10.15252/embj.2020104983] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/26/2022] Open
Abstract
Recent advances in molecular profiling provide descriptive datasets of complex differentiation landscapes including the haematopoietic system, but the molecular mechanisms defining progenitor states and lineage choice remain ill-defined. Here, we employed a cellular model of murine multipotent haematopoietic progenitors (Hoxb8-FL) to knock out 39 transcription factors (TFs) followed by RNA-Seq analysis, to functionally define a regulatory network of 16,992 regulator/target gene links. Focussed analysis of the subnetworks regulated by the B-lymphoid TF Ebf1 and T-lymphoid TF Gata3 revealed a surprising role in common activation of an early myeloid programme. Moreover, Gata3-mediated repression of Pax5 emerges as a mechanism to prevent precocious B-lymphoid differentiation, while Hox-mediated activation of Meis1 suppresses myeloid differentiation. To aid interpretation of large transcriptomics datasets, we also report a new method that visualises likely transitions that a progenitor will undergo following regulatory network perturbations. Taken together, this study reveals how molecular network wiring helps to establish a multipotent progenitor state, with experimental approaches and analysis tools applicable to dissecting a broad range of both normal and perturbed cellular differentiation landscapes.
Collapse
Affiliation(s)
- Iwo Kucinski
- Wellcome–MRC Cambridge Stem Cell InstituteDepartment of HaematologyJeffrey Cheah Biomedical CentreUniversity of CambridgeCambridgeUK
| | - Nicola K Wilson
- Wellcome–MRC Cambridge Stem Cell InstituteDepartment of HaematologyJeffrey Cheah Biomedical CentreUniversity of CambridgeCambridgeUK
| | - Rebecca Hannah
- Wellcome–MRC Cambridge Stem Cell InstituteDepartment of HaematologyJeffrey Cheah Biomedical CentreUniversity of CambridgeCambridgeUK
| | - Sarah J Kinston
- Wellcome–MRC Cambridge Stem Cell InstituteDepartment of HaematologyJeffrey Cheah Biomedical CentreUniversity of CambridgeCambridgeUK
| | - Pierre Cauchy
- Department of Cellular and Molecular ImmunologyMax Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Aurelie Lenaerts
- Department of Cellular and Molecular ImmunologyMax Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
- International Max Planck Research School for Molecular and Cellular BiologyMax Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Rudolf Grosschedl
- Department of Cellular and Molecular ImmunologyMax Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Berthold Göttgens
- Wellcome–MRC Cambridge Stem Cell InstituteDepartment of HaematologyJeffrey Cheah Biomedical CentreUniversity of CambridgeCambridgeUK
| |
Collapse
|
4
|
Rao TN, Kumar S, Pulikkottil AJ, Oliveri F, Hendriks RW, Beckel F, Fehling HJ. Novel, Non-Gene-Destructive Knock-In Reporter Mice Refute the Concept of Monoallelic Gata3 Expression. THE JOURNAL OF IMMUNOLOGY 2020; 204:2600-2611. [PMID: 32213568 DOI: 10.4049/jimmunol.2000025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 02/26/2020] [Indexed: 02/04/2023]
Abstract
Accurately tuned expression levels of the transcription factor GATA-3 are crucial at several stages of T cell and innate lymphoid cell development and differentiation. Moreover, several lines of evidence suggest that Gata3 expression might provide a reliable molecular marker for the identification of elusive progenitor cell subsets at the earliest stages of T lineage commitment. To be able to faithfully monitor Gata3 expression noninvasively at the single-cell level, we have generated a novel strain of knock-in reporter mice, termed GATIR, by inserting an expression cassette encoding a bright fluorescent marker into the 3'-untranslated region of the endogenous Gata3 locus. Importantly, in contrast to three previously published strains of Gata3 reporter mice, GATIR mice preserve physiological Gata3 expression on the targeted allele. In this study, we show that GATIR mice faithfully reflect endogenous Gata3 expression without disturbing the development of GATA-3-dependent lymphoid cell populations. We further show that GATIR mice provide an ideal tool for noninvasive monitoring of Th2 polarization and straightforward identification of innate lymphoid cell 2 progenitor populations. Finally, as our reporter is non-gene-destructive, GATIR mice can be bred to homozygosity, not feasible with previously published strains of Gata3 reporter mice harboring disrupted alleles. The availability of hetero- and homozygous Gata3 reporter mice with an exceptionally bright fluorescent marker, allowed us to visualize allelic Gata3 expression in individual cells simply by flow cytometry. The unambiguous results obtained provide compelling evidence against previously postulated monoallelic Gata3 expression in early T lineage and hematopoietic stem cell subsets.
