Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Stem Cells Transl Med ; 12(11): 720-726, 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37706539

ABSTRACT

Hematopoiesis is maintained by hematopoietic stem cells (HSCs) that replenish all blood lineages throughout life. It is well-established that the HSC pool is functionally heterogeneous consisting of cells differing in longevity, self-renewal ability, cell proliferation, and lineage differentiation. Although HSCs can be identified through the Lineage-Sca-1+c-Kit+CD48-CD34-CD150+ immunophenotype, the cell surface marker combination does not permit absolute purification of functional HSCs with long-term reconstituting ability. Therefore, prospective isolation of long-term HSCs is crucial for mechanistic understanding of the biological functions of HSCs and for resolving functional heterogeneity within the HSC population. Here, we show that the combination of CD229 and CD49b cell surface markers within the phenotypic HSC compartment identifies a subset of multipotent progenitor (MPP) cells with high proliferative activity and short-term reconstituting ability. Thus, the addition of CD229 and CD49b to conventional HSC markers permits prospective isolation of functional HSCs by distinguishing MPPs in the HSC compartment.


Subject(s)
Hematopoietic Stem Cells , Integrin alpha2 , Animals , Mice , Integrin alpha2/metabolism , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells , Cell Differentiation , Hematopoiesis , Mice, Inbred C57BL
2.
BMC Genomics ; 24(1): 205, 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37069502

ABSTRACT

BACKGROUND: RNA sequencing has become the mainstay for studies of gene expression. Still, analysis of rare cells with random hexamer priming - to allow analysis of a broader range of transcripts - remains challenging. RESULTS: We here describe a tagmentation-based, rRNA blocked, random hexamer primed RNAseq approach (T-RHEX-RNAseq) for generating stranded RNAseq libraries from very low numbers of FACS sorted cells without RNA purification steps. CONCLUSION: T-RHEX-RNAseq provides an easy-to-use, time efficient and automation compatible method for generating stranded RNAseq libraries from rare cells.


Subject(s)
High-Throughput Nucleotide Sequencing , RNA, Ribosomal , High-Throughput Nucleotide Sequencing/methods , RNA, Ribosomal/genetics , Base Sequence , Sequence Analysis, RNA/methods , Gene Expression Profiling/methods
3.
Stem Cell Reports ; 17(7): 1546-1560, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35714596

ABSTRACT

Hematopoiesis is maintained by functionally diverse lineage-biased hematopoietic stem cells (HSCs). The functional significance of HSC heterogeneity and the regulatory mechanisms underlying lineage bias are not well understood. However, absolute purification of HSC subtypes with a pre-determined behavior remains challenging, highlighting the importance of continued efforts toward prospective isolation of homogeneous HSC subsets. In this study, we demonstrate that CD49b subdivides the most primitive HSC compartment into functionally distinct subtypes: CD49b- HSCs are highly enriched for myeloid-biased and the most durable cells, while CD49b+ HSCs are enriched for multipotent cells with lymphoid bias and reduced self-renewal ability. We further demonstrate considerable transcriptional similarities between CD49b- and CD49b+ HSCs but distinct differences in chromatin accessibility. Our studies highlight the diversity of HSC functional behaviors and provide insights into the molecular regulation of HSC heterogeneity through transcriptional and epigenetic mechanisms.


Subject(s)
Hematopoietic Stem Cells , Integrin alpha2 , Cell Differentiation/genetics , Cell Lineage/genetics , Hematopoiesis/genetics , Multipotent Stem Cells
4.
Front Immunol ; 13: 880668, 2022.
Article in English | MEDLINE | ID: mdl-35603175

ABSTRACT

The development of B cells relies on an intricate network of transcription factors critical for developmental progression and lineage commitment. In the B cell developmental trajectory, a temporal switch from predominant Foxo3 to Foxo1 expression occurs at the CLP stage. Utilizing VAV-iCre mediated conditional deletion, we found that the loss of FOXO3 impaired B cell development from LMPP down to B cell precursors, while the loss of FOXO1 impaired B cell commitment and resulted in a complete developmental block at the CD25 negative proB cell stage. Strikingly, the combined loss of FOXO1 and FOXO3 resulted in the failure to restrict the myeloid potential of CLPs and the complete loss of the B cell lineage. This is underpinned by the failure to enforce the early B-lineage gene regulatory circuitry upon a predominantly pre-established open chromatin landscape. Altogether, this demonstrates that FOXO3 and FOXO1 cooperatively govern early lineage restriction and initiation of B-lineage commitment in CLPs.


