Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Leukemia ; 30(6): 1375-87, 2016 06.
Article in English | MEDLINE | ID: mdl-26932576

ABSTRACT

The Ets family transcription factor PU.1 and the interferon regulatory factor (IRF)4 and IRF8 regulate gene expression by binding to composite DNA sequences known as Ets/interferon consensus elements. Although all three factors are expressed from the onset of B-cell development, single deficiency of these factors in B-cell progenitors only mildly impacts on bone marrow B lymphopoiesis. Here we tested whether PU.1 cooperates with IRF factors in regulating early B-cell development. Lack of PU.1 and IRF4 resulted in a partial block in development the pre-B-cell stage. The combined deletion of PU.1 and IRF8 reduced recirculating B-cell numbers. Strikingly, all PU.1/IRF4 and ~50% of PU.1/IRF8 double deficient mice developed pre-B-cell acute lymphoblastic leukemia (B-ALL) associated with reduced expression of the established B-lineage tumor suppressor genes, Ikaros and Spi-B. These genes are directly regulated by PU.1/IRF4/IRF8, and restoration of Ikaros or Spi-B expression inhibited leukemic cell growth. In summary, we demonstrate that PU.1, IRF4 and IRF8 cooperate to regulate early B-cell development and to prevent pre-B-ALL formation.


Subject(s)
Interferon Regulatory Factors/physiology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Animals , B-Lymphocytes/cytology , Gene Expression Regulation , Interferon Regulatory Factors/genetics , Lymphopoiesis , Mice , Mice, Knockout , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/prevention & control , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics
2.
Leukemia ; 29(6): 1301-11, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25655195

ABSTRACT

Activating NOTCH1 mutations occur in ~60% of human T-cell acute lymphoblastic leukemias (T-ALLs), and mutations disrupting the transcription factor IKZF1 (IKAROS) occur in ~5% of cases. To investigate the regulatory interplay between these driver genes, we have used a novel transgenic RNA interference mouse model to produce primary T-ALLs driven by reversible Ikaros knockdown. Restoring endogenous Ikaros expression in established T-ALL in vivo acutely represses Notch1 and its oncogenic target genes including Myc, and in multiple primary leukemias causes disease regression. In contrast, leukemias expressing high levels of endogenous or engineered forms of activated intracellular Notch1 (ICN1) resembling those found in human T-ALL rapidly relapse following Ikaros restoration, indicating that ICN1 functionally antagonizes Ikaros in established disease. Furthermore, we find that IKAROS mRNA expression is significantly reduced in a cohort of primary human T-ALL patient samples with activating NOTCH1/FBXW7 mutations, but is upregulated upon acute inhibition of aberrant NOTCH signaling across a panel of human T-ALL cell lines. These results demonstrate for the first time that aberrant NOTCH activity compromises IKAROS function in mouse and human T-ALL, and provide a potential explanation for the relative infrequency of IKAROS gene mutations in human T-ALL.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Ikaros Transcription Factor/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Notch/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Biomarkers, Tumor/genetics , Blotting, Western , Cell Cycle Proteins/genetics , Chromatin Immunoprecipitation , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Flow Cytometry , High-Throughput Nucleotide Sequencing , Humans , Ikaros Transcription Factor/antagonists & inhibitors , Ikaros Transcription Factor/genetics , Mice , Mice, Transgenic , Mutation/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Notch/genetics , Signal Transduction , Ubiquitin-Protein Ligases/genetics
3.
Clin Exp Immunol ; 167(2): 282-95, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22236005

ABSTRACT

The immunomodulatory effects of probiotics were assessed following exposure of normal peripheral blood mononuclear cells (PBMC), cord blood cells and the spleen-derived monocyte/macrophage cell line CRL-9850 to Lactobacillus acidophilus LAVRI-A1, Lb. rhamnosus GG, exopolysaccharides (EPS)-producing Streptococcus thermophilus St1275, Bifidobacteriun longum BL536, B. lactis B94 and Escherichia coli TG1 strains. The production of a panel of pro- and anti-inflammatory cytokines by PBMC following bacterial stimulation was measured, using live, heat-killed or mock gastrointestinal tract (GIT)-exposed bacteria, and results show that (i) all bacterial strains investigated induced significant secretion of pro- and anti-inflammatory cytokines from PBMC-derived monocytes/macrophages; and (ii) cytokine levels increased relative to the expansion of bacterial cell numbers over time for cells exposed to live cultures. Bifidobacteria and S. thermophilus stimulated significant concentrations of transforming growth factor (TGF)-ß, an interleukin necessary for the differentiation of regulatory T cells (T(reg) )/T helper type 17 (Th17) cells and, as such, the study further examined the induction of Th17 and T(reg) cells after PBMC exposure to selected bacteria for 96 h. Data show a significant increase in the numbers of both cell types in the exposed populations, measured by cell surface marker expression and by cytokine production. Probiotics have been shown to induce cytokines from a range of immune cells following ingestion of these organisms. These studies suggest that probiotics' interaction with immune-competent cells produces a cytokine milieu, exerting immunomodulatory effects on local effector cells, as well as potently inducing differentiation of Th17 and T(reg) cells.


