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










Publication year range
1.
Blood ; 131(15): 1712-1719, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29339402

ABSTRACT

Although an essential role for canonical Notch signaling in generation of hematopoietic stem cells in the embryo and in thymic T-cell development is well established, its role in adult bone marrow (BM) myelopoiesis remains unclear. Some studies, analyzing myeloid progenitors in adult mice with inhibited Notch signaling, implicated distinct roles of canonical Notch signaling in regulation of progenitors for the megakaryocyte, erythroid, and granulocyte-macrophage cell lineages. However, these studies might also have targeted other pathways. Therefore, we specifically deleted, in adult BM, the transcription factor recombination signal-binding protein J κ (Rbpj), through which canonical signaling from all Notch receptors converges. Notably, detailed progenitor staging established that canonical Notch signaling is fully dispensable for all investigated stages of megakaryocyte, erythroid, and myeloid progenitors in steady state unperturbed hematopoiesis, after competitive BM transplantation, and in stress-induced erythropoiesis. Moreover, expression of key regulators of these hematopoietic lineages and Notch target genes were unaffected by Rbpj deficiency in BM progenitor cells.


Subject(s)
Bone Marrow/metabolism , Erythropoiesis , Myelopoiesis , Receptors, Notch/metabolism , Signal Transduction , Stress, Physiological , Animals , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Mice, Transgenic , Receptors, Notch/genetics
2.
Nat Immunol ; 17(6): 666-676, 2016 06.
Article in English | MEDLINE | ID: mdl-27043410

ABSTRACT

According to current models of hematopoiesis, lymphoid-primed multi-potent progenitors (LMPPs) (Lin(-)Sca-1(+)c-Kit(+)CD34(+)Flt3(hi)) and common myeloid progenitors (CMPs) (Lin(-)Sca-1(+)c-Kit(+)CD34(+)CD41(hi)) establish an early branch point for separate lineage-commitment pathways from hematopoietic stem cells, with the notable exception that both pathways are proposed to generate all myeloid innate immune cell types through the same myeloid-restricted pre-granulocyte-macrophage progenitor (pre-GM) (Lin(-)Sca-1(-)c-Kit(+)CD41(-)FcγRII/III(-)CD150(-)CD105(-)). By single-cell transcriptome profiling of pre-GMs, we identified distinct myeloid differentiation pathways: a pathway expressing the gene encoding the transcription factor GATA-1 generated mast cells, eosinophils, megakaryocytes and erythroid cells, and a pathway lacking expression of that gene generated monocytes, neutrophils and lymphocytes. These results identify an early hematopoietic-lineage bifurcation that separates the myeloid lineages before their segregation from other hematopoietic-lineage potential.


Subject(s)
Cell Differentiation , Cell Lineage , Lymphocytes/physiology , Myeloid Cells/physiology , Myeloid Progenitor Cells/physiology , Animals , Antigens, CD/metabolism , Cells, Cultured , Computational Biology , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Hematopoiesis , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Sequence Analysis, RNA , Single-Cell Analysis , Tissue Array Analysis , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
3.
Stem Cell Res ; 11(3): 1129-36, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23978475

ABSTRACT

Hematopoietic stem cells (HSC)(1) supply organisms with life-long output of mature blood cells. To do so, the HSC pool size has to be maintained by HSC self-renewing divisions. PRDM3 and PRDM16 have been documented to regulate HSC self-renewal, maintenance and function. We found Prdm11 to have similar expression patterns in the hematopoietic stem and progenitor cell (HSPC) compartments as Prdm3 and Prdm16. Therefore, we undertook experiments to test if PRDM11 regulates HSC self-renewal, maintenance and function by investigating the Prdm11(-/-) mice. Our data shows that phenotypic HSPCs are intact in bone marrow (BM) of one-year-old Prdm11(-/-) mice. In addition, Prdm11(-/-) mice were able to fully regenerate the hematopoietic system upon BM transplantation (BMT) into lethally irradiated mice with a mild drop in lymphoid output only. Taken together, this suggests that PRDM11, in contrast to PRDM3 and PRDM16, is not directly involved in regulation of HSPCs in mice.


