Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
2.
J Immunol ; 206(11): 2700-2713, 2021 06 01.
Article En | MEDLINE | ID: mdl-34021049

B lymphocyte development is dependent on the interplay between the chromatin landscape and lineage-specific transcription factors. It has been suggested that B lineage commitment is associated with major changes in the nuclear chromatin environment, proposing a critical role for lineage-specific transcription factors in the formation of the epigenetic landscape. In this report, we have used chromosome conformation capture in combination with assay for transposase-accessible chromatin sequencing analysis to enable highly efficient annotation of both proximal and distal transcriptional control elements to genes activated in B lineage specification in mice. A large majority of these genes were annotated to at least one regulatory element with an accessible chromatin configuration in multipotent progenitors. Furthermore, the majority of binding sites for the key regulators of B lineage specification, EBF1 and PAX5, occurred in already accessible regions. EBF1 did, however, cause a dynamic change in assay for transposase-accessible chromatin accessibility and was critical for an increase in distal promoter-enhancer interactions. Our data unravel an extensive epigenetic priming at regulatory elements annotated to lineage-restricted genes and provide insight into the interplay between the epigenetic landscape and transcription factors in cell specification.


B-Lymphocytes/immunology , Epigenesis, Genetic/immunology , PAX5 Transcription Factor/immunology , Trans-Activators/immunology , Animals , Epigenesis, Genetic/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , PAX5 Transcription Factor/deficiency , PAX5 Transcription Factor/genetics , Trans-Activators/deficiency , Trans-Activators/genetics
3.
Blood ; 137(22): 3037-3049, 2021 06 03.
Article En | MEDLINE | ID: mdl-33619557

Genes encoding B lineage-restricted transcription factors are frequently mutated in B-lymphoid leukemias, suggesting a close link between normal and malignant B-cell development. One of these transcription factors is early B-cell factor 1 (EBF1), a protein of critical importance for lineage specification and survival of B-lymphoid progenitors. Here, we report that impaired EBF1 function in mouse B-cell progenitors results in reduced expression of Myc. Ectopic expression of MYC partially rescued B-cell expansion in the absence of EBF1 both in vivo and in vitro. Using chromosome conformation analysis in combination with ATAC-sequencing, chromatin immunoprecipitation-sequencing, and reporter gene assays, six EBF1-responsive enhancer elements were identified within the Myc locus. CRISPR-Cas9-mediated targeting of EBF1-binding sites identified one element of key importance for Myc expression and pro-B cell expansion. These data provide evidence that Myc is a direct target of EBF1. Furthermore, chromatin immunoprecipitation-sequencing analysis revealed that several regulatory elements in the Myc locus are targets of PAX5. However, ectopic expression of PAX5 in EBF1-deficient cells inhibits the cell cycle and reduces Myc expression, suggesting that EBF1 and PAX5 act in an opposing manner to regulate Myc levels. This hypothesis is further substantiated by the finding that Pax5 inactivation reduces requirements for EBF1 in pro-B-cell expansion. The binding of EBF1 and PAX5 to regulatory elements in the human MYC gene in a B-cell acute lymphoblastic leukemia cell line indicates that the EBF1:PAX5:MYC regulatory loop is conserved and may control both normal and malignant B-cell development.


Gene Expression Regulation, Leukemic , PAX5 Transcription Factor/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cells, B-Lymphoid/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Trans-Activators/metabolism , Animals , Cell Proliferation , Mice , Mice, Knockout , PAX5 Transcription Factor/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cells, B-Lymphoid/pathology , Proto-Oncogene Proteins c-myc/genetics , Response Elements , Trans-Activators/genetics
4.
J Immunol ; 205(5): 1419-1432, 2020 09 01.
Article En | MEDLINE | ID: mdl-32747500