Subject(s)
Cytokines/metabolism , Monocytes/immunology , Probiotics/pharmacology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Bacterial Load , Bifidobacterium/immunology , Bile , Cell Line , Escherichia coli/immunology , Fetal Blood/cytology , Gastric Acid , Humans , Inflammation , Intestines/microbiology , Lactobacillus acidophilus/immunology , Lacticaseibacillus rhamnosus/immunology , Spleen/cytology , Stomach/microbiology , Streptococcus thermophilus/immunology , Th17 Cells/metabolism
4.
Article in English | MEDLINE | ID: mdl-19022754

ABSTRACT

One critical issue for cancer biology is the nature of the cells that drive the inexorable growth of malignant tumors. Reports that only rare cell populations within human leukemias seeded leukemia in mice stimulated the now widely embraced hypothesis that only such "cancer stem cells" maintain all tumor growth. However, the mouse microenvironment might instead fail to support the dominant human tumor cell populations. Indeed, on syngeneic transplantation of mouse lymphomas and leukemias, we and other investigators have found that a substantial proportion (>10%) of their cells drive tumor growth. Thus, dominant clones rather than rare cancer stem cells appear to sustain many tumors. Another issue is the role of cell survival in tumorigenesis. Because tumor development can be promoted by the overexpression of prosurvival genes such as bcl-2, we are exploring the role of endogenous Bcl-2-like proteins in lymphomagenesis. The absence of endogenous Bcl-2 in mice expressing an Emu-myc transgene reduced mature B-cell numbers and enhanced their apoptosis, but unexpectedly, lymphoma development was undiminished or even delayed. This suggests that these tumors originate in an earlier cell type, such as the pro-B or pre-B cell, and that the nascent neoplastic clones do not require Bcl-2 but may instead be protected by a Bcl-2 relative.


Subject(s)
Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Apoptosis , B-Lymphocytes/pathology , Cell Proliferation , Cell Survival , Genes, bcl-2 , Genes, myc , Humans , Leukemia/etiology , Leukemia/pathology , Lymphoma/etiology , Lymphoma/pathology , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Neoplasm Transplantation , Neoplasms/etiology , Transplantation, Heterologous
5.
Genesis ; 30(3): 110-3, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11477685

ABSTRACT

Morpholino (MO) based inhibition of translational initiation represents an attractive methodology to eliminate gene function during Xenopus development (Heasman et al., 2000). However, the degree to which a given target protein can be eliminated and the longevity of this effect during embryogenesis has not been documented. To examine the efficacy of MOs, we have used transgenic Xenopus lines that harbour known numbers of integrations of a GFP reporter under the control of the ubiquitous and highly expressed CMV promoter (Fig. 1a). In addition we have investigated the longevity of the inhibitory effect by using transgenic lines expressing GFP specifically in the lens of tadpoles. These transgenic lines represent the ideal control for the technique as the promoters are highly expressed and GFP can be easily detected by fluorescence and immunoblotting. Moreover, as GFP has no function in development, the levels of inhibition can be tested in an otherwise normal individual. Here we report that MOs are able to efficiently and specifically inhibit the translation of GFP in transgenic lines from Xenopus laevis and Xenopus tropicalis and the inhibitory effect is long-lived, lasting into the tadpole stages. genesis 30:110--113, 2001.


Subject(s)
Morpholines/metabolism , Oligonucleotides, Antisense/metabolism , Protein Biosynthesis , Xenopus laevis/growth & development , Xenopus laevis/genetics , Xenopus/growth & development , Xenopus/genetics , Animals , Animals, Genetically Modified , Crystallins/genetics , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Female , Gene Expression Regulation, Developmental , Genes, Reporter/genetics , Green Fluorescent Proteins , Larva/genetics , Larva/growth & development , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Oligonucleotides, Antisense/genetics , Promoter Regions, Genetic/genetics , Xenopus/embryology , Xenopus laevis/embryology
6.
Int Rev Immunol ; 20(1): 65-82, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11342298

ABSTRACT

Despite being one of the most intensively studied cell types, the molecular basis of B cell specification is largely unknown. The Pax5 gene encoding the transcription factor BSAP is required for progression of B-lymphopoiesis beyond the pro-B cell stage. Pax5-deficient pro-B cells are, however, not yet committed to the B-lymphoid lineage, but instead have a broad lymphomyeloid developmental potential. Pax5 appears to mediate B-lineage commitment by repressing the transcription of non-B-lymphoid genes and by simultaneously activating the expression of B-lineage-specific genes. Pax5 thus functions both as a transcriptional repressor and activator, depending on its interactions with corepressors of the Groucho protein family or with positive regulators such as the TATA-binding protein. Once committed to the B-lineage, B cells require Pax5 function to maintain their B-lymphoid identity throughout B cell development.