Subject(s)
Carrier Proteins/metabolism , Hematopoietic Stem Cells/cytology , Repressor Proteins/metabolism , Stem Cells/cytology , Animals , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Carrier Proteins/genetics , Hematopoietic Stem Cells/metabolism , Megakaryocytes/cytology , Mice , Mice, Knockout , Platelet Count , Repressor Proteins/genetics , Stem Cells/metabolism , Transcription Factors , Whole-Body Irradiation
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Blood ; 117(13): 3521-8, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21278354

ABSTRACT

Hoxb4 overexpression promotes dramatic expansion of bone marrow (BM) hematopoietic stem cells (HSCs) without leukemic transformation and induces development of definitive HSCs from early embryonic yolk sac and differentiating embryonic stem cells. Knockout studies of Hoxb4 showed little effect on hematopoiesis, but interpretation of these results is obscured by the lack of direct evidence that Hoxb4 is expressed in HSCs and possible compensatory effects of other (Hox) genes. To evaluate accurately the pattern of Hoxb4 expression and to gain a better understanding of the physiologic role of Hoxb4 in the hemato-poietic system, we generated a knock-in Hoxb4-yellow fluorescent protein (YFP) reporter mouse model. We show that BM Lin(-)Sca1(+)c-Kit(+) cells express Hoxb4-YFP and demonstrate functionally in the long-term repopulation assay that definitive HSCs express Hoxb4. Similarly, aorta-gonad-mesonephrous-derived CD45(+)CD144(+) cells, enriched for HSCs, express Hoxb4. Furthermore, yolk sac and placental HSC populations express Hoxb4. Unexpectedly, Hoxb4 expression in the fetal liver HSCs is lower than in the BM, reaching negligible levels in some HSCs, suggesting an insignificant role of Hoxb4 in expansion of fetal liver HSCs. Hoxb4 expression therefore would not appear to correlate with the cycling status of fetal liver HSCs, although highly proliferative HSCs from young BM show strong Hoxb4 expression.


Subject(s)
Bacterial Proteins/genetics , Cell Tracking/methods , Genes, Reporter , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Luminescent Proteins/genetics , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Bacterial Proteins/metabolism , Embryo, Mammalian , Female , Genes, Reporter/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Models, Biological , Pregnancy , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism
11.
PLoS Genet ; 7(12): e1002404, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22216009

ABSTRACT

There is much interest in the mechanisms that regulate adult tissue homeostasis and their relationship to processes governing foetal development. Mice deleted for the Wilms' tumour gene, Wt1, lack kidneys, gonads, and spleen and die at mid-gestation due to defective coronary vasculature. Wt1 is vital for maintaining the mesenchymal-epithelial balance in these tissues and is required for the epithelial-to-mesenchyme transition (EMT) that generates coronary vascular progenitors. Although Wt1 is only expressed in rare cell populations in adults including glomerular podocytes, 1% of bone marrow cells, and mesothelium, we hypothesised that this might be important for homeostasis of adult tissues; hence, we deleted the gene ubiquitously in young and adult mice. Within just a few days, the mice suffered glomerulosclerosis, atrophy of the exocrine pancreas and spleen, severe reduction in bone and fat, and failure of erythropoiesis. FACS and culture experiments showed that Wt1 has an intrinsic role in both haematopoietic and mesenchymal stem cell lineages and suggest that defects within these contribute to the phenotypes we observe. We propose that glomerulosclerosis arises in part through down regulation of nephrin, a known Wt1 target gene. Protein profiling in mutant serum showed that there was no systemic inflammatory or nutritional response in the mutant mice. However, there was a dramatic reduction in circulating IGF-1 levels, which is likely to contribute to the bone and fat phenotypes. The reduction of IGF-1 did not result from a decrease in circulating GH, and there is no apparent pathology of the pituitary and adrenal glands. These findings 1) suggest that Wt1 is a major regulator of the homeostasis of some adult tissues, through both local and systemic actions; 2) highlight the differences between foetal and adult tissue regulation; 3) point to the importance of adult mesenchyme in tissue turnover.


Subject(s)
Glomerulonephritis/genetics , Homeostasis/genetics , Multiple Organ Failure/genetics , WT1 Proteins/physiology , Animals , Atrophy/genetics , Atrophy/pathology , Cell Lineage/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Deletion , Gene Expression Regulation , Glomerulonephritis/pathology , Gonads/embryology , Gonads/metabolism , Gonads/pathology , Hematopoiesis/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Kidney Glomerulus/embryology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Multiple Organ Failure/pathology , Pancreas, Exocrine/embryology , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , Podocytes/metabolism , Podocytes/pathology , Spleen/embryology , Spleen/metabolism , Spleen/pathology , Tamoxifen/pharmacology , WT1 Proteins/genetics
12.
N Engl J Med ; 363(11): 1025-37, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20825315