Collapse
Affiliation(s)
| | - Suresh Kumar
- Institute of Immunology, University Hospital, D-89081 Ulm, Germany; and
| | | | - Franziska Oliveri
- Institute of Immunology, University Hospital, D-89081 Ulm, Germany; and
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus Medical Center, NL-3000 CA Rotterdam, the Netherlands
| | - Franziska Beckel
- Institute of Immunology, University Hospital, D-89081 Ulm, Germany; and
| | | |
Collapse
|
5
|
Zaidan N, Ottersbach K. The multi-faceted role of Gata3 in developmental haematopoiesis. Open Biol 2018; 8:rsob.180152. [PMID: 30463912 PMCID: PMC6282070 DOI: 10.1098/rsob.180152] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
The transcription factor Gata3 is crucial for the development of several tissues and cell lineages both during development as well as postnatally. This importance is apparent from the early embryonic lethality following germline Gata3 deletion, with embryos displaying a number of phenotypes, and from the fact that Gata3 has been implicated in several cancer types. It often acts at the level of stem and progenitor cells in which it controls the expression of key lineage-determining factors as well as cell cycle genes, thus being one of the main drivers of cell fate choice and tissue morphogenesis. Gata3 is involved at various stages of haematopoiesis both in the adult as well as during development. This review summarizes the various contributions of Gata3 to haematopoiesis with a particular focus on the emergence of the first haematopoietic stem cells in the embryo—a process that appears to be influenced by Gata3 at various levels, thus highlighting the complex nature of Gata3 action.
Collapse
Affiliation(s)
- Nada Zaidan
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK.,King Abdullah International Medical Research Centre, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Katrin Ottersbach
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
6
|
The transcription factor Zfp90 regulates the self-renewal and differentiation of hematopoietic stem cells. Cell Death Dis 2018; 9:677. [PMID: 29880802 PMCID: PMC5992204 DOI: 10.1038/s41419-018-0721-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 05/09/2018] [Accepted: 05/16/2018] [Indexed: 12/21/2022]
Abstract
Hematopoietic stem cells (HSCs) can give rise to all blood cells that are essential to defend against pathogen invasion. The defective capability of HSC self-renewal is linked to many serious diseases, such as anemia. However, the potential mechanism regulating HSC self-renewal has not been thoroughly elucidated to date. In this study, we showed that Zfp90 was highly expressed in HSCs. Zfp90 deficiency in the hematopoietic system caused impaired HSPC pools and led to HSC dysfunction. We showed that Zfp90 deletion inhibited HSC proliferation, while HSC apoptosis was not affected. Regarding the mechanism of this effect on HSC proliferation, we found that Zfp90 interacted with Snf2l, a subunit of the NURF complex, to regulate Hoxa9 expression. Ectopic expression of Hoxa9 rescued the HSC repopulation capacity in Zfp90-deficient mice, which indicates that Hoxa9 is the downstream effector of Zfp90. In summary, our findings identify Zfp90 as a key transcription factor in determining the fate of HSCs.