Subject(s)
B-Lymphocyte Subsets/cytology , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Proteins/physiology , Animals , Antigens, CD19/biosynthesis , Antigens, CD19/genetics , Cell Differentiation , Cell Lineage , Cells, Cultured , Coculture Techniques , DNA-Binding Proteins/physiology , Gene Rearrangement, B-Lymphocyte , Genes, Immunoglobulin , Genes, myc , Humans , Interleukin-7/physiology , Mice , Mice, Knockout , Models, Biological , PAX5 Transcription Factor , Proteins/genetics , Repressor Proteins/physiology , Stromal Cells/cytology , Transcription Factor TFIID , Transcription Factors/physiology , Transcription Factors, TFII/metabolism , Transcription, Genetic
7.
Genes Dev ; 15(9): 1152-66, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11331610

ABSTRACT

Signal transduction through the FGF receptor is essential for the specification of the vertebrate body plan. Blocking the FGF pathway in early Xenopus embryos inhibits mesoderm induction and results in truncation of the anterior-posterior axis. The Drosophila gene sprouty encodes an antagonist of FGF signaling, which is transcriptionally induced by the pathway, but whose molecular functions are poorly characterized. We have cloned Xenopus sprouty2 and show that it is expressed in a similar pattern to known FGFs and is dependent on the FGF/Ras/MAPK pathway for its expression. Overexpression of Xsprouty2 in both embryos and explant assays results in the inhibition of the cell movements of convergent extension. Although blocking FGF/Ras/MAPK signaling leads to an inhibition of mesodermal gene expression, these markers are unaffected by Xsprouty2, indicating that mesoderm induction and patterning occurs normally in these embryos. Finally, using Xenopus oocytes we show that Xsprouty2 is an intracellular antagonist of FGF-dependent calcium signaling. These results provide evidence for at least two distinct FGF-dependent signal transduction pathways: a Sprouty-insensitive Ras/MAPK pathway required for the transcription of most mesodermal genes, and a Sprouty-sensitive pathway required for coordination of cellular morphogenesis.


Subject(s)
Body Patterning , Embryonic Induction , Fibroblast Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Xenopus laevis/embryology , Amino Acid Sequence , Animals , Calcium , Embryo, Nonmammalian , Gastrula , Gene Expression Regulation, Developmental , Mesoderm , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/genetics , Phosphorylation , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Xenopus laevis/genetics
8.
Immunol Rev ; 175: 104-11, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10933595

