Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
EMBO J ; 42(15): e112741, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37337907

ABSTRACT

While extended loop extrusion across the entire Igh locus controls VH -DJH recombination, local regulatory sequences, such as the PAIR elements, may also activate VH gene recombination in pro-B-cells. Here, we show that PAIR-associated VH 8 genes contain a conserved putative regulatory element (V8E) in their downstream sequences. To investigate the function of PAIR4 and its V8.7E, we deleted 890 kb containing all 14 PAIRs in the Igh 5' region, which reduced distal VH gene recombination over a 100-kb distance on either side of the deletion. Reconstitution by insertion of PAIR4-V8.7E strongly activated distal VH gene recombination. PAIR4 alone resulted in lower induction of recombination, indicating that PAIR4 and V8.7E function as one regulatory unit. The pro-B-cell-specific activity of PAIR4 depends on CTCF, as mutation of its CTCF-binding site led to sustained PAIR4 activity in pre-B and immature B-cells and to PAIR4 activation in T-cells. Notably, insertion of V8.8E was sufficient to activate VH gene recombination. Hence, enhancers of the PAIR4-V8.7E module and V8.8E element activate distal VH gene recombination and thus contribute to the diversification of the BCR repertoire in the context of loop extrusion.


Subject(s)
Precursor Cells, B-Lymphoid , Regulatory Sequences, Nucleic Acid , Regulatory Sequences, Nucleic Acid/genetics , Binding Sites , Recombination, Genetic
2.
Nat Commun ; 14(1): 2316, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37085514

ABSTRACT

Extended loop extrusion across the immunoglobulin heavy-chain (Igh) locus facilitates VH-DJH recombination following downregulation of the cohesin-release factor Wapl by Pax5, resulting in global changes in the chromosomal architecture of pro-B cells. Here, we demonstrate that chromatin looping and VK-JK recombination at the Igk locus were insensitive to Wapl upregulation in pre-B cells. Notably, the Wapl protein was expressed at a 2.2-fold higher level in pre-B cells compared with pro-B cells, which resulted in a distinct chromosomal architecture with normal loop sizes in pre-B cells. High-resolution chromosomal contact analysis of the Igk locus identified multiple internal loops, which likely juxtapose VK and JK elements to facilitate VK-JK recombination. The higher Wapl expression in Igµ-transgenic pre-B cells prevented extended loop extrusion at the Igh locus, leading to recombination of only the 6 most 3' proximal VH genes and likely to allelic exclusion of all other VH genes in pre-B cells. These results suggest that pro-B and pre-B cells with their distinct chromosomal architectures use different chromatin folding principles for V gene recombination, thereby enabling allelic exclusion at the Igh locus, when the Igk locus is recombined.


Subject(s)
Immunoglobulin Heavy Chains , Precursor Cells, B-Lymphoid , V(D)J Recombination , Chromatin/genetics , Chromatin/metabolism , Immunoglobulin Heavy Chains/genetics , Immunoglobulins/genetics , Immunoglobulins/metabolism , Precursor Cells, B-Lymphoid/metabolism , Recombination, Genetic , V(D)J Recombination/genetics
3.
Sci Immunol ; 6(61)2021 07 23.
Article in English | MEDLINE | ID: mdl-34301800

ABSTRACT

The transcription factor Pax5 controls B cell development, but its role in mature B cells is largely enigmatic. Here, we demonstrated that the loss of Pax5 by conditional mutagenesis in peripheral B lymphocytes led to the strong reduction of B-1a, marginal zone (MZ), and germinal center (GC) B cells as well as plasma cells. Follicular (FO) B cells tolerated the loss of Pax5 but had a shortened half-life. The Pax5-deficient FO B cells failed to proliferate upon B cell receptor or Toll-like receptor stimulation due to impaired PI3K-AKT signaling, which was caused by increased expression of PTEN, a negative regulator of the PI3K pathway. Pax5 restrained PTEN protein expression at the posttranscriptional level, likely involving Pten-targeting microRNAs. Additional PTEN loss in Pten,Pax5 double-mutant mice rescued FO B cell numbers and the development of MZ B cells but did not restore GC B cell formation. Hence, the posttranscriptional down-regulation of PTEN expression is an important function of Pax5 that facilitates the differentiation and survival of mature B cells, thereby promoting humoral immunity.