Collapse
|
7
|
Liu S, Chan HL, Bai F, Ma J, Scott A, Robbins DJ, Capobianco AJ, Zhu P, Pei XH. Gata3 restrains B cell proliferation and cooperates with p18INK4c to repress B cell lymphomagenesis. Oncotarget 2018; 7:64007-64020. [PMID: 27588406 PMCID: PMC5325421 DOI: 10.18632/oncotarget.11746] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/24/2016] [Indexed: 12/13/2022] Open
Abstract
GATA3, a lineage specifier, controls lymphoid cell differentiation and its function in T cell commitment and development has been extensively studied. GATA3 promotes T cell specification by repressing B cell potential in pro T cells and decreased GATA3 expression is essential for early B cell commitment. Inherited genetic variation in GATA3 has been associated with lymphoma susceptibility. However, it remains elusive how the loss of function of GATA3 promotes B cell development and induces B cell lymphomas. In this study, we found that haploid loss of Gata3 by heterozygous germline deletion increased B cell populations in the bone marrow (BM) and spleen, and decreased CD4 T cell populations in the thymus, confirming that Gata3 promotes T and suppresses B cell development. We discovered that haploid loss of Gata3 reduced thymocyte proliferation with induction of p18Ink4c (p18), an inhibitor of CDK4 and CDK6, but enhanced B cell proliferation in the BM and spleen independent of p18. Loss of p18 partially restored Gata3 deficient thymocyte proliferation, but further stimulated Gata3 deficient B cell proliferation in the BM and spleen. Furthermore, we discovered that haploid loss of Gata3 in p18 deficient mice led to the development of B cell lymphomas that were capable of rapidly regenerating tumors when transplanted into immunocompromised mice. These results indicate that Gata3 deficiency promotes B cell differentiation and proliferation, and cooperates with p18 loss to induce B cell lymphomas. This study, for the first time, reveals that Gata3 is a tumor suppressor specifically in B cell lymphomagenesis.
Collapse
Affiliation(s)
- Shiqin Liu
- Department of Hematology, Peking University First Hospital, Beijing, 100034, China.,Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Ho Lam Chan
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Feng Bai
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Jinshan Ma
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,Xinjiang Uigur Autonomous Region People's Hospital, Xinjiang, 830001, China
| | - Alexandria Scott
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - David J Robbins
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,Sylvester Cancer Center, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Anthony J Capobianco
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,Sylvester Cancer Center, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Ping Zhu
- Department of Hematology, Peking University First Hospital, Beijing, 100034, China
| | - Xin-Hai Pei
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, FL 33136, Miami.,Sylvester Cancer Center, Miller School of Medicine, University of Miami, FL 33136, Miami
| |
Collapse
|
8
|
Goloviznina NA, Verghese SC, Yoon YM, Taratula O, Marks DL, Kurre P. Mesenchymal Stromal Cell-derived Extracellular Vesicles Promote Myeloid-biased Multipotent Hematopoietic Progenitor Expansion via Toll-Like Receptor Engagement. J Biol Chem 2016; 291:24607-24617. [PMID: 27758863 DOI: 10.1074/jbc.m116.745653] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/06/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) present in the bone marrow microenvironment secrete cytokines and angiogenic factors that support the maintenance and regenerative expansion of hematopoietic stem and progenitor cells (HSPCs). Here, we tested the hypothesis that extracellular vesicles (EVs) released by MSCs contribute to the paracrine crosstalk that shapes hematopoietic function. We systematically characterized EV release by murine stromal cells and demonstrate that MSC-derived EVs prompt a loss of HSPC quiescence with concomitant expansion of murine myeloid progenitors. Our studies reveal that HSPC expansion by MSC EVs is mediated via the MyD88 adapter protein and is partially blocked by treatment with a TLR4 inhibitor. Imaging of fluorescence protein-tagged MSC EVs corroborated their cellular co-localization with TLR4 and endosomal Rab5 compartments in HSPCs. The dissection of downstream responses to TLR4 activation reveals that the mechanism by which MSC EVs impact HSPCs involves canonical NF-κB signaling and downstream activation of Hif-1α and CCL2 target genes. Our aggregate data identify a previously unknown role for MSC-derived EVs in the regulation of hematopoiesis through innate immune mechanisms and illustrate the expansive cell-cell crosstalk in the bone marrow microenvironment.