Maturation of lymphoid cells is controlled by the action of stage and lineage-restricted transcription factors working in concert with the general transcription and chromatin remodeling machinery to regulate gene expression. To better understand this functional interplay, we used Biotin Identification in human embryonic kidney cells to identify proximity interaction partners for GATA3, TCF7 (TCF1), SPI1, HLF, IKZF1, PAX5, ID1, and ID2. The proximity interaction partners shared among the lineage-restricted transcription factors included ARID1a, a BRG1-associated factor complex component. CUT&RUN analysis revealed that ARID1a shared binding with TCF7 and GATA3 at a substantial number of putative regulatory elements in mouse T cell progenitors. In support of an important function for ARID1a in lymphocyte development, deletion of Arid1a in early lymphoid progenitors in mice resulted in a pronounced developmental arrest in early T cell development with a reduction of CD4+CD8+ cells and a 20-fold reduction in thymic cellularity. Exploring gene expression patterns in DN3 cells from Wt and Arid1a-deficient mice suggested that the developmental block resided in the DN3a to DN3b transition, indicating a deficiency in ß-selection. Our work highlights the critical importance of functional interactions between stage and lineage-restricted factors and the basic transcription machinery during lymphocyte differentiation.


CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Lymphocytes/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Line , Cell Lineage/genetics , Cell Lineage/immunology , Chromatin/genetics , Chromatin/immunology , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , Gene Expression/genetics , Gene Expression/immunology , HEK293 Cells , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Transcription, Genetic/genetics , Transcription, Genetic/immunology
6.
Mol Cell Biol ; 38(17)2018 09 01.
Article En | MEDLINE | ID: mdl-29915154

Zinc finger protein 521 (ZFP521), a DNA-binding protein containing 30 Krüppel-like zinc fingers, has been implicated in the differentiation of multiple cell types, including hematopoietic stem and progenitor cells (HSPC) and B lymphocytes. Here, we report a novel role for ZFP521 in regulating the earliest stages of hematopoiesis and lymphoid cell development via a cell-extrinsic mechanism. Mice with inactivated Zfp521 genes (Zfp521-/-) possess reduced frequencies and numbers of hematopoietic stem and progenitor cells, common lymphoid progenitors, and B and T cell precursors. Notably, ZFP521 deficiency changes bone marrow microenvironment cytokine levels and gene expression within resident HSPC, consistent with a skewing of hematopoiesis away from lymphopoiesis. These results advance our understanding of ZFP521's role in normal hematopoiesis, justifying further research to assess its potential as a target for cancer therapies.


Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Stem Cell Niche/physiology , Transcription Factors/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cytokines/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Lymphopoiesis/genetics , Lymphopoiesis/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myelopoiesis/genetics , Myelopoiesis/physiology , Protein Binding , Stem Cell Niche/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
7.
J Exp Med ; 215(7): 1947-1963, 2018 07 02.
Article En | MEDLINE | ID: mdl-29899037

To understand the developmental trajectories in early lymphocyte differentiation, we identified differentially expressed surface markers on lineage-negative lymphoid progenitors (LPs). Single-cell polymerase chain reaction experiments allowed us to link surface marker expression to that of lineage-associated transcription factors (TFs) and identify GFRA2 and BST1 as markers of early B cells. Functional analyses in vitro and in vivo as well as single-cell gene expression analyses supported that surface expression of these proteins defined distinct subpopulations that include cells from both the classical common LPs (CLPs) and Fraction A compartments. The formation of the GFRA2-expressing stages of development depended on the TF EBF1, critical both for the activation of stage-specific target genes and modulation of the epigenetic landscape. Our data show that consecutive expression of Ly6D, GFRA2, and BST1 defines a developmental trajectory linking the CLP to the CD19+ progenitor compartment.


B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Compartmentation , Lymphopoiesis , Stem Cells/cytology , ADP-ribosyl Cyclase/metabolism , Animals , Antigens, CD/metabolism , Antigens, Ly/metabolism , Bone Marrow/metabolism , Cell Lineage , Cell Membrane/metabolism , GPI-Linked Proteins/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Mice , Models, Biological
8.
Genes Dev ; 30(22): 2486-2499, 2016 11 15.
Article En | MEDLINE | ID: mdl-27913602

Even though leukemia is considered to be confined to one specific hematopoietic cell type, cases of acute leukemia of ambiguous lineage and patients relapsing in phenotypically altered disease suggest that a malignant state may be transferred between lineages. Because B-cell leukemia is associated with mutations in transcription factors of importance for stable preservation of lineage identity, we here investigated the potential lineage plasticity of leukemic cells. We report that primary pro-B leukemia cells from mice carrying heterozygous mutations in either or both the Pax5 and Ebf1 genes, commonly mutated in human leukemia, can be converted into T lineage leukemia cells. Even though the conversion process involved global changes in gene expression and lineage-restricted epigenetic reconfiguration, the malignant phenotype of the cells was preserved, enabling them to expand as T lineage leukemia cells in vivo. Furthermore, while the transformed pro-B cells displayed plasticity toward myeloid lineages, the converted cells failed to cause myeloid leukemia after transplantation. These data provide evidence that a malignant phenotype can be transferred between hematopoietic lineages. This has important implications for modern cancer medicine because lineage targeted treatment of leukemia patients can be predicted to provoke the emergence of phenotypically altered subclones, causing clinical relapse.


