Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Immunity ; 54(11): 2465-2480.e5, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34706222

ABSTRACT

Epigenetic reprogramming underlies specification of immune cell lineages, but patterns that uniquely define immune cell types and the mechanisms by which they are established remain unclear. Here, we identified lineage-specific DNA methylation signatures of six immune cell types from human peripheral blood and determined their relationship to other epigenetic and transcriptomic patterns. Sites of lineage-specific hypomethylation were associated with distinct combinations of transcription factors in each cell type. By contrast, sites of lineage-specific hypermethylation were restricted mostly to adaptive immune cells. PU.1 binding sites were associated with lineage-specific hypo- and hypermethylation in different cell types, suggesting that it regulates DNA methylation in a context-dependent manner. These observations indicate that innate and adaptive immune lineages are specified by distinct epigenetic mechanisms via combinatorial and context-dependent use of key transcription factors. The cell-specific epigenomics and transcriptional patterns identified serve as a foundation for future studies on immune dysregulation in diseases and aging.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Epigenomics , Gene Expression Regulation , Immunity , Transcription Factors/metabolism , Transcriptome , Epigenomics/methods , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Transcription Factors/genetics
2.
Genes Dev ; 36(15-16): 901-915, 2022 08 01.
Article in English | MEDLINE | ID: mdl-36167471

ABSTRACT

Transcription factor EBF1 (early B cell factor 1) acts as a key regulator of B cell specification. The transcriptional network in which EBF1 operates has been extensively studied; however, the regulation of EBF1 function remains poorly defined. By mass spectrometric analysis of proteins associated with endogenous EBF1 in pro-B cells, we identified the nuclear import receptor Transportin-3 (Tnpo3) and found that it interacts with the immunoglobulin-like fold domain of EBF1. We delineated glutamic acid 271 of EBF1 as a critical residue for the association with Tnpo3. EBF1E271A showed normal nuclear localization; however, it had an impaired B cell programming ability in conditions of Notch signaling, as determined by retroviral transduction of Ebf1 -/- progenitors. By RNA-seq analysis of EBF1E271A-expressing progenitors, we found an up-regulation of T lineage determinants and down-regulation of early B genes, although similar chromatin binding of EBF1E271A and EBF1wt was detected in pro-B cells expressing activated Notch1. B lineage-specific inactivation of Tnpo3 in mice resulted in a block of early B cell differentiation, accompanied by a down-regulation of B lineage genes and up-regulation of T and NK lineage genes. Taken together, our observations suggest that Tnpo3 ensures B cell programming by EBF1 in nonpermissive conditions.


Subject(s)
Glutamic Acid , Trans-Activators , beta Karyopherins , Animals , Mice , beta Karyopherins/metabolism , Cell Lineage/genetics , Chromatin , Immunoglobulins/genetics , Immunoglobulins/metabolism , Karyopherins/genetics , Receptors, Notch/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism
3.
Genes Dev ; 34(21-22): 1503-1519, 2020 11 01.
Article in English | MEDLINE | ID: mdl-33004416

ABSTRACT

EBF1 and PAX5 mutations are associated with the development of B progenitor acute lymphoblastic leukemia (B-ALL) in humans. To understand the molecular networks driving leukemia in the Ebf1+/-Pax5+/- (dHet) mouse model for B-ALL, we interrogated the transcriptional profiles and chromatin status of leukemic cells, preleukemic dHet pro-B, and wild-type pro-B cells with the corresponding EBF1 and Pax5 cistromes. In dHet B-ALL cells, many EBF1 and Pax5 target genes encoding pre-BCR signaling components and transcription factors were down-regulated, whereas Myc and genes downstream from IL-7 signaling or associated with the folate pathway were up-regulated. We show that blockade of IL-7 signaling in vivo and methotrexate treatment of leukemic cells in vitro attenuate the expansion of leukemic cells. Single-cell RNA-sequencing revealed heterogeneity of leukemic cells and identified a subset of wild-type pro-B cells with reduced Ebf1 and enhanced Myc expression that show hallmarks of dHet B-ALL cells. Thus, EBF1 and Pax5 may safeguard early stage B cells from transformation to B-ALL by limiting IL-7 signaling, folate metabolism and Myc expression.