Collapse
Affiliation(s)
| | | | - Young Me Yoon
- From the Department of Pediatrics,; Papé Family Pediatric Research Institute, and
| | - Oleh Taratula
- the Oregon State University, College of Pharmacy, Corvallis, Oregon 97331
| | - Daniel L Marks
- From the Department of Pediatrics,; Papé Family Pediatric Research Institute, and
| | - Peter Kurre
- From the Department of Pediatrics,; Papé Family Pediatric Research Institute, and; Pediatric Cancer Biology Program, Oregon Health & Science University, Portland, Oregon 97239 and.
| |
Collapse
|
9
|
TNF-alpha and Notch signaling regulates the expression of HOXB4 and GATA3 during early T lymphopoiesis. In Vitro Cell Dev Biol Anim 2016; 52:920-934. [PMID: 27251160 DOI: 10.1007/s11626-016-0055-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/04/2016] [Indexed: 10/21/2022]
Abstract
During the early thymus colonization, Notch signaling activation on hematopoietic progenitor cells (HPCs) drives proliferation and T cell commitment. Although these processes are driven by transcription factors such as HOXB4 and GATA3, there is no evidence that Notch directly regulates their transcription. To evaluate the role of NOTCH and TNF signaling in this process, human CD34+ HPCs were cocultured with OP9-DL1 cells, in the presence or absence of TNF. The use of a Notch signaling inhibitor and a protein synthesis inhibitor allowed us to distinguish primary effects, mediated by direct signaling downstream Notch and TNF, from secondary effects, mediated by de novo synthesized proteins. A low and physiologically relevant concentration of TNF promoted T lymphopoiesis in OP9-DL1 cocultures. TNF positively modulated the expression of both transcripts in a Notch-dependent manner; however, GATA3 induction was mediated by a direct mechanism, while HOXB4 induction was indirect. Induction of both transcripts was repressed by a GSK3β inhibitor, indicating that activation of canonical Wnt signaling inhibits rather than induces their expression. Our study provides novel evidences of the mechanisms integrating Notch and TNF-alpha signaling in the transcriptional induction of GATA3 and HOXB4. This mechanism has direct implications in the control of self-renewal, proliferation, commitment, and T cell differentiation.
Collapse
|
10
|
Functional and molecular characterization of mouse Gata2-independent hematopoietic progenitors. Blood 2016; 127:1426-37. [PMID: 26834239 DOI: 10.1182/blood-2015-10-673749] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/20/2016] [Indexed: 01/15/2023] Open
Abstract
The Gata2 transcription factor is a pivotal regulator of hematopoietic cell development and maintenance, highlighted by the fact that Gata2 haploinsufficiency has been identified as the cause of some familial cases of acute myelogenous leukemia/myelodysplastic syndrome and in MonoMac syndrome. Genetic deletion in mice has shown that Gata2 is pivotal to the embryonic generation of hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). It functions in the embryo during endothelial cell to hematopoietic cell transition to affect hematopoietic cluster, HPC, and HSC formation. Gata2 conditional deletion and overexpression studies show the importance of Gata2 levels in hematopoiesis, during all developmental stages. Although previous studies of cell populations phenotypically enriched in HPCs and HSCs show expression of Gata2, there has been no direct study of Gata2 expressing cells during normal hematopoiesis. In this study, we generate a Gata2Venus reporter mouse model with unperturbed Gata2 expression to examine the hematopoietic function and transcriptome of Gata2 expressing and nonexpressing cells. We show that all the HSCs are Gata2 expressing. However, not all HPCs in the aorta, vitelline and umbilical arteries, and fetal liver require or express Gata2. These Gata2-independent HPCs exhibit a different functional output and genetic program, including Ras and cyclic AMP response element-binding protein pathways and other Gata factors, compared with Gata2-dependent HPCs. Our results, indicating that Gata2 is of major importance in programming toward HSC fate but not in all cells with HPC fate, have implications for current reprogramming strategies.