Subject(s)
Hematopoiesis , Lymphoid Progenitor Cells , B-Lymphocytes/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Hematopoiesis/genetics , Lymphoid Progenitor Cells/metabolism , Precursor Cells, B-Lymphoid/metabolism
5.
Nat Immunol ; 17(12): 1424-1435, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27695000

ABSTRACT

The final stages of restriction to the T cell lineage occur in the thymus after the entry of thymus-seeding progenitors (TSPs). The identity and lineage potential of TSPs remains unclear. Because the first embryonic TSPs enter a non-vascularized thymic rudiment, we were able to directly image and establish the functional and molecular properties of embryonic thymopoiesis-initiating progenitors (T-IPs) before their entry into the thymus and activation of Notch signaling. T-IPs did not include multipotent stem cells or molecular evidence of T cell-restricted progenitors. Instead, single-cell molecular and functional analysis demonstrated that most fetal T-IPs expressed genes of and had the potential to develop into lymphoid as well as myeloid components of the immune system. Moreover, studies of embryos deficient in the transcriptional regulator RBPJ demonstrated that canonical Notch signaling was not involved in pre-thymic restriction to the T cell lineage or the migration of T-IPs.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Lymphoid Progenitor Cells/physiology , Myeloid Progenitor Cells/physiology , Receptors, Notch/metabolism , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Cells, Cultured , Fetus , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction
6.
Blood ; 128(19): 2338-2342, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27707736

ABSTRACT

BCL11A, a repressor of human fetal (γ-)globin expression, is required for immune and hematopoietic stem cell functions and brain development. Regulatory sequences within the gene, which are subject to genetic variation affecting fetal globin expression, display hallmarks of an erythroid enhancer in cell lines and transgenic mice. As such, this enhancer is a novel, attractive target for therapeutic gene editing. To explore the roles of such sequences in vivo, we generated mice in which the orthologous 10-kb intronic sequences were removed. Bcl11a enhancer-deleted mice, Bcl11a(Δenh), phenocopy the BCL11A-null state with respect to alterations of globin expression, yet are viable and exhibit no observable blood, brain, or other abnormalities. These preclinical findings provide strong in vivo support for genetic modification of the enhancer for therapy of hemoglobin disorders.


Subject(s)
Carrier Proteins/metabolism , Enhancer Elements, Genetic/genetics , Erythroid Cells/metabolism , Nuclear Proteins/metabolism , Animals , Base Sequence , Cell Compartmentation , DNA-Binding Proteins , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Gene Silencing , Humans , Mice , Mice, Transgenic , Repressor Proteins
7.
Cell Rep ; 16(12): 3181-3194, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27653684

ABSTRACT

B cell CLL/lymphoma 11A (BCL11A) is a transcription factor and regulator of hemoglobin switching that has emerged as a promising therapeutic target for sickle cell disease and thalassemia. In the hematopoietic system, BCL11A is required for B lymphopoiesis, yet its role in other hematopoietic cells, especially hematopoietic stem cells (HSCs) remains elusive. The extensive expression of BCL11A in hematopoiesis implicates context-dependent roles, highlighting the importance of fully characterizing its function as part of ongoing efforts for stem cell therapy and regenerative medicine. Here, we demonstrate that BCL11A is indispensable for normal HSC function. Bcl11a deficiency results in HSC defects, typically observed in the aging hematopoietic system. We find that downregulation of cyclin-dependent kinase 6 (Cdk6), and the ensuing cell-cycle delay, correlate with HSC dysfunction. Our studies define a mechanism for BCL11A in regulation of HSC function and have important implications for the design of therapeutic approaches to targeting BCL11A.