ABSTRACT

During B-lymphocyte development in mouse fetal liver and bone marrow, a pre-B I cell stage is reached in which the cells express B-lineage-specific genes, such as CD19, Ig alpha and Igbeta and VpreB and lambda5, which encode the surrogate light (SL) chain. In these pre-B I cells both alleles of the immunoglobulin heavy (IgH) chain locus are D(H)J(H) rearranged. Transplantation of pre-B I cells from wild-type (e.g. C57Bl/6) mice in histocompatible RAG-deficient hosts leads to long-term reconstitution of some of the mature B-cell compartments and to the establishment of normal IgM levels, a third of the normal serum IgA levels, and IgG levels below the detection limit. Neither T-lineage nor myeloid cells of donor origin can be detected in the transplanted hosts, indicating that the pre-B I cells are committed to B-lineage differentiation. Consequently, the B-cell-reconstituted hosts respond to T-cell-independent antigens but not to T-cell-dependent antigens. Responses to T-cell-dependent antigens can be restored in the pre-B I-cell-transplanted, RAG-deficient hosts by the concomitant transplantation of mature CD4+ T cells. The transplanted wild-type pre-B I cells do not home back to the bone marrow and become undetectable shortly after transplantation. B-lymphocyte development in Pax-5-deficient mice becomes arrested at the transition of pre-B I to pre-B II cells i.e. at the stage when V(H) to D(H)J(H) rearrangements occur and when the pre-B-cell receptor, complete with muH chains and SL chains, is normally formed. T-lineage and myeloid cell development in these mice is normal. Pre-B I cells of Pax-5-deficient mice have a wild-type pre-B I-cell-like phenotype: while they do not express Pax-5-controlled CD19 gene, and express Ig alpha to a lesser extent, they express Igbeta, VpreB and lambda5, and proliferate normally in vitro on stromal cells in the presence of interleukin (IL)-7. Clones of these pre-B I cells carry characteristic D(H)J(H) rearrangements on both IgH chain alleles. However, removal of IL-7 from the tissue cultures, unlike wild-type pre-B I cells, does not induce B-cell differentiation to surface IgM-expressing B cells, but induces macrophage differentiation. This differentiation into macrophages requires either the presence of stromal cells or addition of macrophage colony-stimulating factor (M-CSF). Addition of M-CSF followed by granulocyte-macrophage colony-stimulating factor induces the differentiation to MHC class II-expressing, antigen-presenting dendritic cells. In vitro differentiation to granulocytes and osteoclasts can also be observed in the presence of the appropriate cytokines. Moreover, transplantation of Pax-5-deficient pre-B I clones into RAG-deficient hosts, while not allowing B-cell differentiation, leads to the full reconstitution of the thymus with all stages of CD4-CD8- and CD4+CD8+ thymocytes, to normal positive and negative selection of thymocytes in the thymus, and to the development of normal, reactive mature CD4+ and CD8+ T-cell compartments in the peripheral lymphoid tissues, all carrying the clone-specific D(H)J(H) rearrangements. On the other hand, Ig alpha, Igbeta, VpreB and lambda5 are turned off in the thymocytes, demonstrating that the expression of these genes does not commit cells irreversibly to the B lineage. Further more, Pax-5-deficient pre-B I cells are long-term reconstituting cells. They home back to the bone marrow of the RAG-deficient host, can be reisolated and regrown in tissue culture, and can be retransplanted into a secondary RAG-deficient host. This again develops thymocytes and mature T cells and allows the transplanted clonal pre-B I cells to home to the bone marrow.


Subject(s)
B-Lymphocytes/immunology , Transcription Factors , Animals , Antigens/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , B-Lymphocytes/transplantation , Cell Differentiation , Cell Lineage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Deletion , Gene Rearrangement, B-Lymphocyte , Genes, Immunoglobulin , Hematopoietic Stem Cells/immunology , Immunocompromised Host , Mice , Models, Biological , Myeloid Cells/immunology , Nuclear Proteins/genetics , Nuclear Proteins/physiology , PAX5 Transcription Factor , T-Lymphocytes/immunology
9.
Curr Opin Immunol ; 12(2): 151-8, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10712946

ABSTRACT

The mechanisms controlling the commitment of hematopoietic progenitor cells to the lymphoid lineages are still mostly unknown. Recent findings indicate that the earliest phase of B cell development may proceed in two steps. At the onset of B-lymphopoiesis, the transcription factors E2A and EBF coordinately activate the B-cell-specific gene expression program. Subsequently, Pax5 appears to repress the promiscuous transcription of lineage-inappropriate genes and thus commits progenitor cells to the B-lymphoid pathway by suppressing alternative cell fates. B-lineage commitment by Pax5 seems to occur in a stochastic manner in the bone marrow, as indicated by the random activation of only one of the two Pax5 alleles in early pro-B cells. In contrast, loss- and gain-of-function analyses have implicated the Notch1 receptor in the specification of the T cell fate, which may thus be controlled by instructive signals in the thymus.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Hematopoiesis , Hematopoietic Stem Cells/cytology , Lymphocyte Subsets/cytology , Nuclear Proteins/physiology , Receptors, Cell Surface , Trans-Activators/physiology , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Cell Lineage , Cell Survival , DNA-Binding Proteins/genetics , Helix-Loop-Helix Motifs/genetics , Helix-Loop-Helix Motifs/physiology , Hematopoiesis/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Nuclear Proteins/genetics , PAX5 Transcription Factor , Phenotype , Receptor, Notch1 , Signal Transduction , Stochastic Processes , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic
10.
Nature ; 401(6753): 556-62, 1999 Oct 07.
Article in English | MEDLINE | ID: mdl-10524622

ABSTRACT

The Pax5 gene encoding the B-cell-specific activator protein (BSAP) is expressed within the haematopoietic system exclusively in the B-lymphoid lineage, where it is required in vivo for progression beyond the pro-B-cell stage. However, Pax5 is not essential for in vitro propagation of pro-B cells in the presence of interleukin-7 and stromal cells. Here we show that pro-B cells lacking Pax5 are also incapable of in vitro B-cell differentiation unless Pax5 expression is restored by retroviral transduction. Pax5-/- pro-B cells are not restricted in their lineage fate, as stimulation with appropriate cytokines induces them to differentiate into functional macrophages, osteoclasts, dendritic cells, granulocytes and natural killer cells. As expected for a clonogenic haematopoietic progenitor with lymphomyeloid developmental potential, the Pax5-/- pro-B cell expresses genes of different lineage-affiliated programmes, and restoration of Pax5 activity represses this lineage-promiscuous transcription. Pax5 therefore plays an essential role in B-lineage commitment by suppressing alternative lineage choices.