Subject(s)
Folic Acid/metabolism , Interleukin-7/physiology , PAX5 Transcription Factor/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Proto-Oncogene Proteins c-myc/genetics , Signal Transduction/genetics , Trans-Activators/metabolism , Animals , Carbon/metabolism , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Mice , PAX5 Transcription Factor/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cells, B-Lymphoid/pathology , Protein Binding , Single-Cell Analysis , Trans-Activators/genetics
4.
Genes Dev ; 32(2): 96-111, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29440261

ABSTRACT

B-cell fate determination requires the action of transcription factors that operate in a regulatory network to activate B-lineage genes and repress lineage-inappropriate genes. However, the dynamics and hierarchy of events in B-cell programming remain obscure. To uncouple the dynamics of transcription factor expression from functional consequences, we generated induction systems in developmentally arrested Ebf1-/- pre-pro-B cells to allow precise experimental control of EBF1 expression in the genomic context of progenitor cells. Consistent with the described role of EBF1 as a pioneer transcription factor, we show in a time-resolved analysis that EBF1 occupancy coincides with EBF1 expression and precedes the formation of chromatin accessibility. We observed dynamic patterns of EBF1 target gene expression and sequential up-regulation of transcription factors that expand the regulatory network at the pro-B-cell stage. A continuous EBF1 function was found to be required for Cd79a promoter activity and for the maintenance of an accessible chromatin domain that is permissive for binding of other transcription factors. Notably, transient EBF1 occupancy was detected at lineage-inappropriate genes prior to their silencing in pro-B cells. Thus, persistent and transient functions of EBF1 allow for an ordered sequence of epigenetic and transcriptional events in B-cell programming.


Subject(s)
B-Lymphocytes/metabolism , Epigenesis, Genetic , Stem Cells/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Animals , CD79 Antigens/genetics , Cell Lineage/genetics , Cells, Cultured , Chromatin/metabolism , DNA/metabolism , Mice , PAX5 Transcription Factor/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism
5.
Immunity ; 44(3): 527-541, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26982363

ABSTRACT

Lymphopoiesis requires the activation of lineage-specific genes embedded in naive, inaccessible chromatin or in primed, accessible chromatin. The mechanisms responsible for de novo gain of chromatin accessibility, known as "pioneer" function, remain poorly defined. Here, we showed that the EBF1 C-terminal domain (CTD) is required for the regulation of a specific gene set involved in B cell fate decision and differentiation, independently of activation and repression functions. Using genome-wide analysis of DNaseI hypersensitivity and DNA methylation in multipotent Ebf1(-/-) progenitors and derivative EBF1wt- or EBF1ΔC-expressing cells, we found that the CTD promoted chromatin accessibility and DNA demethylation in previously naive chromatin. The CTD allowed EBF1 to bind at inaccessible genomic regions that offer limited co-occupancy by other transcription factors, whereas the CTD was dispensable for EBF1 binding at regions that are occupied by multiple transcription factors. Thus, the CTD enables EBF1 to confer permissive lineage-specific changes in progenitor chromatin landscape.


Subject(s)
B-Lymphocytes/physiology , Chromatin/metabolism , Lymphoid Progenitor Cells/physiology , Trans-Activators/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , DNA Methylation/genetics , Gene Regulatory Networks/genetics , Lymphopoiesis , Mice , Mice, Knockout , Mice, Transgenic , Protein Structure, Tertiary/genetics , Trans-Activators/genetics
6.
Genes Dev ; 30(20): 2310-2324, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27807034