Collapse
|
11
|
Wong WM, Dolinska M, Sigvardsson M, Ekblom M, Qian H. A novel Lin-CD34+CD38- integrin α2- bipotential megakaryocyte-erythrocyte progenitor population in the human bone marrow. Leukemia 2015; 30:1399-402. [PMID: 26500141 PMCID: PMC4895173 DOI: 10.1038/leu.2015.300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- W M Wong
- Hematopoietic Stem Cell Laboratory, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - M Dolinska
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - M Sigvardsson
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - M Ekblom
- Hematopoietic Stem Cell Laboratory, Department of Laboratory Medicine, Lund University, Lund, Sweden.,Department of Hematology and Coagulation, Skåne University Hospital, Lund, Sweden
| | - H Qian
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
12
|
Tindemans I, Serafini N, Di Santo JP, Hendriks RW. GATA-3 function in innate and adaptive immunity. Immunity 2014; 41:191-206. [PMID: 25148023 DOI: 10.1016/j.immuni.2014.06.006] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/19/2014] [Indexed: 02/07/2023]
Abstract
The zinc-finger transcription factor GATA-3 has received much attention as a master regulator of T helper 2 (Th2) cell differentiation, during which it controls interleukin-4 (IL-4), IL-5, and IL-13 expression. More recently, GATA-3 was shown to contribute to type 2 immunity through regulation of group 2 innate lymphoid cell (ILC2) development and function. Furthermore, during thymopoiesis, GATA-3 represses B cell potential in early T cell precursors, activates TCR signaling in pre-T cells, and promotes the CD4(+) T cell lineage after positive selection. GATA-3 also functions outside the thymus in hematopoietic stem cells, regulatory T cells, CD8(+) T cells, thymic natural killer cells, and ILC precursors. Here we discuss the varied functions of GATA-3 in innate and adaptive immune cells, with emphasis on its activity in T cells and ILCs, and examine the mechanistic basis for the dose-dependent, developmental-stage- and cell-lineage-specific activity of this transcription factor.
Collapse
Affiliation(s)
- Irma Tindemans
- Department of Pulmonary Medicine, Erasmus MC, 3000 CA Rotterdam, the Netherlands
| | - Nicolas Serafini
- Innate Immunity Unit, Institut Pasteur, 75724 Paris, France; INSERM U668, 75724 Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, 75724 Paris, France; INSERM U668, 75724 Paris, France
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, 3000 CA Rotterdam, the Netherlands.
| |
Collapse
|
13
|
Scripture-Adams DD, Damle SS, Li L, Elihu KJ, Qin S, Arias AM, Butler RR, Champhekar A, Zhang JA, Rothenberg EV. GATA-3 dose-dependent checkpoints in early T cell commitment. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 193:3470-91. [PMID: 25172496 PMCID: PMC4170028 DOI: 10.4049/jimmunol.1301663] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GATA-3 expression is crucial for T cell development and peaks during commitment to the T cell lineage, midway through the CD4(-)CD8(-) (double-negative [DN]) stages 1-3. We used RNA interference and conditional deletion to reduce GATA-3 protein acutely at specific points during T cell differentiation in vitro. Even moderate GATA-3 reduction killed DN1 cells, delayed progression to the DN2 stage, skewed DN2 gene regulation, and blocked appearance of the DN3 phenotype. Although a Bcl-2 transgene rescued DN1 survival and improved DN2 cell generation, it did not restore DN3 differentiation. Gene expression analyses (quantitative PCR, RNA sequencing) showed that GATA-3-deficient DN2 cells quickly upregulated genes, including Spi1 (PU.1) and Bcl11a, and downregulated genes, including Cpa3, Ets1, Zfpm1, Bcl11b, Il9r, and Il17rb with gene-specific kinetics and dose dependencies. These targets could mediate two distinct roles played by GATA-3 in lineage commitment, as revealed by removing wild-type or GATA-3-deficient early T lineage cells from environmental Notch signals. GATA-3 worked as a potent repressor of B cell potential even at low expression levels, so that only full deletion of GATA-3 enabled pro-T cells to reveal B cell potential. The ability of GATA-3 to block B cell development did not require T lineage commitment factor Bcl11b. In prethymic multipotent precursors, however, titration of GATA-3 activity using tamoxifen-inducible GATA-3 showed that GATA-3 inhibits B and myeloid developmental alternatives at different threshold doses. Furthermore, differential impacts of a GATA-3 obligate repressor construct imply that B and myeloid development are inhibited through distinct transcriptional mechanisms. Thus, the pattern of GATA-3 expression sequentially produces B lineage exclusion, T lineage progression, and myeloid-lineage exclusion for commitment.