Subject(s)
Carrier Proteins/genetics , Cellular Senescence/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/pathology , Nuclear Proteins/genetics , Animals , Cyclin-Dependent Kinase 6/biosynthesis , DNA-Binding Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Repressor Proteins
8.
Nature ; 527(7577): 192-7, 2015 Nov 12.
Article in English | MEDLINE | ID: mdl-26375006

ABSTRACT

Enhancers, critical determinants of cellular identity, are commonly recognized by correlative chromatin marks and gain-of-function potential, although only loss-of-function studies can demonstrate their requirement in the native genomic context. Previously, we identified an erythroid enhancer of human BCL11A, subject to common genetic variation associated with the fetal haemoglobin level, the mouse orthologue of which is necessary for erythroid BCL11A expression. Here we develop pooled clustered regularly interspaced palindromic repeat (CRISPR)-Cas9 guide RNA libraries to perform in situ saturating mutagenesis of the human and mouse enhancers. This approach reveals critical minimal features and discrete vulnerabilities of these enhancers. Despite conserved function of the composite enhancers, their architecture diverges. The crucial human sequences appear to be primate-specific. Through editing of primary human progenitors and mouse transgenesis, we validate the BCL11A erythroid enhancer as a target for fetal haemoglobin reinduction. The detailed enhancer map will inform therapeutic genome editing, and the screening approach described here is generally applicable to functional interrogation of non-coding genomic elements.


Subject(s)
CRISPR-Associated Proteins/metabolism , Carrier Proteins/genetics , Enhancer Elements, Genetic/genetics , Genetic Engineering , Mutagenesis/genetics , Nuclear Proteins/genetics , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA-Binding Proteins , Erythroblasts/metabolism , Fetal Hemoglobin/genetics , Genome/genetics , Humans , Mice , Molecular Sequence Data , Organ Specificity , RNA, Guide, Kinetoplastida/genetics , Repressor Proteins , Reproducibility of Results , Species Specificity
9.
Cell Stem Cell ; 13(4): 492-505, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24035353

ABSTRACT

Stem cell differentiation pathways are most often studied at the population level, whereas critical decisions are executed at the level of single cells. We have established a highly multiplexed, quantitative PCR assay to profile in an unbiased manner a panel of all commonly used cell surface markers (280 genes) from individual cells. With this method, we analyzed over 1,500 single cells throughout the mouse hematopoietic system and illustrate its utility for revealing important biological insights. The comprehensive single cell data set permits mapping of the mouse hematopoietic stem cell differentiation hierarchy by computational lineage progression analysis. Further profiling of 180 intracellular regulators enabled construction of a genetic network to assign the earliest differentiation event during hematopoietic lineage specification. Analysis of acute myeloid leukemia elicited by MLL-AF9 uncovered a distinct cellular hierarchy containing two independent self-renewing lineages with different clonal activities. The strategy has broad applicability in other cellular systems.


Subject(s)
Antigens, Surface/analysis , Antigens, Surface/genetics , Single-Cell Analysis , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hematopoietic System/cytology , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Polymerase Chain Reaction
10.
Cell Stem Cell ; 13(5): 535-48, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24054998

ABSTRACT

In jawed vertebrates, development of an adaptive immune-system is essential for protection of the born organism against otherwise life-threatening pathogens. Myeloid cells of the innate immune system are formed early in development, whereas lymphopoiesis has been suggested to initiate much later, following emergence of definitive hematopoietic stem cells (HSCs). Herein, we demonstrate that the embryonic lymphoid commitment process initiates earlier than previously appreciated, prior to emergence of definitive HSCs, through establishment of a previously unrecognized entirely immune-restricted and lymphoid-primed progenitor. Notably, this immune-restricted progenitor appears to first emerge in the yolk sac and contributes physiologically to the establishment of lymphoid and some myeloid components of the immune-system, establishing the lymphomyeloid lineage restriction process as an early and physiologically important lineage-commitment step in mammalian hematopoiesis.


Subject(s)
Hematopoietic Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cells, Cultured , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Hematopoietic Stem Cells/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Male , Mice , Myeloid Cells/cytology , Myeloid Cells/metabolism , Polymerase Chain Reaction
12.
Elife ; 2: e00633, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23795291

ABSTRACT

Here, we describe that lysine-specific demethylase 1 (Lsd1/KDM1a), which demethylates histone H3 on Lys4 or Lys9 (H3K4/K9), is an indispensible epigenetic governor of hematopoietic differentiation. Integrative genomic analysis, combining global occupancy of Lsd1, genome-wide analysis of its substrates H3K4 monomethylation and dimethylation, and gene expression profiling, reveals that Lsd1 represses hematopoietic stem and progenitor cell (HSPC) gene expression programs during hematopoietic differentiation. We found that Lsd1 acts at transcription start sites, as well as enhancer regions. Loss of Lsd1 was associated with increased H3K4me1 and H3K4me2 methylation on HSPC genes and gene derepression. Failure to fully silence HSPC genes compromised differentiation of hematopoietic stem cells as well as mature blood cell lineages. Collectively, our data indicate that Lsd1-mediated concurrent repression of enhancer and promoter activity of stem and progenitor cell genes is a pivotal epigenetic mechanism required for proper hematopoietic maturation. DOI:http://dx.doi.org/10.7554/eLife.00633.001.