Subject(s)
B-Lymphocytes/cytology , DNA-Binding Proteins/physiology , Leukopoiesis , Nuclear Proteins/physiology , Transcription Factors/physiology , Animals , Bone Marrow Cells/cytology , Cell Lineage , Cells, Cultured , DNA-Binding Proteins/genetics , Granulocytes/cytology , Hematopoietic Stem Cells/cytology , Killer Cells, Natural/cytology , Mice , Mice, Inbred C57BL , Monocytes/cytology , Nuclear Proteins/genetics , PAX5 Transcription Factor , Transcription Factors/genetics , Transcription, Genetic
11.
Nature ; 401(6753): 603-6, 1999 Oct 07.
Article in English | MEDLINE | ID: mdl-10524629

ABSTRACT

The mechanisms controlling the commitment of haematopoietic progenitors to the B-lymphoid lineage are poorly understood. The observations that mice deficient in E2A and EBF lack B-lineage cells have implicated these two transcription factors in the commitment process. Moreover, the expression of genes encoding components of the rearrangement machinery (RAG1, RAG2, TdT) or pre-B-cell receptor (lambda5, VpreB, Igalpha, Igbeta) has been considered to indicate B-lineage commitment. All these genes including E2A and EBF are expressed in pro-B cells lacking the transcription factor Pax5. Here we show that cloned Pax5-deficient pro-B cells transferred into RAG2-deficient mice provide long-term reconstitution of the thymus and give rise to mature T cells expressing alpha/beta-T-cell receptors. The bone marrow of these mice contains a population of cells of Pax5-/- origin with the same phenotype as the donor pro-B cells. When transferred into secondary recipients, these pro-B cells again home to the bone marrow and reconstitute the thymus. Hence, B-lineage commitment is determined neither by immunoglobulin DJ rearrangement nor by the expression of E2A, EBF, lambda5, VpreB, Igalpha and Igbeta. Instead, our data implicate Pax5 in the control of B-lineage commitment.


Subject(s)
B-Lymphocytes/physiology , DNA-Binding Proteins/physiology , Hematopoietic Stem Cells/physiology , Leukopoiesis , Nuclear Proteins/physiology , T-Lymphocytes/physiology , Transcription Factors , Animals , Bone Marrow Cells/physiology , Cell Line , Clone Cells , DNA-Binding Proteins/genetics , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , PAX5 Transcription Factor , Thymus Gland/cytology
12.
Biol Chem ; 380(6): 601-11, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10430025

ABSTRACT

It is generally assumed that most mammalian genes are transcribed from both alleles. Hence, the diploid state of the genome offers the advantage that a loss-of-function mutation in one allele can be compensated for by the remaining wild-type allele of the same gene. Indeed, the vast majority of human disease syndromes and engineered mutations in the mouse genome are recessive, indicating that recessiveness is the 'default' state. However, a minority of genes are semi-dominant, as heterozygous loss-of-function mutation in these genes leads to phenotypic abnormalities. This condition, known as haploinsufficiency, has been described for five of the nine mammalian Pax genes, which are associated with mouse developmental mutants and human disease syndromes. Recently we have reported that the Pax5 gene is subject to allele-specific regulation during B cell development. Pax5 is predominantly transcribed from only one of its two alleles in early B-lymphoid progenitors and mature B cells, while it transiently switches to a biallelic mode of transcription in pre-B and immature B cells. As a consequence, B-lymphoid tissues are mosaic with regard to the transcribed allele, and heterozygous mutation of Pax5 therefore results in deletion of B lymphocytes expressing only the mutant allele. The allele-specific regulation of Pax5 raises the intriguing possibility that monoallelic expression may also be the mechanism causing the haploinsufficiency of other Pax genes. In this review, we discuss different models accounting for the haploinsufficiency of mammalian Pax genes, provide further evidence in support of the allele-specific regulation of Pax5 and discuss the implication of these findings in the context of the recent literature describing the stochastic and monoallelic activation of other hematopoietic genes.