Subject(s)
B-Lymphocytes/immunology , PAX5 Transcription Factor/immunology , PTEN Phosphohydrolase/immunology , Phosphatidylinositol 3-Kinases/immunology , Animals , Cell Differentiation , Down-Regulation , Female , Male , Mice, Transgenic , PAX5 Transcription Factor/genetics , PTEN Phosphohydrolase/genetics , Receptors, Antigen, B-Cell/immunology , Signal Transduction , Toll-Like Receptors/immunology
4.
Nature ; 584(7819): 142-147, 2020 08.
Article in English | MEDLINE | ID: mdl-32612238

ABSTRACT

Nuclear processes, such as V(D)J recombination, are orchestrated by the three-dimensional organization of chromosomes at multiple levels, including compartments1 and topologically associated domains (TADs)2,3 consisting of chromatin loops4. TADs are formed by chromatin-loop extrusion5-7, which depends on the loop-extrusion function of the ring-shaped cohesin complex8-12. Conversely, the cohesin-release factor Wapl13,14 restricts loop extension10,15. The generation of a diverse antibody repertoire, providing humoral immunity to pathogens, requires the participation of all V genes in V(D)J recombination16, which depends on contraction of the 2.8-Mb-long immunoglobulin heavy chain (Igh) locus by Pax517,18. However, how Pax5 controls Igh contraction in pro-B cells remains unknown. Here we demonstrate that locus contraction is caused by loop extrusion across the entire Igh locus. Notably, the expression of Wapl is repressed by Pax5 specifically in pro-B and pre-B cells, facilitating extended loop extrusion by increasing the residence time of cohesin on chromatin. Pax5 mediates the transcriptional repression of Wapl through a single Pax5-binding site by recruiting the polycomb repressive complex 2 to induce bivalent chromatin at the Wapl promoter. Reduced Wapl expression causes global alterations in the chromosome architecture, indicating that the potential to recombine all V genes entails structural changes of the entire genome in pro-B cells.


Subject(s)
Genes, Immunoglobulin Heavy Chain/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Variable Region/genetics , PAX5 Transcription Factor/metabolism , Proteins/genetics , Repressor Proteins/metabolism , V(D)J Recombination/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Binding Sites , Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Variable Region/chemistry , Mice , Polycomb Repressive Complex 2/metabolism , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Promoter Regions, Genetic/genetics , Cohesins
5.
Nat Immunol ; 20(11): 1517-1529, 2019 11.
Article in English | MEDLINE | ID: mdl-31591571

ABSTRACT

The establishment of a diverse B cell antigen receptor (BCR) repertoire by V(D)J recombination also generates autoreactive B cells. Anergy is one tolerance mechanism; it renders autoreactive B cells insensitive to stimulation by self-antigen, whereas Toll-like receptor (TLR) signaling can reactivate anergic B cells. Here, we describe a critical role of the transcription factor Ikaros in controlling BCR anergy and TLR signaling. Mice with specific deletion of Ikaros in mature B cells developed systemic autoimmunity. Ikaros regulated many anergy-associated genes, including Zfp318, which is implicated in the attenuation of BCR responsiveness by promoting immunoglobulin D expression in anergic B cells. TLR signaling was hyperactive in Ikaros-deficient B cells, which failed to upregulate feedback inhibitors of the MyD88-nuclear factor κB signaling pathway. Systemic inflammation was lost on expression of a non-self-reactive BCR or loss of MyD88 in Ikaros-deficient B cells. Thus, Ikaros acts as a guardian preventing autoimmunity by promoting BCR anergy and restraining TLR signaling.