Subject(s)
Hematopoietic Stem Cells/cytology , Histone Demethylases/physiology , Cell Differentiation , Gene Expression Profiling , Histone Demethylases/genetics , Humans
13.
Cell Rep ; 3(6): 1766-76, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23727242

ABSTRACT

Whether signals mediated via growth factor receptors (GFRs) might influence lineage fate in multipotent progenitors (MPPs) is unclear. We explored this issue in a mouse knockin model of gain-of-function Flt3-ITD mutation because FLT3-ITDs are paradoxically restricted to acute myeloid leukemia even though Flt3 primarily promotes lymphoid development during normal hematopoiesis. When expressed in MPPs, Flt3-ITD collaborated with Runx1 mutation to induce high-penetrance aggressive leukemias that were exclusively of the myeloid phenotype. Flt3-ITDs preferentially expanded MPPs with reduced lymphoid and increased myeloid transcriptional priming while compromising early B and T lymphopoiesis. Flt3-ITD-induced myeloid lineage bias involved upregulation of the transcription factor Pu.1, which is a direct target gene of Stat3, an aberrantly activated target of Flt3-ITDs, further establishing how lineage bias can be inflicted on MPPs through aberrant GFR signaling. Collectively, these findings provide new insights into how oncogenic mutations might subvert the normal process of lineage commitment and dictate the phenotype of resulting malignancies.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Multipotent Stem Cells/cytology , Myeloid Cells/cytology , fms-Like Tyrosine Kinase 3/physiology , Animals , Cell Differentiation/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Flow Cytometry/methods , Gene Expression , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Microarray Analysis , Multipotent Stem Cells/immunology , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/pathology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Cells/pathology , Signal Transduction , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
14.
Blood ; 120(12): 2412-6, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22869792

ABSTRACT

MicroRNAs (miRs) are involved in many aspects of normal and malignant hematopoiesis, including hematopoietic stem cell (HSC) self-renewal, proliferation, and terminal differentiation. However, a role for miRs in the generation of the earliest stages of lineage committed progenitors from HSCs has not been identified. Using Dicer inactivation, we show that the miR complex is not only essential for HSC maintenance but is specifically required for their erythroid programming and subsequent generation of committed erythroid progenitors. In bipotent pre-MegEs, loss of Dicer up-regulated transcription factors preferentially expressed in megakaryocyte progenitors (Gata2 and Zfpm1) and decreased expression of the erythroid-specific Klf1 transcription factor. These results show a specific requirement for Dicer in acquisition of erythroid lineage programming and potential in HSCs and their subsequent erythroid lineage differentiation, and in particular indicate a role for the miR complex in achieving proper balance of lineage-specific transcriptional regulators necessary for HSC multilineage potential to be maintained.


Subject(s)
Cell Lineage , DEAD-box RNA Helicases/physiology , Erythroid Cells/cytology , Erythroid Cells/metabolism , Hematopoietic Stem Cells/cytology , Megakaryocyte Progenitor Cells/cytology , Ribonuclease III/physiology , Animals , Biomarkers/metabolism , Blotting, Western , Cell Differentiation , DEAD-box RNA Helicases/antagonists & inhibitors , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Integrases/metabolism , Megakaryocyte Progenitor Cells/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ribonuclease III/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Nat Immunol ; 13(4): 412-9, 2012 Feb 19.
Article in English | MEDLINE | ID: mdl-22344248

ABSTRACT

The stepwise commitment from hematopoietic stem cells in the bone marrow to T lymphocyte-restricted progenitors in the thymus represents a paradigm for understanding the requirement for distinct extrinsic cues during different stages of lineage restriction from multipotent to lineage-restricted progenitors. However, the commitment stage at which progenitors migrate from the bone marrow to the thymus remains unclear. Here we provide functional and molecular evidence at the single-cell level that the earliest progenitors in the neonatal thymus had combined granulocyte-monocyte, T lymphocyte and B lymphocyte lineage potential but not megakaryocyte-erythroid lineage potential. These potentials were identical to those of candidate thymus-seeding progenitors in the bone marrow, which were closely related at the molecular level. Our findings establish the distinct lineage-restriction stage at which the T cell lineage-commitment process transits from the bone marrow to the remote thymus.