Subject(s)
Alleles , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Animals , B-Lymphocytes/cytology , Cell Lineage , DNA-Binding Proteins/metabolism , Haplotypes , Heterozygote , Humans , In Situ Hybridization, Fluorescence/methods , Mice , Models, Genetic , Nuclear Proteins/metabolism , PAX5 Transcription Factor , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Nat Genet ; 21(4): 390-5, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10192389

ABSTRACT

The developmental control genes of the Pax family are frequently associated with mouse mutants and human disease syndromes. The function of these transcription factors is sensitive to gene dosage, as mutation of one allele or a modest increase in gene number results in phenotypic abnormalities. Pax5 has an important role in B-cell and midbrain development. By following the expression of individual Pax5 alleles at the single-cell level, we demonstrate here that Pax5 is subject to allele-specific regulation during B-lymphopoiesis. Pax5 is predominantly transcribed from only one allele in early progenitors and mature B cells, whereas it switches to a biallelic transcription mode in immature B cells. The allele-specific regulation of Pax5 is stochastic, reversible, independent of parental origin and correlates with synchronous replication, in contrast with imprinted and other monoallelically expressed genes. As a consequence, B-lymphoid tissues are mosaics with respect to the transcribed Pax5 allele, and thus mutation of one allele in heterozygous mice results in deletion of the cell population expressing the mutant allele due to loss of Pax5 function at the single-cell level. Similar allele-specific regulation may be a common mechanism causing the haploinsufficiency and frequent association of other Pax genes with human disease.


Subject(s)
B-Lymphocytes/physiology , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Nuclear Proteins/genetics , Alleles , Animals , Bone Marrow/metabolism , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Ikaros Transcription Factor , In Situ Hybridization, Fluorescence , Liver/cytology , Liver/embryology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Mutant Strains , Mutation , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , PAX5 Transcription Factor , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology , Spleen/embryology , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Exp Med ; 188(4): 735-44, 1998 Aug 17.
Article in English | MEDLINE | ID: mdl-9705955

ABSTRACT

The formation of the pre-B cell receptor (BCR) corresponds to an important checkpoint in B cell development that selects pro-B (pre-BI) cells expressing a functionally rearranged immunoglobulin mu (Igmu) heavy chain protein to undergo the transition to the pre-B (pre-BII) cell stage. The pre-BCR contains, in addition to Igmu, the surrogate light chains lambda5 and VpreB and the signal transducing proteins Igalpha and Igbeta. The absence of one of these pre-BCR components is known to arrest B cell development at the pre-BI cell stage. Disruption of the Pax5 gene, which codes for the B cell-specific activator protein (BSAP), also blocks adult B lymphopoiesis at the pre-BI cell stage. Moreover, expression of the mb-1 (Igalpha) gene and VH-to-DHJH recombination at the IgH locus are reduced in Pax5-deficient B lymphocytes approximately 10- and approximately 50-fold, respectively. Here we demonstrate that complementation of these deficiencies in pre-BCR components by expression of functionally rearranged Ig mu and chimeric Igmu-Igbeta transgenes fails to advance B cell development to the pre-BII cell stage in Pax5 (-/-) mice in contrast to RAG2 (-/-) mice. Furthermore, the pre-BCR is stably expressed on cultured pre-BI cells from Igmu transgenic, Pax5-deficient bone marrow, but is unable to elicit its normal signaling responses. In addition, the early developmental block is unlikely to be caused by the absence of a survival signal, as it could not be rescued by expression of a bcl2 transgene in Pax5-deficient pre-BI cells. Together, these data demonstrate that the absence of Pax5 arrests adult B lymphopoiesis at an early developmental stage that is unresponsive to pre-BCR signaling.


Subject(s)
B-Lymphocytes/physiology , DNA-Binding Proteins/physiology , Hematopoietic Stem Cells/physiology , Leukopoiesis/physiology , Nuclear Proteins/physiology , Receptors, Antigen, B-Cell/physiology , Transcription Factors , Animals , B-Lymphocytes/cytology , Cells, Cultured , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/cytology , Humans , Immunoglobulin mu-Chains/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/genetics , PAX5 Transcription Factor , Signal Transduction
16.
Semin Immunol ; 10(2): 133-42, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9618759

ABSTRACT

Pax-5 codes for the transcription factor BSAP which is expressed throughout B cell development except in terminally differentiated plasma cells. Gene targeting experiments in the mouse revealed a differential dependency of fetal and adult B-lymphopoiesis on this transcription factor. BSAP is required for B-lineage commitment in the fetal liver and for progression beyond an early pro-B cell stage in adult bone marrow. The characterization of Pax-5-deficient pro-B cells demonstrated an important role of BSAP in the regulation of the CD19, mb-1 (Ig alpha) and N-myc genes as well as in the developmental pathway controlling VH-to-DHJH recombination at the immunoglobulin heavy-chain (IgH) locus. The human PAX-5 gene was recently shown to participate together with the IgH locus in the chromosomal translocation t(9;14)(p13;q32). This translocation is characteristic of a small subset of non-Hodgkin lymphomas exhibiting plasmacytoid differentiation. The translocated PAX-5 gene is deregulated by the insertion of IgH regulatory elements into its 5' region, which may contribute to tumorigenesis by interfering with the shut-down of PAX-5 transcription and thus with the completion of plasma cell differentiation.