ABSTRACT

BACKGROUND: The in vivo clinical significance of malignant stem cells remains unclear. METHODS: Patients who have the 5q deletion (del[5q]) myelodysplastic syndrome (interstitial deletions involving the long arm of chromosome 5) have complete clinical and cytogenetic remissions in response to lenalidomide treatment, but they often have relapse. To determine whether the persistence of rare but distinct malignant stem cells accounts for such relapses, we examined bone marrow specimens obtained from seven patients with the del(5q) myelodysplastic syndrome who became transfusion-independent while receiving lenalidomide treatment and entered cytogenetic remission. RESULTS: Virtually all CD34+, CD38+ progenitor cells and stem cells that were positive for CD34 and CD90, with undetectable or low CD38 (CD38−/low), had the 5q deletion before treatment. Although lenalidomide efficiently reduced these progenitors in patients in complete remission, a larger fraction of the minor, quiescent, CD34+,CD38-/low, CD90+ del(5q) stem cells as well as functionally defined del(5q) stem cells remained distinctly resistant to lenalidomide. Over time, lenalidomide resistance developed in most of the patients in partial and complete remission, with recurrence or expansion of the del(5q) clone and clinical and cytogenetic progression. CONCLUSIONS: In these patients with the del(5q) myelodysplastic syndrome, we identified rare and phenotypically distinct del(5q) myelodysplastic syndrome stem cells that were also selectively resistant to therapeutic targeting at the time of complete clinical and cytogenetic remission. (Funded by the EuroCancerStemCell Consortium and others.)


Subject(s)
Antineoplastic Agents/pharmacology , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Drug Resistance/genetics , Myelodysplastic Syndromes/pathology , Neoplastic Stem Cells/drug effects , Thalidomide/analogs & derivatives , ADP-ribosyl Cyclase 1/analysis , Aged , Aged, 80 and over , Antigens, CD34/analysis , Antineoplastic Agents/therapeutic use , Female , Gene Expression , Humans , Lenalidomide , Male , Middle Aged , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Neoplastic Stem Cells/immunology , Phenotype , Remission Induction , Thalidomide/pharmacology , Thalidomide/therapeutic use , Thy-1 Antigens/analysis
13.
Blood ; 115(24): 5061-8, 2010 Jun 17.
Article in English | MEDLINE | ID: mdl-20393130

ABSTRACT

Mice deficient in c-fms-like tyrosine kinase 3 (FLT3) signaling have reductions in early multipotent and lymphoid progenitors, whereas no evident myeloid phenotype has been reported. However, activating mutations of Flt3 are among the most common genetic events in acute myeloid leukemia and mice harboring internal tandem duplications within Flt3 (Flt3-ITD) develop myeloproliferative disease, with characteristic expansion of granulocyte-monocyte (GM) progenitors (GMP), possibly compatible with FLT3-ITD promoting a myeloid fate of multipotent progenitors. Alternatively, FLT3 might be expressed at the earliest stages of GM development. Herein, we investigated the expression, function, and role of FLT3 in recently identified early GMPs. Flt3-cre fate-mapping established that most progenitors and mature progeny of the GM lineage are derived from Flt3-expressing progenitors. A higher expression of FLT3 was found in preGMP compared with GMP, and preGMPs were more responsive to stimulation with FLT3 ligand (FL). Whereas preGMPs and GMPs were reduced in Fl(-/-) mice, megakaryocyte-erythroid progenitors were unaffected and lacked FLT3 expression. Notably, mice deficient in both thrombopoietin (THPO) and FL had a more pronounced GMP phenotype than Thpo(-/-) mice, establishing a role of FL in THPO-dependent and -independent regulation of GMPs, of likely significance for myeloid malignancies with Flt3-ITD mutations.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid, Acute/physiopathology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , Animals , Cell Lineage/physiology , Gene Expression/physiology , Granulocytes/cytology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Myeloid Cells/cytology , Signal Transduction/physiology , Thrombopoietin/genetics
14.
Blood ; 113(15): 3453-60, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19188666

ABSTRACT

Originally cloned from hematopoietic stem cell (HSC) populations and its ligand being extensively used to promote ex vivo HSC expansion, the FMS-like tyrosine kinase 3 (FLT3; also called FLK2) receptor and its ligand (FL) were expected to emerge as an important physiologic regulator of HSC maintenance and expansion. However, the role of FLT3 receptor and ligand in HSC regulation remains unclear and disputed. Herein, using Fl-deficient mice, we establish for the first time that HSC expansion in fetal liver and after transplantation is FL independent. Because previous findings in Flk2(-/-) mice were compatible with an important role of FLT3 receptor in HSC regulation and because alternative ligands might potentially interact directly or indirectly with FLT3 receptor, we here also characterized HSCs in Flk2(-/-) mice. Advanced phenotypic as well as functional evaluation of Flk2(-/-) HSCs showed that the FLT3 receptor is dispensable for HSC steady-state maintenance and expansion after transplantation. Taken together, these studies show that the FLT3 receptor and ligand are not critical regulators of mouse HSCs, neither in steady state nor during fetal or posttransplantation expansion.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Membrane Proteins/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Cell Division/physiology , Flow Cytometry , Graft Survival/physiology , Hematopoiesis/physiology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Phenotype , fms-Like Tyrosine Kinase 3/genetics
15.
Semin Immunol ; 20(4): 213-20, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18752972