Subject(s)
Autoimmunity/genetics , B-Lymphocytes/immunology , Clonal Anergy/genetics , Ikaros Transcription Factor/metabolism , Toll-Like Receptors/metabolism , Animals , B-Lymphocytes/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/immunology , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/immunology , Immunoglobulin D/immunology , Immunoglobulin D/metabolism , Mice , Models, Animal , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptors/immunology
6.
Nat Immunol ; 20(9): 1161-1173, 2019 09.
Article in English | MEDLINE | ID: mdl-31406378

ABSTRACT

Induction of the transcription factor Irf8 in the common dendritic cell progenitor (CDP) is required for classical type 1 dendritic cell (cDC1) fate specification, but the mechanisms controlling this induction are unclear. In the present study Irf8 enhancers were identified via chromatin profiling of dendritic cells and CRISPR/Cas9 genome editing was used to assess their roles in Irf8 regulation. An enhancer 32 kilobases (kb) downstream of the Irf8 transcriptional start site (+32-kb Irf8) that was active in mature cDC1s was required for the development of this lineage, but not for its specification. Instead, a +41-kb Irf8 enhancer, previously thought to be active only in plasmacytoid dendritic cells, was found to also be transiently accessible in cDC1 progenitors, and deleting this enhancer prevented the induction of Irf8 in CDPs and abolished cDC1 specification. Thus, cryptic activation of the +41-kb Irf8 enhancer in dendritic cell progenitors is responsible for cDC1 fate specification.


Subject(s)
Dendritic Cells/cytology , Enhancer Elements, Genetic/genetics , Interferon Regulatory Factors/metabolism , Macrophages/cytology , Monocytes/cytology , Animals , CRISPR-Cas Systems/genetics , Cell Differentiation , Cell Lineage , Dendritic Cells/immunology , Gene Expression Regulation , Interferon Regulatory Factors/genetics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Stem Cells/cytology , Tumor Cells, Cultured
7.
EMBO J ; 38(2)2019 01 15.
Article in English | MEDLINE | ID: mdl-30498131

ABSTRACT

The transcription factor Blimp1 is not only an essential regulator of plasma cells, but also a risk factor for the development of autoimmune disease in humans. Here, we demonstrate in the mouse that the Prdm1 (Blimp1) gene was partially activated at the chromatin and transcription level in early B cell development, although mature Prdm1 mRNA did not accumulate due to posttranscriptional regulation. By analyzing a mouse model that facilitated ectopic Blimp1 protein expression throughout B lymphopoiesis, we could demonstrate that Blimp1 impaired B cell development by interfering with the B cell gene expression program, while leading to an increased abundance of plasma cells by promoting premature plasmablast differentiation of immature and mature B cells. With progressing age, these mice developed an autoimmune disease characterized by the presence of autoantibodies and glomerulonephritis. Hence, these data identified ectopic Blimp1 expression as a novel mechanism, through which Blimp1 can act as a risk factor in the development of autoimmune disease.


Subject(s)
B-Lymphocytes/metabolism , Glomerulonephritis/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Animals , Autoantibodies/metabolism , B-Lymphocytes/cytology , Cell Differentiation , Disease Models, Animal , Female , Gene Regulatory Networks , Glomerulonephritis/genetics , Humans , Male , Mice , Transcriptional Activation
8.
Nat Immunol ; 18(4): 442-455, 2017 04.
Article in English | MEDLINE | ID: mdl-28250425

ABSTRACT

Innate-like B-1a cells provide a first line of defense against pathogens, yet little is known about their transcriptional control. Here we identified an essential role for the transcription factor Bhlhe41, with a lesser contribution by Bhlhe40, in controlling B-1a cell differentiation. Bhlhe41-/-Bhlhe40-/- B-1a cells were present at much lower abundance than were their wild-type counterparts. Mutant B-1a cells exhibited an abnormal cell-surface phenotype and altered B cell receptor (BCR) repertoire exemplified by loss of the phosphatidylcholine-specific VH12Vκ4 BCR. Expression of a pre-rearranged VH12Vκ4 BCR failed to 'rescue' the mutant phenotype and revealed enhanced proliferation accompanied by increased cell death. Bhlhe41 directly repressed the expression of cell-cycle regulators and inhibitors of BCR signaling while enabling pro-survival cytokine signaling. Thus, Bhlhe41 controls the development, BCR repertoire and self-renewal of B-1a cells.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Self Renewal , Receptors, Antigen, B-Cell/metabolism , Animals , B-Lymphocyte Subsets/immunology , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , Biomarkers , Cell Differentiation/genetics , Cell Self Renewal/genetics , Gene Expression Regulation , Genes, Immunoglobulin , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Nucleotide Motifs , Organ Specificity/genetics , Organ Specificity/immunology , Phenotype , Position-Specific Scoring Matrices , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/metabolism , Signal Transduction
9.
J Exp Med ; 213(7): 1201-21, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27261530