B-Lymphocytes/pathology , Cell Transformation, Neoplastic/genetics , Leukemia, Lymphoid/physiopathology , Animals , Cell Line , Cell Line, Tumor , Cell Lineage , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Leukemia, T-Cell/physiopathology , Mice , Mice, Inbred C57BL , Mutation , Myeloid Cells/pathology , Precursor Cells, B-Lymphoid/metabolism , Protein Binding , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction
9.
J Exp Med ; 212(7): 1109-23, 2015 Jun 29.
Article En | MEDLINE | ID: mdl-26056231

To investigate how transcription factor levels impact B-lymphocyte development, we generated mice carrying transheterozygous mutations in the Pax5 and Ebf1 genes. Whereas combined reduction of Pax5 and Ebf1 had minimal impact on the development of the earliest CD19(+) progenitors, these cells displayed an increased T cell potential in vivo and in vitro. The alteration in lineage fate depended on a Notch1-mediated conversion process, whereas no signs of de-differentiation could be detected. The differences in functional response to Notch signaling in Wt and Pax5(+/-)Ebf1(+/-) pro-B cells were reflected in the transcriptional response. Both genotypes responded by the generation of intracellular Notch1 and activation of a set of target genes, but only the Pax5(+/-)Ebf1(+/-) pro-B cells down-regulated genes central for the preservation of stable B cell identity. This report stresses the importance of the levels of transcription factor expression during lymphocyte development, and suggests that Pax5 and Ebf1 collaborate to modulate the transcriptional response to Notch signaling. This provides an insight on how transcription factors like Ebf1 and Pax5 preserve cellular identity during differentiation.


B-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Lineage/immunology , Gene Expression Regulation/immunology , PAX5 Transcription Factor/deficiency , T-Lymphocytes/cytology , Trans-Activators/deficiency , Animals , Antigens, CD19/immunology , B-Lymphocytes/immunology , Base Sequence , Blotting, Western , Chromatin Immunoprecipitation , DNA Primers/genetics , Flow Cytometry , Gene Expression Regulation/genetics , Loss of Heterozygosity , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation/genetics , PAX5 Transcription Factor/genetics , Real-Time Polymerase Chain Reaction , Receptors, Notch/metabolism , Sequence Analysis, RNA , Trans-Activators/genetics
10.
Exp Hematol ; 43(7): 554-64, 2015 Jul.
Article En | MEDLINE | ID: mdl-25931014

The phosphatidylinositol 3-kinase/AKT pathway is an integral component of signaling involved in the development of many cancers, including myeloid leukemias such as chronic myeloid leukemia and acute myeloid leukemia (AML). Increased AKT1 activity is frequently seen in AML patients, providing leukemic cells with growth and survival promoting signals. An important aspect of AKT1 function is its involvement in cellular metabolism and energy production. Under some circumstances, strong activation of AKT1 increases oxidative stress, which can cause apoptosis when cells progressively build up excess free radicals. This has been described in hematopoietic cells overexpressing activated AKT1; however, whether this is true in cells coexpressing other genetic events involved in leukemia is not known. This prompted us to investigate the effect of constitutively active AKT1 (myristoylated AKT1) in hematopoietic progenitor cells expressing constitutively active signal transducer and activator of transcription 5, Fms-related tyrosine kinase 3-internal tandem duplication, or antiapoptotic B-cell lymphoma 2. Surprisingly, myristoylated AKT1 was incompatible with proliferation driven by both signal transducer and activator of transcription 5 and Fms-related tyrosine kinase 3-internal tandem duplication, which triggered cell cycle block and apoptosis. Moreover, transplantable cells of B-cell lymphoma 2-transgenic mice were impaired in their engraftment ability to recipient mice when expressing hyperactivated AKT1. This was linked to AKT1-mediated proapoptotic functions and not to impairment in homing to the bone marrow. Although cells expressing hyperactivated AKT1 displayed higher levels of reactive oxygen species both in vitro and in vivo, the addition of the antioxidant N-acetyl-L-cysteine significantly reduced apoptosis. Taken together, the results indicate that constitutive AKT1 activity is incompatible with growth- and survival-promoting ability of other activated genes in AML.