Subject(s)
B-Lymphocytes/cytology , DNA-Binding Proteins/physiology , Leukopoiesis/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Adult , Animals , DNA-Binding Proteins/genetics , Down-Regulation , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Gene Rearrangement, B-Lymphocyte , Genes, Immunoglobulin , Humans , Immunoglobulin Heavy Chains/genetics , Liver/embryology , Lymphoma, Non-Hodgkin/genetics , Mice , Mutation , Nuclear Proteins/genetics , PAX5 Transcription Factor , Transcription Factors/genetics , Translocation, Genetic
17.
EMBO J ; 17(8): 2319-33, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9545244

ABSTRACT

The Pax-5 gene codes for the transcription factor BSAP which is essential for the progression of adult B lymphopoiesis beyond an early progenitor (pre-BI) cell stage. Although several genes have been proposed to be regulated by BSAP, CD19 is to date the only target gene which has been genetically confirmed to depend on this transcription factor for its expression. We have now taken advantage of cultured pre-BI cells of wild-type and Pax-5 mutant bone marrow to screen a large panel of B lymphoid genes for additional BSAP target genes. Four differentially expressed genes were shown to be under the direct control of BSAP, as their expression was rapidly regulated in Pax-5-deficient pre-BI cells by a hormone-inducible BSAP-estrogen receptor fusion protein. The genes coding for the B-cell receptor component Ig-alpha (mb-1) and the transcription factors N-myc and LEF-1 are positively regulated by BSAP, while the gene coding for the cell surface protein PD-1 is efficiently repressed. Distinct regulatory mechanisms of BSAP were revealed by reconstituting Pax-5-deficient pre-BI cells with full-length BSAP or a truncated form containing only the paired domain. IL-7 signalling was able to efficiently induce the N-myc gene only in the presence of full-length BSAP, while complete restoration of CD19 synthesis was critically dependent on the BSAP protein concentration. In contrast, the expression of the mb-1 and LEF-1 genes was already reconstituted by the paired domain polypeptide lacking any transactivation function, suggesting that the DNA-binding domain of BSAP is sufficient to recruit other transcription factors to the regulatory regions of these two genes. In conclusion, these loss- and gain-of-function experiments demonstrate that BSAP regulates four newly identified target genes as a transcriptional activator, repressor or docking protein depending on the specific regulatory sequence context.


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Antigens, CD/genetics , Antigens, CD19/genetics , Antigens, Surface/genetics , Apoptosis Regulatory Proteins , B-Lymphocytes/chemistry , CD79 Antigens , Cell Membrane/metabolism , Gene Expression Regulation , Genes, myc , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Interleukin-7/pharmacology , Lymphoid Enhancer-Binding Factor 1 , Mice , Mutagenesis , PAX5 Transcription Factor , Programmed Cell Death 1 Receptor , Receptors, Antigen, B-Cell/genetics
18.
Genes Dev ; 11(4): 476-91, 1997 Feb 15.
Article in English | MEDLINE | ID: mdl-9042861

ABSTRACT

The Pax5 gene coding for the transcription factor BSAP has an essential role in B lymphopoiesis and midbrain development. Here we present a detailed analysis of the B-cell phenotype of Pax5 mutant mice that revealed a differential dependency of fetal and adult B lymphopoiesis on this transcriptional regulator. B-cell development is arrested in the bone marrow at the early pro-B (pre-BI) cell stage, which is characterized by expression of the early markers c-kit, CD43, lambda5, VpreB, and HSA and the absence of the later markers CD25 and BP-1. These pre-BI cells fail to express the BSAP target gene CD19 and are capable of long-term proliferation in vitro in the presence of stromal cells and IL-7. B-lymphoid progenitors could not be detected in the fetal liver of Pax5 mutant embryos. However, Pax5-deficient fetal liver cells gave rise to the development of pre-BI cells in bone marrow on transplantation into lethally irradiated mice. These data indicate different functions of Pax5 in the distinctive microenvironments of fetal liver and adult bone marrow. As shown by PCR analyses, the pre-BI cells in Pax5-deficient bone marrow have undergone D(H)-to-J(H) rearrangement of the immunoglobulin heavy-chain locus at normal frequency. In contrast, V(H)-to-D(H)J(H) rearrangements were reduced approximately 50-fold in Pax5-deficient pre-BI cells, suggesting a role for Pax5 in the developmental pathway controlling V-to-DJ recombination.