ABSTRACT

E2A is an essential regulator of early B cell development. Here, we have demonstrated that E2A together with E2-2 controlled germinal center (GC) B cell and plasma cell development. As shown by the identification of regulated E2A,E2-2 target genes in activated B cells, these E-proteins directly activated genes with important functions in GC B cells and plasma cells by inducing and maintaining DNase I hypersensitive sites. Through binding to multiple enhancers in the Igh 3' regulatory region and Aicda locus, E-proteins regulated class switch recombination by inducing both Igh germline transcription and AID expression. By regulating 3' Igk and Igh enhancers and a distal element at the Prdm1 (Blimp1) locus, E-proteins contributed to Igk, Igh, and Prdm1 activation in plasmablasts. Together, these data identified E2A and E2-2 as central regulators of B cell immunity.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , Germinal Center/immunology , Plasma Cells/immunology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , Enhancer Elements, Genetic/immunology , Germinal Center/cytology , Immunoglobulin Class Switching/immunology , Mice , Mice, Knockout , Plasma Cells/cytology , Transcription Factor 4
10.
Nat Immunol ; 17(3): 331-43, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26779602

ABSTRACT

The transcription factor Blimp-1 is necessary for the generation of plasma cells. Here we studied its functions in plasmablast differentiation by identifying regulated Blimp-1 target genes. Blimp-1 promoted the migration and adhesion of plasmablasts. It directly repressed genes encoding several transcription factors and Aicda (which encodes the cytidine deaminase AID) and thus silenced B cell-specific gene expression, antigen presentation and class-switch recombination in plasmablasts. It directly activated genes, which led to increased expression of the plasma cell regulator IRF4 and proteins involved in immunoglobulin secretion. Blimp-1 induced the transcription of immunoglobulin genes by controlling the 3' enhancers of the loci encoding the immunoglobulin heavy chain (Igh) and κ-light chain (Igk) and, furthermore, regulated the post-transcriptional expression switch from the membrane-bound form of the immunoglobulin heavy chain to its secreted form by activating Ell2 (which encodes the transcription-elongation factor ELL2). Notably, Blimp-1 recruited chromatin-remodeling and histone-modifying complexes to regulate its target genes. Hence, many essential functions of plasma cells are under the control of Blimp-1.


Subject(s)
Cell Differentiation/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin kappa-Chains/immunology , Interferon Regulatory Factors/immunology , Plasma Cells/immunology , Transcription Factors/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Adhesion/genetics , Cell Adhesion/immunology , Cell Differentiation/genetics , Cell Migration Assays, Leukocyte , Cell Movement/genetics , Cell Movement/immunology , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin kappa-Chains/genetics , Interferon Regulatory Factors/genetics , Mass Spectrometry , Mice , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Analysis, RNA , Transcription Factors/genetics
11.
Nat Immunol ; 15(3): 283-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24509509