Apoptosis/physiology , Hematopoietic Stem Cells/cytology , Leukemia, Myeloid, Acute/enzymology , Neoplasm Proteins/physiology , Proto-Oncogene Proteins c-akt/physiology , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Cycle , Cell Division , Cell Movement/drug effects , Enzyme Activation , Gene Expression Regulation, Leukemic , Genes, bcl-2 , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myristic Acid , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplastic Stem Cells/cytology , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Recombinant Fusion Proteins/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/physiology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/physiology
11.
Blood ; 125(26): 4052-9, 2015 Jun 25.
Article En | MEDLINE | ID: mdl-25838350

Early B-cell factor 1 (Ebf1) is a transcription factor with documented dose-dependent functions in normal and malignant B-lymphocyte development. To understand more about the roles of Ebf1 in malignant transformation, we investigated the impact of reduced functional Ebf1 dosage on mouse B-cell progenitors. Gene expression analysis suggested that Ebf1 was involved in the regulation of genes important for DNA repair and cell survival. Investigation of the DNA damage in steady state, as well as after induction of DNA damage by UV light, confirmed that pro-B cells lacking 1 functional allele of Ebf1 display signs of increased DNA damage. This correlated to reduced expression of DNA repair genes including Rad51, and chromatin immunoprecipitation data suggested that Rad51 is a direct target for Ebf1. Although reduced dosage of Ebf1 did not significantly increase tumor formation in mice, a dramatic increase in the frequency of pro-B cell leukemia was observed in mice with combined heterozygous mutations in the Ebf1 and Pax5 genes, revealing a synergistic effect of combined dose reduction of these proteins. Our data suggest that Ebf1 controls DNA repair in a dose-dependent manner providing a possible explanation to the frequent involvement of EBF1 gene loss in human leukemia.


Cell Transformation, Neoplastic/genetics , DNA Damage/genetics , PAX5 Transcription Factor/genetics , Precursor Cells, B-Lymphoid/metabolism , Trans-Activators/genetics , Animals , Blotting, Western , Chromatin Immunoprecipitation , Comet Assay , Flow Cytometry , Fluorescent Antibody Technique , Haploinsufficiency/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction
12.
J Biol Chem ; 288(46): 33449-61, 2013 Nov 15.
Article En | MEDLINE | ID: mdl-24078629

Transcription factor doses are of importance for normal and malignant B-lymphocyte development; however, the understanding of underlying mechanisms and functional consequences of reduced transcription factor levels is limited. We have analyzed progenitor and B-lineage compartments in mice carrying heterozygote mutations in the E2a, Ebf1, or Pax5 gene. Although lymphoid progenitors from Ebf1 or Pax5 heterozygote mice were specified and lineage-restricted in a manner comparable with Wt progenitors, this process was severely impaired in E2a heterozygote mutant mice. This defect was not significantly enhanced upon combined deletion of E2a with Ebf1 or Pax5. Analysis of the pre-B-cell compartment in Ebf1 heterozygote mice revealed a reduction in cell numbers. These cells expressed Pax5 and other B-lineage-associated genes, and global gene expression analysis suggested that the reduction of the pre-B-cell compartment was a result of impaired pre-B-cell expansion. This idea was supported by a reduction in IL2Rα-expressing late pre-B-cells as well as by cell cycle analysis and by the finding that the complexity of the VDJ rearrangement patterns was comparable in Wt and Ebf1(+/-) pre-B-cells, although the number of progenitors was reduced. Heterozygote deletion of Ebf1 resulted in impaired response to IL7 in vitro and reduced expression levels of pre-BCR on the cell surface, providing possible explanations for the observed stage-specific reduction in cellular expansion. Thus, transcription factor doses are critical for specification as well as expansion of B-lymphoid progenitors, providing increased insight into the molecular regulation of B-cell development.