Subject(s)
B-Lymphocytes/cytology , Cell Lineage/immunology , Lymphoid Progenitor Cells/cytology , Myeloid Cells/cytology , Precursor Cells, B-Lymphoid/cytology , T-Lymphocytes/cytology , Animals , Cell Separation , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Lymphoid Progenitor Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Thymus Gland/cytology
16.
EMBO J ; 31(2): 351-65, 2012 Jan 18.
Article in English | MEDLINE | ID: mdl-22068055

ABSTRACT

The transcription factors that control lineage specification of haematopoietic stem cells (HSCs) have been well described for the myeloid and lymphoid lineages, whereas transcriptional control of erythroid (E) and megakaryocytic (Mk) fate is less understood. We here use conditional removal of the GATA-1 and FOG-1 transcription factors to identify FOG-1 as required for the formation of all committed Mk- and E-lineage progenitors, whereas GATA-1 was observed to be specifically required for E-lineage commitment. FOG-1-deficient HSCs and preMegEs, the latter normally bipotent for the Mk and E lineages, underwent myeloid transcriptional reprogramming, and formed myeloid, but not erythroid and megakaryocytic cells in vitro. These results identify FOG-1 and GATA-1 as required for formation of bipotent Mk/E progenitors and their E-lineage commitment, respectively, and show that FOG-1 mediates transcriptional Mk/E programming of HSCs as well as their subsequent Mk/E-lineage commitment. Finally, C/EBPs and FOG-1 exhibited transcriptional cross-regulation in early myelo-erythroid progenitors making their functional antagonism a potential mechanism for separation of the myeloid and Mk/E lineages.


Subject(s)
Erythropoiesis/genetics , GATA1 Transcription Factor/physiology , Gene Expression Regulation, Developmental/genetics , Megakaryocyte-Erythroid Progenitor Cells/cytology , Nuclear Proteins/physiology , Thrombopoiesis/genetics , Transcription Factors/physiology , Animals , Bone Marrow Cells/cytology , CCAAT-Enhancer-Binding Protein-beta/deficiency , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Proteins/deficiency , CCAAT-Enhancer-Binding Proteins/genetics , Cell Lineage , Cells, Cultured/cytology , Colony-Forming Units Assay , Erythroid Precursor Cells/cytology , GATA1 Transcription Factor/genetics , Megakaryocyte Progenitor Cells/cytology , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic
17.
Blood ; 118(13): 3613-21, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21813452

ABSTRACT

Acquisition of homozygous activating growth factor receptor mutations might accelerate cancer progression through a simple gene-dosage effect. Internal tandem duplications (ITDs) of FLT3 occur in approximately 25% cases of acute myeloid leukemia and induce ligand-independent constitutive signaling. Homozygous FLT3-ITDs confer an adverse prognosis and are frequently detected at relapse. Using a mouse knockin model of Flt3-internal tandem duplication (Flt3-ITD)-induced myeloproliferation, we herein demonstrate that the enhanced myeloid phenotype and expansion of granulocyte-monocyte and primitive Lin(-)Sca1(+)c-Kit(+) progenitors in Flt3-ITD homozygous mice can in part be mediated through the loss of the second wild-type allele. Further, whereas autocrine FLT3 ligand production has been implicated in FLT3-ITD myeloid malignancies and resistance to FLT3 inhibitors, we demonstrate here that the mouse Flt3(ITD/ITD) myeloid phenotype is FLT3 ligand-independent.