ABSTRACT

The transcription factor Ikaros is an essential regulator of lymphopoiesis. Here we studied its B cell-specific function by conditional inactivation of the gene encoding Ikaros (Ikzf1) in pro-B cells. B cell development was arrested at an aberrant 'pro-B cell' stage characterized by increased cell adhesion and loss of signaling via the pre-B cell signaling complex (pre-BCR). Ikaros activated genes encoding signal transducers of the pre-BCR and repressed genes involved in the downregulation of pre-BCR signaling and upregulation of the integrin signaling pathway. Unexpectedly, derepression of expression of the transcription factor Aiolos did not compensate for the loss of Ikaros in pro-B cells. Ikaros induced or suppressed active chromatin at regulatory elements of activated or repressed target genes. Notably, binding of Ikaros and expression of its target genes were dynamically regulated at distinct stages of early B lymphopoiesis.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation/immunology , Ikaros Transcription Factor/immunology , Lymphopoiesis/immunology , Precursor Cells, B-Lymphoid/cytology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Chromatin Immunoprecipitation , Flow Cytometry , Gene Expression Regulation/immunology , Gene Knockdown Techniques , Ikaros Transcription Factor/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism
12.
Immunity ; 39(2): 229-44, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973221

ABSTRACT

The immunoglobulin heavy-chain (Igh) locus undergoes large-scale contraction in pro-B cells, which facilitates VH-DJH recombination by juxtaposing distal VH genes next to the DJH-rearranged gene segment in the 3' proximal Igh domain. By using high-resolution mapping of long-range interactions, we demonstrate that local interaction domains established the three-dimensional structure of the extended Igh locus in lymphoid progenitors. In pro-B cells, these local domains engaged in long-range interactions across the Igh locus, which depend on the regulators Pax5, YY1, and CTCF. The large VH gene cluster underwent flexible long-range interactions with the more rigidly structured proximal domain, which probably ensures similar participation of all VH genes in VH-DJH recombination to generate a diverse antibody repertoire. These long-range interactions appear to be an intrinsic feature of the VH gene cluster, because they are still generated upon mutation of the Eµ enhancer, IGCR1 insulator, or 3' regulatory region in the proximal Igh domain.


Subject(s)
Antibody Diversity/genetics , Gene Rearrangement, B-Lymphocyte, Heavy Chain/genetics , Genes, Immunoglobulin Heavy Chain , Immunoglobulin Variable Region/genetics , Precursor Cells, B-Lymphoid/immunology , Animals , Base Sequence , Binding Sites , CCCTC-Binding Factor , Chromosome Mapping , Gene Rearrangement , Mice , Mice, Inbred C57BL , PAX5 Transcription Factor/metabolism , Protein Binding , Repressor Proteins/metabolism , Sequence Analysis, DNA , YY1 Transcription Factor/metabolism
13.
Blood ; 122(5): 759-69, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23616623

ABSTRACT

The activation of B-cell-specific genes, such as CD19 and PAX5, is a hallmark of t(8;21) acute myeloid leukemia (AML) which expresses the translocation product RUNX1/ETO. PAX5 is an important regulator of B-lymphoid development and blocks myeloid differentiation when ectopically expressed. To understand the molecular mechanism of PAX5 deregulation, we examined its chromatin structure and regulation in t(8;21) AML cells, non-t(8;21) myeloid precursor control cells, and pre-B cells. In non-t(8;21) myeloid precursors, PAX5 is poised for transcription, but is repressed by polycomb complexes. In t(8;21) AML, PAX5 is not directly activated by RUNX1/ETO, but expression requires constitutive mitogen-activated protein (MAP) kinase signaling. Using a model of t(8;21) carrying an activating KIT mutation, we demonstrate that deregulated MAP kinase signaling in t(8;21) AML abrogates the association of polycomb complexes to PAX5 and leads to aberrant gene activation. Our findings therefore suggest a novel role of activating tyrosine kinase mutations in lineage-inappropriate gene expression in AML.