ABSTRACT

The prevailing model for adult hematopoiesis postulates that the first lineage commitment step results in a strict separation of common myeloid and common lymphoid pathways. However, the recent identification of granulocyte/monocyte (GM)-lymphoid restricted lymphoid-primed multipotent progenitors (LMPPs) and primitive common myeloid progenitors (CMPs) within the "HSC" compartment provide compelling support for establishment of independent GM-megakaryocyte/erythroid (GM-MkE) and GM-lymphoid commitment pathways as decisive early lineage fate decisions. These changes in lineage potentials are corroborated by corresponding changes in multilineage transcriptional priming, as LMPPs down-regulate MkE priming but become GM-lymphoid transcriptionally primed, whereas CMPs are GM-MkE primed. These distinct biological and molecular relationships are established already in the fetal liver.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Models, Biological , Adult Stem Cells/classification , Animals , Fetal Stem Cells/classification , Hematopoietic Stem Cells/classification , Humans , Signal Transduction
16.
Blood ; 111(7): 3424-34, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18218856

ABSTRACT

Evidence for a novel route of adult hematopoietic stem-cell lineage commitment through Lin-Sca-1+Kit+Flt3hi (LSKFlt3hi) lymphoid-primed multipotent progenitors (LMPPs) with granulocyte/monocyte (GM) and lymphoid but little or no megakaryocyte/erythroid (MkE) potential was recently challenged, as LSKFlt3hi cells were reported to possess MkE potential. Herein, residual (1%-2%) MkE potential segregated almost entirely with LSKFlt3hi cells expressing the thrombopoietin receptor (Mpl), whereas LSKFlt3hiMpl- LMPPs lacked significant MkE potential in vitro and in vivo, but sustained combined GM and lymphoid potentials, and coexpressed GM and lymphoid but not MkE transcriptional lineage programs. Gradually increased transcriptional lymphoid priming in single LMPPs from Rag1GFP mice was shown to occur in the presence of maintained GM lineage priming, but gradually reduced GM lineage potential. These functional and molecular findings reinforce the existence of GM/lymphoid-restricted progenitors with dramatically down-regulated probability for committing toward MkE fates, and support that lineage restriction occurs through gradual rather than abrupt changes in specific lineage potentials.


Subject(s)
Lymphocytes/metabolism , Multipotent Stem Cells/metabolism , Myeloid Cells/metabolism , Receptors, Thrombopoietin/metabolism , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Down-Regulation/genetics , Homeodomain Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Thrombopoietin/genetics , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
17.
Cancer Cell ; 12(4): 367-80, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17936561

ABSTRACT

Despite their known transforming properties, the effects of leukemogenic FLT3-ITD mutations on hematopoietic stem and multipotent progenitor cells and on hematopoietic differentiation are not well understood. We report a mouse model harboring an ITD in the murine Flt3 locus that develops myeloproliferative disease resembling CMML and further identified FLT3-ITD mutations in a subset of human CMML. These findings correlated with an increase in number, cell cycling, and survival of multipotent stem and progenitor cells in an ITD dose-dependent manner in animals that exhibited alterations within their myeloid progenitor compartments and a block in normal B cell development. This model provides insights into the consequences of constitutive signaling by an oncogenic tyrosine kinase on hematopoietic progenitor quiescence, function, and cell fate.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cells/metabolism , Leukemia, Myelomonocytic, Chronic/metabolism , Multipotent Stem Cells/metabolism , Mutation , Myeloproliferative Disorders/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Exons , Gene Expression Regulation, Neoplastic , Genotype , Hematopoietic Stem Cells/pathology , Humans , Kaplan-Meier Estimate , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/pathology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Phenotype , Signal Transduction , fms-Like Tyrosine Kinase 3/genetics
18.
Blood ; 110(8): 2955-64, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17540845