ABSTRACT

Transcription factor EBF1 (early B-cell factor 1) regulates early B-cell differentiation by poising or activating lineage-specific genes and repressing genes associated with alternative cell fates. To identify proteins that regulate the diverse functions of EBF1, we used SILAC (stable isotope labeling by amino acids in cell culture)-based mass spectrometry of proteins associated with endogenous EBF1 in pro-B cells. This analysis identified most components of the multifunctional CCR4-NOT complex, which regulates transcription and mRNA degradation. CNOT3 interacts with EBF1, and we identified histidine 240 in EBF1 as a critical residue for this interaction. Complementation of Ebf1-/- progenitors with EBF1H240A revealed a partial block of pro-B-cell differentiation and altered expression of specific EBF1 target genes that show either reduced transcription or increased mRNA stability. Most deregulated EBF1 target genes show normal occupancy by EBF1H240A, but we also detected genes with altered occupancy, suggesting that the CCR4-NOT complex affects multiple activities of EBF1. Mice with conditional Cnot3 inactivation recapitulate the block of early B-cell differentiation, which we found to be associated with an impaired autoregulation of Ebf1 and reduced expression of pre-B-cell receptor components. Thus, the interaction of the CCR4-NOT complex with EBF1 diversifies the function of EBF1 in a context-dependent manner and may coordinate transcriptional and post-transcriptional gene regulation.


Subject(s)
B-Lymphocytes/physiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation/genetics , Homeodomain Proteins/metabolism , Lymphopoiesis/genetics , Nuclear Proteins/metabolism , RNA Stability/genetics , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Gene Silencing , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Mice , Molecular Chaperones/genetics , Mutation , Nuclear Proteins/genetics , Protein Binding , Transcription Factors/genetics , Transgenes
7.
Trends Genet ; 34(4): 257-269, 2018 04.
Article in English | MEDLINE | ID: mdl-29336845

ABSTRACT

Hematopoiesis is regulated by signals from the microenvironment, transcription factor networks, and changes of the epigenetic landscape. Transcription factors interact with and shape chromatin to allow for lineage- and cell type-specific changes in gene expression. During B lymphopoiesis, epigenetic regulation is observed in multilineage progenitors in which a specific chromatin context is established, at the onset of the B cell differentiation when early B cell factor 1 (EBF1) induces lineage-specific changes in chromatin, during V(D)J recombination and after antigen-driven activation of B cells and terminal differentiation. In this review, we discuss the epigenetic changes underlying B cell differentiation, focusing on the role of transcription factor EBF1 in B cell lineage priming.


Subject(s)
B-Lymphocytes/immunology , Chromatin/chemistry , Epigenesis, Genetic , Gene Regulatory Networks , Lymphopoiesis/genetics , Trans-Activators/genetics , Animals , B-Lymphocytes/cytology , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Chromatin/metabolism , Gene Expression , Histones/genetics , Histones/immunology , Humans , Stem Cells/cytology , Stem Cells/immunology , Trans-Activators/immunology , V(D)J Recombination
8.
Genes Dev ; 26(7): 668-82, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22431510

ABSTRACT

The transcription factor Ebf1 is an important determinant of early B lymphopoiesis. To gain insight into the functions of Ebf1 at distinct stages of differentiation, we conditionally inactivated Ebf1. We found that Ebf1 is required for the proliferation, survival, and signaling of pro-B cells and peripheral B-cell subsets, including B1 cells and marginal zone B cells. The proliferation defect of Ebf1-deficient pro-B cells and the impaired expression of multiple cell cycle regulators are overcome by transformation with v-Abl. The survival defect of transformed Ebf1(fl/fl) pro-B cells can be rescued by the forced expression of the Ebf1 targets c-Myb or Bcl-x(L). In mature B cells, Ebf1 deficiency interferes with signaling via the B-cell-activating factor receptor (BAFF-R)- and B-cell receptor (BCR)-dependent Akt pathways. Moreover, Ebf1 is required for germinal center formation and class switch recombination. Genome-wide analyses of Ebf1-mediated gene expression and chromatin binding indicate that Ebf1 regulates both common and distinct sets of genes in early and late stage B cells. By regulating important components of transcription factor and signaling networks, Ebf1 appears to be involved in the coordination of cell proliferation, survival, and differentiation at multiple stages of B lymphopoiesis.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation , Cell Proliferation , Signal Transduction , Trans-Activators/metabolism , Animals , B-Lymphocytes/immunology , Cell Survival , Gene Expression Regulation , Genome-Wide Association Study , Immunoglobulin Heavy Chains/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Trans-Activators/genetics , Trans-Activators/immunology , Transcription, Genetic
9.
J Proteome Res ; 17(1): 76-85, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29181981