Gene Dosage/immunology , Precursor Cells, B-Lymphoid/immunology , Trans-Activators/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Dosage/genetics , Gene Expression Regulation/immunology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7/genetics , Interleukin-7/immunology , Interleukin-7/metabolism , Mice , Mice, Mutant Strains , Mutation , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/immunology , PAX5 Transcription Factor/metabolism , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
13.
Proc Natl Acad Sci U S A ; 109(51): 21028-33, 2012 Dec 18.
Article En | MEDLINE | ID: mdl-23213261

Recent studies have identified a number of transcriptional regulators, including E2A, early B-cell factor 1 (EBF1), FOXO1, and paired box gene 5 (PAX5), that promote early B-cell development. However, how this ensemble of regulators mechanistically promotes B-cell fate remains poorly understood. Here we demonstrate that B-cell development in FOXO1-deficient mice is arrested in the common lymphoid progenitor (CLP) LY6D(+) cell stage. We demonstrate that this phenotype closely resembles the arrest in B-cell development observed in EBF1-deficient mice. Consistent with these observations, we find that the transcription signatures of FOXO1- and EBF1-deficient LY6D(+) progenitors are strikingly similar, indicating a common set of target genes. Furthermore, we found that depletion of EBF1 expression in LY6D(+) CLPs severely affects FOXO1 mRNA abundance, whereas depletion of FOXO1 activity in LY6D(+) CLPs ablates EBF1 transcript levels. We generated a global regulatory network from EBF1 and FOXO1 genome-wide transcription factor occupancy and transcription signatures derived from EBF1- and FOXO1-deficient CLPs. This analysis reveals that EBF1 and FOXO1 act in a positive feedback circuitry to promote and stabilize specification to the B-cell lineage.


B-Lymphocytes/immunology , Forkhead Transcription Factors/metabolism , Trans-Activators/physiology , Animals , B-Lymphocytes/cytology , Cell Lineage , Cell Separation , Enhancer Elements, Genetic , Feedback, Physiological , Flow Cytometry , Forkhead Box Protein O1 , Gene Expression Regulation , Luciferases/metabolism , Mice , Models, Biological , Models, Genetic , Transcription, Genetic
14.
Proc Natl Acad Sci U S A ; 109(39): 15871-6, 2012 Sep 25.
Article En | MEDLINE | ID: mdl-23019372

To better understand the process of B-lymphocyte lineage restriction, we have investigated molecular and functional properties in early B-lineage cells from Pax-5-deficient animals crossed to a B-lineage-restricted reporter mouse, allowing us to identify B-lineage-specified progenitors independently of conventional surface markers. Pax-5 deficiency resulted in a dramatic increase in the frequency of specified progenitor B-cells marked by expression of a λ5 (Igll1) promoter-controlled reporter gene. Gene expression analysis of ex vivo isolated progenitor cells revealed that Pax-5 deficiency has a minor impact on B-cell specification. However, single-cell in vitro differentiation analysis of ex vivo isolated cells revealed that specified B-lineage progenitors still displayed a high degree of plasticity for development into NK or T lineage cells. In contrast, we were unable to detect any major changes in myeloid lineage potential in specified Pax-5-deficient cells. By comparison of gene expression patterns in ex vivo isolated Pax-5- and Ebf-1-deficient progenitors, it was possible to identify a set of B-cell-restricted genes dependent on Ebf-1 but not Pax-5, supporting the idea that B-cell specification and commitment is controlled by distinct regulatory networks.


B-Lymphocytes/immunology , Cell Differentiation/physiology , Gene Expression Regulation/immunology , Animals , B-Lymphocytes/cytology , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Mice , Mice, Transgenic , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Trans-Activators/genetics , Trans-Activators/immunology
15.
Semin Immunol ; 23(5): 335-40, 2011 Oct.
Article En | MEDLINE | ID: mdl-21944938