Subject(s)
Gene Dosage/physiology , Gene Duplication/physiology , Loss of Heterozygosity/physiology , Membrane Proteins/genetics , Myeloproliferative Disorders/genetics , fms-Like Tyrosine Kinase 3/physiology , Alleles , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Cells/physiology , Cell Proliferation , Cells, Cultured , Gene Knock-In Techniques , Loss of Heterozygosity/genetics , Male , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Tandem Repeat Sequences/genetics , Tandem Repeat Sequences/physiology , fms-Like Tyrosine Kinase 3/metabolism
18.
Blood ; 118(6): 1544-8, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21628405

ABSTRACT

Lymphoid-primed multipotent progenitors with down-regulated megakaryocyte-erythroid (MkE) potential are restricted to cells with high levels of cell-surface FLT3 expression, whereas HSCs and MkE progenitors lack detectable cell-surface FLT3. These findings are compatible with FLT3 cell-surface expression not being detectable in the fully multipotent stem/progenitor cell compartment in mice. If so, this process could be distinct from human hematopoiesis, in which FLT3 already is expressed in multipotent stem/progenitor cells. The expression pattern of Flt3 (mRNA) and FLT3 (protein) in multipotent progenitors is of considerable relevance for mouse models in which prognostically important Flt3 mutations are expressed under control of the endogenous mouse Flt3 promoter. Herein, we demonstrate that mouse Flt3 expression initiates in fully multipotent progenitors because in addition to lymphoid and granulocyte-monocyte progenitors, FLT3(-) Mk- and E-restricted downstream progenitors are also highly labeled when Flt3-Cre fate mapping is applied.


Subject(s)
Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , fms-Like Tyrosine Kinase 3/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Lineage/genetics , Cell Membrane/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Flow Cytometry , Granulocyte Precursor Cells/cytology , Granulocyte Precursor Cells/metabolism , Hematopoietic Stem Cells/cytology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/cytology , Monocytes/metabolism , Multipotent Stem Cells/cytology , Reverse Transcriptase Polymerase Chain Reaction , fms-Like Tyrosine Kinase 3/metabolism
19.
Epigenetics Chromatin ; 4(1): 9, 2011 Jun 06.
Article in English | MEDLINE | ID: mdl-21645363

ABSTRACT

BACKGROUND: In self-renewing, pluripotent cells, bivalent chromatin modification is thought to silence (H3K27me3) lineage control genes while 'poising' (H3K4me3) them for subsequent activation during differentiation, implying an important role for epigenetic modification in directing cell fate decisions. However, rather than representing an equivalently balanced epigenetic mark, the patterns and levels of histone modifications at bivalent genes can vary widely and the criteria for identifying this chromatin signature are poorly defined. RESULTS: Here, we initially show how chromatin status alters during lineage commitment and differentiation at a single well characterised bivalent locus. In addition we have determined how chromatin modifications at this locus change with gene expression in both ensemble and single cell analyses. We also show, on a global scale, how mRNA expression may be reflected in the ratio of H3K4me3/H3K27me3. CONCLUSIONS: While truly 'poised' bivalently modified genes may exist, the original hypothesis that all bivalent genes are epigenetically premarked for subsequent expression might be oversimplistic. In fact, from the data presented in the present work, it is equally possible that many genes that appear to be bivalent in pluripotent and multipotent cells may simply be stochastically expressed at low levels in the process of multilineage priming. Although both situations could be considered to be forms of 'poising', the underlying mechanisms and the associated implications are clearly different.

20.
Blood ; 118(5): 1291-3, 2011 Aug 04.
Article in English | MEDLINE | ID: mdl-21670475

ABSTRACT

GATA3 has been identified as a master regulator of T helper cells, as well as being important for early thymic progenitors and T-cell commitment. However, Gata3 expression initiates already at the hematopoietic stem cell (HSC) level, implicating a potential role also in the regulation of HSCs. Herein we used a conditional Gata3 knockout strategy in which Gata3 expression was completely deleted from the earliest stage of embryonic hematopoietic development after emergence of HSCs from hemogenic endothelium. Through a detailed analysis of HSCs at the phenotypic and functional level, we demonstrate that steady-state levels of HSCs are normal in Gata3(fl/fl)Vav-Cre(tg/+) mice. Moreover, through long-term primary and secondary transplantation experiments, we also unequivocally demonstrate that Gata3 has a redundant role in post-transplantation HSC self-renewal.


Subject(s)
Cell Proliferation , GATA3 Transcription Factor/physiology , Hematopoietic Stem Cells/physiology , Animals , Cells, Cultured , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...