ABSTRACT

The molecular pathways regulating lymphoid priming, fate, and development of multipotent bone marrow (BM) stem/progenitor cells that continuously replace thymic progenitors remain largely unknown. Herein, we show that fms-like tyrosine kinase 3 (Flt3) ligand (Fl)-deficient mice have distinct reductions in the earliest thymic progenitors in fetal, postnatal, and adult thymus. A critical role of FL in thymopoiesis was particularly evident in the absence of interleukin-7 receptor alpha (IL-7Ralpha) signaling. Fl-/-Il-7r-/- mice have extensive reductions in fetal and postnatal thymic progenitors that result in a loss of active thymopoiesis in adult mice, demonstrating an indispensable role of FL in IL-7Ralpha-independent fetal and adult T lymphopoiesis. Moreover, we establish a unique and critical role of FL, distinct from that of IL-7Ralpha, in regulation of the earliest lineage-negative (Lin(-)) Lin(-)SCA1+KIT+ (LSK) FLT3(hi) lymphoid-primed multipotent progenitors in BM, demonstrating a key role of FLT3 signaling in regulating the very earliest stages of lymphoid progenitors.


Subject(s)
Hematopoietic Stem Cells/cytology , Lymphopoiesis/physiology , Membrane Proteins/metabolism , Receptors, Interleukin-7/metabolism , T-Lymphocytes/cytology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Base Sequence , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Fetus , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression/immunology , Hematopoietic Stem Cells/immunology , Image Processing, Computer-Assisted , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/embryology , Thymus Gland/immunology , Thymus Gland/pathology
19.
Curr Opin Hematol ; 14(4): 315-21, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17534155

ABSTRACT

PURPOSE OF REVIEW: This review addresses recently reported evidence for alternative cellular pathways for haematopoietic stem cell lineage commitment. RECENT FINDINGS: Using various approaches, several laboratories suggested the existence of adult as well as foetal multipotent progenitor cells with combined B cell, T cell and granulocyte/macrophage potential, but little or no megakaryocyte/erythroid potential. Compared with haematopoietic stem cells, these multipotent progenitor cells exhibited downregulated transcriptional expression of genes of the megakaryocyte/erythroid lineages and upregulated expression of lymphoid lineage genes. The existence of these lineage-restricted multipotent progenitor cells suggests that the first lineage commitment step of haematopoietic stem cells does not result in strict separation into myelopoiesis and lymphopoiesis, and that there might be alternative pathways for commitment toward different lineage fates. These findings have been questioned by other studies, however. To resolve this controversy and establish the complete road map for haematopoietic lineage commitment, improved tools and more stringent standards for how to identify and characterize lineage fate options of distinct stem and progenitor cells are needed. SUMMARY: Current and future progress in establishing the complete cellular roadmap for haematopoietic lineage commitment will permit identification and characterization of key regulators of lineage fate decisions in haematopoietic stem cells.


Subject(s)
Cell Lineage , Hematopoietic Stem Cells/cytology , Adult Stem Cells/cytology , Fetal Stem Cells/cytology , Humans , Lymphopoiesis , Multipotent Stem Cells/cytology , Myelopoiesis
20.
Immunity ; 26(4): 407-19, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17433729

ABSTRACT

Recent studies implicated the existence of adult lymphoid-primed multipotent progenitors (LMPPs) with little or no megakaryocyte-erythroid potential, questioning common myeloid and lymphoid progenitors as obligate intermediates in hematopoietic stem cell (HSC) lineage commitment. However, the existence of LMPPs remains contentious. Herein, global and single-cell analyses revealed a hierarchical organization of transcriptional lineage programs, with downregulation of megakaryocyte-erythroid genes from HSCs to LMPPs, sustained granulocyte-monocyte priming, and upregulation of common lymphoid (but not B and T cell-specific) genes. These biological and molecular relationships, implicating almost mutual exclusion of megakaryocyte-erythroid and lymphoid pathways, are established already in fetal hematopoiesis, as evidenced by existence of LMPPs in fetal liver. The identification of LMPPs and hierarchically ordered transcriptional activation and downregulation of distinct lineage programs is compatible with a model for HSC lineage commitment in which the probability for undergoing different lineage commitment fates changes gradually when progressing from HSCs to LMPPs.


Subject(s)
Adult Stem Cells/immunology , Cell Lineage/genetics , Fetus/immunology , Gene Expression Regulation, Developmental , Lymphocytes/immunology , Multipotent Stem Cells/immunology , Adult Stem Cells/cytology , Animals , Fetal Development/genetics , Fetus/cytology , Gene Expression Profiling , Lymphocytes/cytology , Mice , Multipotent Stem Cells/cytology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...