Even though B-lymphocyte development is one of the best understood models for cell differentiation in the hematopoetic system, recent advances in cell sorting and functional genomics has increased this understanding further. This has suggested that already early lymphoid primed multipotent progenitor cells (LMPPs) express low levels of lymphoid restricted transcripts. The expression of these genes becomes more pronounced when cells enter the FLT-3/IL-7 receptor positive common lymphoid progenitor (CLP) stage. However, the expression of B-lineage specific genes is limited to a B-cell restricted Ly6D surface positive subpopulation of the CLP compartment. The gene expression patterns also reflect differences in lineage potential and while Ly6D negative FLT-3/IL-7 receptor positive cells represents true CLPs with an ability to generate B/T and NK cells, the Ly6D positive cells lack NK cell potential and display a reduced T-cell potential in vivo. These recent findings suggest that the CLP compartment is highly heterogenous and that the point of no return in B-cell development may occur already in B220(-)CD19(-) cells. These findings have allowed for a better understanding of the interplay between transcription factors like EBF-1, PAX-5 and E47, all known as crucial for normal B-cell development. In this review, we aim to provide a comprehensive overview of B-cell fate specification and commitment based on the recent advances in the understanding of molecular networks as well as functional properties of early progenitor populations.


B-Lymphocytes , Lymphoid Progenitor Cells/cytology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation , Cell Lineage , Hematopoietic Stem Cells/cytology , Humans , Lymphoid Progenitor Cells/immunology
16.
Blood ; 118(5): 1283-90, 2011 Aug 04.
Article En | MEDLINE | ID: mdl-21652681

Deficiencies in the IL-7 signaling pathway result in severe disruptions of lymphoid development in adult mice. To understand more about how IL-7 deficiency impacts early lymphoid development, we have investigated lineage restriction events within the common lymphoid progenitor (CLP) compartment in IL-7 knockout mice. This revealed that although IL-7 deficiency had a minor impact on the development of LY6D(-) multipotent CLPs, the formation of the lineage restricted LY6D(+) CLP population was dramatically reduced. This was reflected in a low-level transcription of B-lineage genes as well as in a loss of functional B-cell commitment. The few Ly6D(+) CLPs developed in the absence of IL-7 displayed increased lineage plasticity and low expression of Ebf-1. Absence of Ebf-1 could be linked to increased plasticity because even though Ly6D(+) cells develop in Ebf-1-deficient mice, these cells retain both natural killer and dendritic cell potential. This reveals that IL-7 is essential for normal development of Ly6D(+) CLPs and that Ebf-1 is crucial for lineage restriction in early lymphoid progenitors.


Cell Lineage/genetics , Interleukin-7/physiology , Lymphoid Progenitor Cells/physiology , Trans-Activators/physiology , Animals , Antigens, Ly/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/physiology , Cell Differentiation/genetics , Cells, Cultured , GPI-Linked Proteins/metabolism , Gene Expression Profiling , Interleukin-7/genetics , Killer Cells, Natural/metabolism , Killer Cells, Natural/physiology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , T-Lymphocytes/metabolism , T-Lymphocytes/physiology , Trans-Activators/genetics , Trans-Activators/metabolism
17.
J Biol Chem ; 285(47): 36275-84, 2010 Nov 19.
Article En | MEDLINE | ID: mdl-20829349

The development of lymphoid cells from bone marrow progenitors is dictated by interplay between internal cues such as transcription factors and external signals like the cytokines Flt-3 ligand and Il-7. These proteins are both of large importance for normal lymphoid development; however, it is unclear if they act in direct synergy to expand a transient Il-7R(+)Flt-3(+) population or if the collaboration is created through sequential activities. We report here that Flt-3L and Il-7 synergistically stimulated the expansion of primary Il-7R(+)Flt-3(+) progenitor cells and a hematopoietic progenitor cell line ectopically expressing the receptors. The stimulation resulted in a reduced expression of pro-apoptotic genes and also mediated survival of primary progenitor cells in vitro. However, functional analysis of single cells suggested that the anti-apoptotic effect was additive indicating that the synergy observed mainly depends on stimulation of proliferation. Analysis of downstream signaling events suggested that although Il-7 induced Stat-5 phosphorylation, Flt-3L caused activation of the ERK and AKT signaling pathways. Flt-3L could also drive proliferation in synergy with ectopically expressed constitutively active Stat-5. This synergy could be inhibited with either receptor tyrosine kinase or MAPK inhibitors suggesting that Flt-3L and Il-7 act in synergy by activation of independent signaling pathways to expand early hematopoietic progenitors.


Cell Proliferation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Interleukin-7/pharmacology , STAT5 Transcription Factor/metabolism , Signal Transduction , fms-Like Tyrosine Kinase 3/metabolism , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Drug Synergism , Flow Cytometry , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/genetics , fms-Like Tyrosine Kinase 3/genetics
...