Collapse
Affiliation(s)
- Deirdre D Scripture-Adams
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Sagar S Damle
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Long Li
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Koorosh J Elihu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Shuyang Qin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Alexandra M Arias
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Robert R Butler
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Ameya Champhekar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Jingli A Zhang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| | - Ellen V Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125
| |
Collapse
|
14
|
Mirshekar-Syahkal B, Fitch SR, Ottersbach K. Concise Review: From Greenhouse to Garden: The Changing Soil of the Hematopoietic Stem Cell Microenvironment During Development. Stem Cells 2014; 32:1691-700. [DOI: 10.1002/stem.1680] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 01/29/2014] [Accepted: 02/18/2014] [Indexed: 01/27/2023]
Affiliation(s)
- Bahar Mirshekar-Syahkal
- Department of Haematology; Cambridge Institute for Medical Research; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute; University of Cambridge; Cambridge England, United Kingdom
| | - Simon R. Fitch
- Department of Haematology; Cambridge Institute for Medical Research; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute; University of Cambridge; Cambridge England, United Kingdom
| | - Katrin Ottersbach
- Department of Haematology; Cambridge Institute for Medical Research; Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute; University of Cambridge; Cambridge England, United Kingdom
| |
Collapse
|
15
|
Yoshida T, Georgopoulos K. GATA-3 controls self-renewal in stressed HSCs. Nat Immunol 2013; 14:1032-3. [PMID: 24048133 DOI: 10.1038/ni.2715] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Toshimi Yoshida
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | | |
Collapse
|
16
|
Guitart AV, Subramani C, Armesilla-Diaz A, Smith G, Sepulveda C, Gezer D, Vukovic M, Dunn K, Pollard P, Holyoake TL, Enver T, Ratcliffe PJ, Kranc KR. Hif-2α is not essential for cell-autonomous hematopoietic stem cell maintenance. Blood 2013; 122:1741-5. [PMID: 23894152 DOI: 10.1182/blood-2013-02-484923] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Local hypoxia in hematopoietic stem cell (HSC) niches is thought to regulate HSC functions. Hypoxia-inducible factor-1 (Hif-1) and Hif-2 are key mediators of cellular responses to hypoxia. Although oxygen-regulated α-subunits of Hifs, namely Hif-1α and Hif-2α, are closely related, they play overlapping and also distinct functions in nonhematopoietic tissues. Although Hif-1α-deficient HSCs lose their activity on serial transplantation, the role for Hif-2α in cell-autonomous HSC maintenance remains unknown. Here, we demonstrate that constitutive or inducible hematopoiesis-specific Hif-2α deletion does not affect HSC numbers and steady-state hematopoiesis. Furthermore, using serial transplantations and 5-fluorouracil treatment, we demonstrate that HSCs do not require Hif-2α to self-renew and recover after hematopoietic injury. Finally, we show that Hif-1α deletion has no major impact on steady-state maintenance of Hif-2α-deficient HSCs and their ability to repopulate primary recipients, indicating that Hif-1α expression does not account for normal behavior of Hif-2α-deficient HSCs.
Collapse
Affiliation(s)
- Amelie V Guitart
- Medical Research Council Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Frelin C, Herrington R, Janmohamed S, Barbara M, Tran G, Paige CJ, Benveniste P, Zuñiga-Pflücker JC, Souabni A, Busslinger M, Iscove NN. GATA-3 regulates the self-renewal of long-term hematopoietic stem cells. Nat Immunol 2013; 14:1037-44. [PMID: 23974957 PMCID: PMC4972578 DOI: 10.1038/ni.2692] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 07/22/2013] [Indexed: 01/07/2023]
Abstract
The transcription factor GATA-3 is expressed and required for differentiation and function throughout the T lymphocyte lineage. Despite evidence it may also be expressed in multipotent hematopoietic stem cells (HSCs), any role for GATA-3 in these cells has remained unclear. Here we found GATA-3 was in the cytoplasm in quiescent long-term stem cells from steady-state bone marrow but relocated to the nucleus when HSCs cycled. Relocation depended on signaling via the mitogen-activated protein kinase p38 and was associated with a diminished capacity for long-term reconstitution after transfer into irradiated mice. Deletion of Gata3 enhanced the repopulating capacity and augmented the self-renewal of long-term HSCs in cell-autonomous fashion without affecting the cell cycle. Our observations position GATA-3 as a regulator of the balance between self-renewal and differentiation in HSCs that acts downstream of the p38 signaling pathway.