Subject(s)
Cell Lineage/genetics , Leukemia, Myeloid, Acute/genetics , PAX5 Transcription Factor/genetics , Polycomb-Group Proteins/physiology , Cell Line, Tumor , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 8/genetics , Down-Regulation/physiology , Gene Expression Regulation, Leukemic , HL-60 Cells , HeLa Cells , Humans , Leukemia, Myeloid, Acute/pathology , MAP Kinase Signaling System/physiology , Models, Biological , PAX5 Transcription Factor/metabolism , Polycomb-Group Proteins/metabolism , Protein Binding , Signal Transduction/genetics , Signal Transduction/physiology , Translocation, Genetic
14.
PLoS One ; 8(1): e54935, 2013.
Article in English | MEDLINE | ID: mdl-23383005

ABSTRACT

The gene encoding the receptor for macrophage colony-stimulating factor (CSF-1R) is expressed exclusively in cells of the myeloid lineages as well as trophoblasts. A conserved element in the second intron, Fms-Intronic Regulatory Element (FIRE), is essential for macrophage-specific transcription of the gene. However, the molecular details of how FIRE activity is regulated and how it impacts the Csf1r promoter have not been characterised. Here we show that agents that down-modulate Csf1r mRNA transcription regulated promoter activity altered the occupancy of key FIRE cis-acting elements including RUNX1, AP1, and Sp1 binding sites. We demonstrate that FIRE acts as an anti-sense promoter in macrophages and reversal of FIRE orientation within its native context greatly reduced enhancer activity in macrophages. Mutation of transcription initiation sites within FIRE also reduced transcription. These results demonstrate that FIRE is an orientation-specific transcribed enhancer element.


Subject(s)
Conserved Sequence/genetics , Genetic Loci/genetics , Introns/genetics , Receptor, Macrophage Colony-Stimulating Factor/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Base Sequence , Binding Sites , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Genetic Loci/drug effects , Humans , Introns/drug effects , Lipopolysaccharides/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/metabolism , Mice , Molecular Sequence Data , RNA Polymerase II/genetics , RNA, Antisense/genetics , RNA, Messenger/genetics , Sp1 Transcription Factor/metabolism , Time Factors , Transcription Factor AP-1/metabolism
15.
Article in English | MEDLINE | ID: mdl-24584058

ABSTRACT

Lymphocytes recognize a vast variety of pathogens by expressing a diverse repertoire of antigen receptor genes that are assembled by V(D)J recombination in immature B cells (Igh, Igk) and T cells (Tcrb, Tcra/d). V(D)J recombination takes place in the 3' proximal domain containing the D, J, and C gene segments, whereas 31 (Tcrb) to 200 (Igh) V genes are spread over a large region of 0.67 (Tcrb) to 3 (Igk) Mb pairs. All antigen receptor loci undergo reversible contraction at the developmental stage, where they engage in V-(D)J recombination. This long-range looping promotes the participation of all V genes in V-(D)J recombination by juxtaposing distant V genes next to (D)J segments in the proximal recombination center. The B-cell-specific Pax5, ubiquitous YY1, and architectural CTCF/cohesin proteins promote Igh locus contraction in pro-B cells by binding to multiple sites in the VH gene cluster. These regulators also control the pro-B-cell-specific activity of the distally located PAIR elements, which are likely involved in the regulation of VH-DJH recombination by mediating locus contraction. Notably, the large VH gene cluster of the Igh locus undergoes flexible long-range looping that ensures similar participation of all VH genes in VH-DJH recombination to generate a diverse antibody repertoire.


Subject(s)
Receptors, Antigen/genetics , V(D)J Recombination , VDJ Recombinases/metabolism , Alleles , Animals , Antibodies/immunology , B-Lymphocytes/immunology , Chromatin/metabolism , Enhancer Elements, Genetic , Humans , Lymphocytes/immunology , Mice , Models, Genetic , Multigene Family , Mutagenesis , Protein Binding , Receptors, Antigen/immunology , Transcription Factors/metabolism
16.
EMBO J ; 31(14): 3130-46, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22669466

ABSTRACT

Pax5 controls the identity and development of B cells by repressing lineage-inappropriate genes and activating B-cell-specific genes. Here, we used genome-wide approaches to identify Pax5 target genes in pro-B and mature B cells. In these cell types, Pax5 bound to 40% of the cis-regulatory elements defined by mapping DNase I hypersensitive (DHS) sites, transcription start sites and histone modifications. Although Pax5 bound to 8000 target genes, it regulated only 4% of them in pro-B and mature B cells by inducing enhancers at activated genes and eliminating DHS sites at repressed genes. Pax5-regulated genes in pro-B cells account for 23% of all expression changes occurring between common lymphoid progenitors and committed pro-B cells, which identifies Pax5 as an important regulator of this developmental transition. Regulated Pax5 target genes minimally overlap in pro-B and mature B cells, which reflects massive expression changes between these cell types. Hence, Pax5 controls B-cell identity and function by regulating distinct target genes in early and late B lymphopoiesis.