Subject(s)
B-Lymphocytes/cytology , DNA Nucleotidyltransferases/metabolism , DNA-Binding Proteins/physiology , Immunoglobulin Heavy Chains/genetics , Nuclear Proteins/physiology , Recombination, Genetic , Transcription Factors , Animals , Bone Marrow/embryology , Bone Marrow Cells , Cell Lineage , DNA Nucleotidyltransferases/genetics , Gene Expression Regulation, Developmental , Liver/cytology , Liver/embryology , Mice , Mice, Inbred C57BL , PAX5 Transcription Factor , VDJ Recombinases
19.
Immunobiology ; 198(1-3): 227-35, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9442394

ABSTRACT

Pax-5 codes for the transcription factor BSAP which is expressed in all B-lymphoid tissues in addition to the developing central nervous system and testis. Within the B-lymphoid lineage, Pax-5 expression is already detected in the earliest B cell progenitors and persists up to the mature B cell stage. Targeted inactivation of the Pax-5 gene in the mouse germline revealed a differential dependency of fetal and adult B-lymphopoiesis on this transcription factor. Pax-5 is required for the differentiation of the earliest B-lineage-committed precursor cells in the fetal liver. In contrast, B cell development in the adult bone marrow progresses up to an early pro-B cell stage in the absence of Pax-5 function. The expression of CD19, Ig alpha (mb-1) and N-myc is severely reduced in Pax-5-deficient pro-B cells. These BSAP target genes are, however, unlikely to explain the early developmental block based on their known function in B cell development. Moreover, VH-to-DHJH rearrangements at the immunoglobulin heavy-chain locus are approximately 50-fold reduced in Pax-5-deficient pro B-cells, while the DH-to-JH rearrangements occur at a normal frequency. However, the expression of rearranged mu heavy-chain transgenes does not allow Pax-5-deficient pro-B cells to develop further to the pre-B cell stage. Together these data demonstrate therefore that B cell development in the Pax-5 deficient bone marrow is arrested at an early pro-B cell stage which is not yet responsive to pre-B cell receptor signaling.


Subject(s)
B-Lymphocytes/cytology , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors , Animals , Cell Differentiation , DNA-Binding Proteins/genetics , Gene Expression Regulation , Hematopoietic Stem Cells , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Joining Region/genetics , Immunoglobulin Variable Region/genetics , Mice , Nuclear Proteins/genetics , PAX5 Transcription Factor
20.
Recept Channels ; 3(1): 21-31, 1995.
Article in English | MEDLINE | ID: mdl-8589990

ABSTRACT

A member of the ionotropic family of glutamate receptors, hGluR4, was isolated from a human cDNA library and characterized following expression in mammalian cell lines. Human GluR4 possessed a 99% amino acid and 92% nucleotide homology to that of its rat counterpart with sequence differences restricted to the carboxy and amino terminal regions of the molecule. Transfection of simian kidney cells (COS-1) with an hGluR4 expression plasmid resulted in the transient formation of a membrane protein that possessed high specific binding for [3H](RS)-alpha-amino- 3-hydroxy-5-methylisoxazole-4-propionic acid ([3H]AMPA) but not [3H]kainate. Competition studies yielded a displacement profile of AMPA = quisqualate > glutamate > domoate > kainate >> N-methyl-D-aspartate (NMDA) or dihydrokainate. Whole-cell, voltage-clamp recordings from a human embryonic kidney cell line (HEK 293) stably expressing hGluR4 confirmed the presence of constitutively active, ligand-gated ion channels activated by AMPA, glutamate and kainate but not N-methyl-D-aspartate. Kainate-evoked currents were reversibly attenuated by 6-cyano-7-nitro- quinoxaline-2,3-dione (CNQX) but not DL-2-amino-5- phosphonovalerate (DL-AP5). Agonist-evoked currents exhibited inward rectification and ion substitution experiments indicated that hGluR4 receptor-linked ion channels in their homomeric state are permeable to both CA2+ and Na+ ions. In the same cell line antibody to rat GluR4 immunoprecipitated a major protein band at approximately 108 kDa and a minor one at approximately 340 kDa. The immunoblot analysis of membranes chemically crosslinked with dithiobis(succinimidylpropionate) showed a broad band at 550-600 kDa suggesting that the GluR4 receptor forms a pentamer in situ. This is the first report of the cloning of hGluR4 receptor and its stable expression in a human cell line.


Subject(s)
Receptors, Glutamate/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Electrophysiology , Gene Expression , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Humans , Kainic Acid/metabolism , Kainic Acid/pharmacology , Ligands , Molecular Sequence Data , Rats , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Sequence Homology, Amino Acid , Species Specificity , Transfection , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...