Collapse
Affiliation(s)
- Catherine Frelin
- 1] Ontario Cancer Institute, University Health Network, Toronto, Canada. [2] Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Magnusson M, Sierra MI, Sasidharan R, Prashad SL, Romero M, Saarikoski P, Van Handel B, Huang A, Li X, Mikkola HKA. Expansion on stromal cells preserves the undifferentiated state of human hematopoietic stem cells despite compromised reconstitution ability. PLoS One 2013; 8:e53912. [PMID: 23342037 PMCID: PMC3547050 DOI: 10.1371/journal.pone.0053912] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 12/04/2012] [Indexed: 12/15/2022] Open
Abstract
Lack of HLA-matched hematopoietic stem cells (HSC) limits the number of patients with life-threatening blood disorders that can be treated by HSC transplantation. So far, insufficient understanding of the regulatory mechanisms governing human HSC has precluded the development of effective protocols for culturing HSC for therapeutic use and molecular studies. We defined a culture system using OP9M2 mesenchymal stem cell (MSC) stroma that protects human hematopoietic stem/progenitor cells (HSPC) from differentiation and apoptosis. In addition, it facilitates a dramatic expansion of multipotent progenitors that retain the immunophenotype (CD34+CD38-CD90+) characteristic of human HSPC and proliferative potential over several weeks in culture. In contrast, transplantable HSC could be maintained, but not significantly expanded, during 2-week culture. Temporal analysis of the transcriptome of the ex vivo expanded CD34+CD38-CD90+ cells documented remarkable stability of most transcriptional regulators known to govern the undifferentiated HSC state. Nevertheless, it revealed dynamic fluctuations in transcriptional programs that associate with HSC behavior and may compromise HSC function, such as dysregulation of PBX1 regulated genetic networks. This culture system serves now as a platform for modeling human multilineage hematopoietic stem/progenitor cell hierarchy and studying the complex regulation of HSC identity and function required for successful ex vivo expansion of transplantable HSC.
Collapse
Affiliation(s)
- Mattias Magnusson
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Maria I. Sierra
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Rajkumar Sasidharan
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Sacha L. Prashad
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Melissa Romero
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Pamela Saarikoski
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Ben Van Handel
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Andy Huang
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Xinmin Li
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Hanna K. A. Mikkola
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, United States of America
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
19
|
GATA-3 promotes T-cell specification by repressing B-cell potential in pro-T cells in mice. Blood 2013; 121:1749-59. [PMID: 23287858 DOI: 10.1182/blood-2012-06-440065] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transcription factors orchestrate T-lineage differentiation in the thymus. One critical checkpoint involves Notch1 signaling that instructs T-cell commitment at the expense of the B-lineage program. While GATA-3 is required for T-cell specification, its mechanism of action is poorly understood. We show that GATA-3 works in concert with Notch1 to commit thymic progenitors to the T-cell lineage via 2 distinct pathways. First, GATA-3 orchestrates a transcriptional “repertoire” that is required for thymocyte maturation up to and beyond the pro-T-cell stage. Second, GATA-3 critically suppresses a latent B-cell potential in pro–T cells. As such, GATA-3 is essential to sealing in Notch-induced T-cell fate in early thymocyte precursors by promoting T-cell identity through the repression of alternative developmental options.
Collapse
|
20
|
Tang P, Gao C, Li A, Aster J, Sun L, Chai L. Differential roles of Kras and Pten in murine leukemogenesis. Leukemia 2012. [PMID: 23183424 DOI: 10.1038/leu.2012.316] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
21
|
Signaling from the sympathetic nervous system regulates hematopoietic stem cell emergence during embryogenesis. Cell Stem Cell 2012; 11:554-66. [PMID: 23040481 PMCID: PMC3510442 DOI: 10.1016/j.stem.2012.07.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 04/25/2012] [Accepted: 07/02/2012] [Indexed: 02/02/2023]
Abstract
The first adult-repopulating hematopoietic stem cells (HSCs) emerge in the aorta-gonads-mesonephros (AGM) region of the embryo. We have recently identified the transcription factor Gata3 as being upregulated in this tissue specifically at the time of HSC emergence. We now demonstrate that the production of functional and phenotypic HSCs in the AGM is impaired in the absence of Gata3. Furthermore, we show that this effect on HSC generation is secondary to the role of Gata3 in the production of catecholamines, the mediators of the sympathetic nervous system (SNS), thus making these molecules key components of the AGM HSC niche. These findings demonstrate that the recently described functional interplay between the hematopoietic system and the SNS extends to the earliest stages of their codevelopment and highlight the fact that HSC development needs to be viewed in the context of the development of other organs.