Subject(s)
Gene Expression Regulation/physiology , Lymphopoiesis/physiology , PAX5 Transcription Factor/metabolism , Precursor Cells, B-Lymphoid/metabolism , Response Elements/physiology , Transcription, Genetic/physiology , Animals , Mice , PAX5 Transcription Factor/genetics , Precursor Cells, B-Lymphoid/cytology
17.
Adv Immunol ; 111: 179-206, 2011.
Article in English | MEDLINE | ID: mdl-21970955

ABSTRACT

The B cell lineage of the hematopoietic system is responsible for the generation of high-affinity antibodies, which provide humoral immunity for protection against foreign pathogens. B cell commitment and development depend on many transcription factors including Pax5. Here, we review the different functions of Pax5 in regulating various aspects of B lymphopoiesis. At B cell commitment, Pax5 restricts the developmental potential of lymphoid progenitors to the B cell pathway by repressing B-lineage-inappropriate genes, while it simultaneously promotes B cell development by activating B-lymphoid-specific genes. Pax5 thereby controls gene transcription by recruiting chromatin-remodeling, histone-modifying, and basal transcription factor complexes to its target genes. Moreover, Pax5 contributes to the diversity of the antibody repertoire by controlling V(H)-DJ(H) recombination by inducing contraction of the immunoglobulin heavy-chain locus in pro-B cells, which is likely mediated by PAIR elements in the 5' region of the V(H) gene cluster. Importantly, all mature B cell types depend on Pax5 for their differentiation and function. Pax5 thus controls the identity of B lymphocytes throughout B cell development. Consequently, conditional loss of Pax5 allows mature B cells from peripheral lymphoid organs to develop into functional T cells in the thymus via dedifferentiation to uncommitted progenitors in the bone marrow. Pax5 has also been implicated in human B cell malignancies because it can function as a haploinsufficient tumor suppressor or oncogenic translocation fusion protein in B cell precursor acute lymphoblastic leukemia.


Subject(s)
Leukemia/pathology , PAX5 Transcription Factor/metabolism , Animals , Antibody Diversity , Antibody Formation , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Gene Expression Regulation , Gene Rearrangement, B-Lymphocyte , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Leukemia/immunology , Lymphopoiesis , PAX5 Transcription Factor/genetics
18.
Exp Hematol ; 39(7): 730-40.e1-2, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21549805

ABSTRACT

OBJECTIVE: The transcription factor PAX5 is essential for the activation of B-cell-specific genes and for the silencing of myeloid-specific genes. We previously determined the molecular mechanism by which PAX5 silences the myeloid-specific colony-stimulating-factor-receptor (Csf1R) gene and showed that PAX5 directly binds to the Csf1r promoter as well as to an intronic enhancer that generates an antisense transcript in B cells. Here we examine the role of PAX5 in the regulation of sense and antisense transcription in B cells. MATERIALS AND METHODS: We performed PAX5-specific chromatin immunoprecipitation analyses across the Csfr1 locus. We investigated the role of PAX5 in regulating Csf1r sense and antisense promoter activity by transient transfections and by employing a Pax5(-/-) pro-B-cell line expressing an inducible PAX5 protein. PAX5 interacting factors were identified by pull-down experiments. The role of the transcription factor Sp3 in driving antisense promoter expression was examined in B cells from Sp3 knockout mice. RESULTS: PAX5 differentially regulates the Csf1r promoter and the promoter of the antisense transcript. PAX5 interferes with PU.1 transactivation at the sense promoter by binding to a PAX5 consensus sequence. At the antisense promoter, PAX5 does not specifically recognize DNA, but interacts with Sp3 to upregulate antisense promoter activity. Antisense promoter activation by PAX5 is dependent on the presence of its partial homeo-domain. CONCLUSIONS: We demonstrate that PAX5 regulates Csf1r in B cells by reducing the frequency of binding of the basal transcription machinery to the promoter and by activating antisense RNA expression.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation , PAX5 Transcription Factor/genetics , Promoter Regions, Genetic/genetics , Receptor, Macrophage Colony-Stimulating Factor/genetics , Animals , Base Sequence , Binding Sites/genetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Chromatin Immunoprecipitation , DNA, Antisense/genetics , Mice , Mice, Knockout , Mutation , PAX5 Transcription Factor/metabolism , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
19.
EMBO J ; 30(12): 2388-404, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21552207