Collapse
|
22
|
Pérez-Montarelo D, Hudson NJ, Fernández AI, Ramayo-Caldas Y, Dalrymple BP, Reverter A. Porcine tissue-specific regulatory networks derived from meta-analysis of the transcriptome. PLoS One 2012; 7:e46159. [PMID: 23049964 PMCID: PMC3458843 DOI: 10.1371/journal.pone.0046159] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 08/28/2012] [Indexed: 11/19/2022] Open
Abstract
The processes that drive tissue identity and differentiation remain unclear for most tissue types. So are the gene networks and transcription factors (TF) responsible for the differential structure and function of each particular tissue, and this is particularly true for non model species with incomplete genomic resources. To better understand the regulation of genes responsible for tissue identity in pigs, we have inferred regulatory networks from a meta-analysis of 20 gene expression studies spanning 480 Porcine Affymetrix chips for 134 experimental conditions on 27 distinct tissues. We developed a mixed-model normalization approach with a covariance structure that accommodated the disparity in the origin of the individual studies, and obtained the normalized expression of 12,320 genes across the 27 tissues. Using this resource, we constructed a network, based on the co-expression patterns of 1,072 TF and 1,232 tissue specific genes. The resulting network is consistent with the known biology of tissue development. Within the network, genes clustered by tissue and tissues clustered by site of embryonic origin. These clusters were significantly enriched for genes annotated in key relevant biological processes and confirm gene functions and interactions from the literature. We implemented a Regulatory Impact Factor (RIF) metric to identify the key regulators in skeletal muscle and tissues from the central nervous systems. The normalization of the meta-analysis, the inference of the gene co-expression network and the RIF metric, operated synergistically towards a successful search for tissue-specific regulators. Novel among these findings are evidence suggesting a novel key role of ERCC3 as a muscle regulator. Together, our results recapitulate the known biology behind tissue specificity and provide new valuable insights in a less studied but valuable model species.
Collapse
Affiliation(s)
- Dafne Pérez-Montarelo
- Computational and Systems Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO) Animal, Food and Health Sciences, Queensland Bioscience Precinct, St. Lucia, Brisbane, Queensland, Australia
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Nicholas J. Hudson
- Computational and Systems Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO) Animal, Food and Health Sciences, Queensland Bioscience Precinct, St. Lucia, Brisbane, Queensland, Australia
| | - Ana I. Fernández
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Madrid, Spain
| | - Yuliaxis Ramayo-Caldas
- Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Brian P. Dalrymple
- Computational and Systems Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO) Animal, Food and Health Sciences, Queensland Bioscience Precinct, St. Lucia, Brisbane, Queensland, Australia
| | - Antonio Reverter
- Computational and Systems Biology, Commonwealth Scientific and Industrial Research Organisation (CSIRO) Animal, Food and Health Sciences, Queensland Bioscience Precinct, St. Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
23
|
Abstract
Maintaining hematopoietic stem cell (HSC) quiescence is a critical property for the life-long generation of blood cells. Approximately 75% of cells in a highly enriched long-term repopulating HSC (LT-HSC) pool (Lin(-)Sca1(+)c-Kit(hi)CD150(+)CD48(-)) are quiescent, with only a small percentage of the LT-HSCs in cycle. Transcription factor GATA-3 is known to be vital for the development of T cells at multiple stages in the thymus and for Th2 differentiation in the peripheral organs. Although it is well documented that GATA-3 is expressed in HSCs, a role for GATA-3 in any prethymic progenitor cell has not been established. In the present study, we show that Gata3-null mutant mice generate fewer LT-HSCs and that fewer Gata3-null LT-HSCs are in cycle. Furthermore, Gata3 mutant hematopoietic progenitor cells fail to be recruited into an increased cycling state after 5-fluorouracil-induced myelosuppression. Therefore, GATA-3 is required for the maintenance of a normal number of LT-HSCs and for their entry into the cell cycle.
Collapse
|