ABSTRACT

Pax5 is a critical regulator of B-cell commitment. Here, we identified direct Pax5 target genes by streptavidin-mediated ChIP-chip analysis of pro-B cells expressing in vivo biotinylated Pax5. By binding to promoters and enhancers, Pax5 directly regulates the expression of multiple transcription factor, cell surface receptor and signal transducer genes. One of the newly identified enhancers was shown by transgenic analysis to confer Pax5-dependent B-cell-specific activity to the Nedd9 gene controlling B-cell trafficking. Profiling of histone modifications in Pax5-deficient and wild-type pro-B cells demonstrated that Pax5 induces active chromatin at activated target genes, while eliminating active chromatin at repressed genes in committed pro-B cells. Pax5 rapidly induces these chromatin and transcription changes by recruiting chromatin-remodelling, histone-modifying and basal transcription factor complexes to its target genes. These data provide novel insight into the regulatory network and epigenetic regulation, by which Pax5 controls B-cell commitment.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/metabolism , Cell Differentiation , Chromatin/metabolism , Gene Targeting , PAX5 Transcription Factor/physiology , Animals , Cell Differentiation/genetics , Cell Line , Gene Knock-In Techniques , Gene Targeting/methods , Mice , Mice, Knockout , Mice, Transgenic , PAX5 Transcription Factor/genetics , Protein Binding/genetics , Protein Transport/genetics , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/metabolism
20.
Immunity ; 34(2): 175-87, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21349430

ABSTRACT

V(H)-DJ(H) recombination of the immunoglobulin heavy chain (Igh) locus is temporally and spatially controlled during early B cell development, and yet no regulatory elements other than the V(H) gene promoters have been identified throughout the entire V(H) gene cluster. Here, we discovered regulatory sequences that are interspersed in the distal V(H) gene region. These conserved repeat elements were characterized by the presence of Pax5 transcription factor-dependent active chromatin by binding of the regulators Pax5, E2A, CTCF, and Rad21, as well as by Pax5-dependent antisense transcription in pro-B cells. The Pax5-activated intergenic repeat (PAIR) elements were no longer bound by Pax5 in pre-B and B cells consistent with the loss of antisense transcription, whereas E2A and CTCF interacted with PAIR elements throughout early B cell development. The pro-B cell-specific and Pax5-dependent activity of the PAIR elements suggests that they are involved in the regulation of distal V(H)-DJ(H) recombination at the Igh locus.


Subject(s)
Chromatin/genetics , DNA, Intergenic/genetics , Gene Rearrangement, B-Lymphocyte, Heavy Chain , Genes, Immunoglobulin/genetics , Immunoglobulin Heavy Chains/genetics , PAX5 Transcription Factor/physiology , Regulatory Sequences, Nucleic Acid/genetics , Animals , B-Lymphocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/physiology , Binding Sites , CCCTC-Binding Factor , Chromatin Immunoprecipitation , Conserved Sequence , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Genetic , Oligonucleotide Array Sequence Analysis , PAX5 Transcription Factor/deficiency , PAX5 Transcription Factor/genetics , Precursor Cells, B-Lymphoid/metabolism , RNA, Antisense/biosynthesis , RNA, Antisense/genetics , Repressor Proteins/physiology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...