ABSTRACT

Early B cell factor 1 (EBF1) is one of the key transcription factors required for orchestrating B-cell lineage development. Although studies have shown that Ebf1 haploinsufficiency is involved in the development of leukemia, no study has been conducted that characterizes the global effect of Ebf1 heterozygosity on the proteome of pro-B lymphocytes. Here, we employ both data independent acquisition (DIA) and shotgun data dependent acquisition (DDA) workflows for profiling proteins that are differently expressed between Ebf1+/+ and Ebf1+/- cells. Both DDA and DIA were able to reveal the downregulation of the EBF1 transcription factor in Ebf1+/- pro-B lymphocytes. Further examination of differentially expressed proteins by DIA revealed that, similar to EBF1, the expression of other B-cell lineage regulators, such as TCF3 and Pax5, is also downregulated in Ebf1 heterozygous cells. Functional DIA analysis of differentially expressed proteins showed that EBF1 heterozygosity resulted in the deregulation of at least eight transcription factors involved in lymphopoiesis and the deregulation of key proteins playing crucial roles in survival, development, and differentiation of pro-B lymphocytes.


Subject(s)
Data Collection/methods , Precursor Cells, B-Lymphoid/chemistry , Proteomics/methods , Trans-Activators/genetics , Animals , Gene Expression Regulation , Heterozygote , Humans , Lymphopoiesis/genetics , Mice , Transcription Factors/metabolism
10.
Immunol Rev ; 261(1): 102-15, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25123279

ABSTRACT

During the last decades, many studies have investigated the transcriptional and epigenetic regulation of lineage decision in the hematopoietic system. These efforts led to a model in which extrinsic signals and intrinsic cues establish a permissive chromatin context upon which a regulatory network of transcription factors and epigenetic modifiers act to guide the differentiation of hematopoietic lineages. These networks include lineage-specific factors that further modify the epigenetic landscape and promote the generation of specific cell types. The process of B lymphopoiesis requires a set of transcription factors, including Ikaros, PU.1, E2A, and FoxO1 to 'prime' cis-regulatory regions for subsequent activation by the B-lineage-specific transcription factors EBF1 and Pax-5. The expression of EBF1 is initiated by the combined action of E2A and FoxO1, and it is further enhanced and maintained by several positive feedback loops that include Pax-5 and IL-7 signaling. EBF1 acts in concert with Ikaros, PU.1, Runx1, E2A, FoxO1, and Pax-5 to establish the B cell-specific transcription profile. EBF1 and Pax-5 also collaborate to repress alternative cell fates and lock cells into the B-lineage fate. In addition to the functions of EBF1 in establishing and maintaining B-cell identity, EBF1 is required to coordinate differentiation with cell proliferation and survival.


Subject(s)
B-Lymphocytes/immunology , Histone Code/physiology , Trans-Activators/metabolism , Animals , Cell Lineage , Cell Proliferation , Cell Survival , Epigenesis, Genetic , Feedback, Physiological , Gene Expression Regulation , Gene Regulatory Networks/immunology , Homeostasis , Humans , Interleukin-7/metabolism , Lymphopoiesis
11.
Nat Genet ; 46(2): 176-181, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24362818

ABSTRACT

Follicular lymphoma is an incurable malignancy, with transformation to an aggressive subtype representing a critical event during disease progression. Here we performed whole-genome or whole-exome sequencing on 10 follicular lymphoma-transformed follicular lymphoma pairs followed by deep sequencing of 28 genes in an extension cohort, and we report the key events and evolutionary processes governing tumor initiation and transformation. Tumor evolution occurred through either a 'rich' or 'sparse' ancestral common progenitor clone (CPC). We identified recurrent mutations in linker histone, JAK-STAT signaling, NF-κB signaling and B cell developmental genes. Longitudinal analyses identified early driver mutations in chromatin regulator genes (CREBBP, EZH2 and KMT2D (MLL2)), whereas mutations in EBF1 and regulators of NF-κB signaling (MYD88 and TNFAIP3) were gained at transformation. Collectively, this study provides new insights into the genetic basis of follicular lymphoma and the clonal dynamics of transformation and suggests that personalizing therapies to target key genetic alterations in the CPC represents an attractive therapeutic strategy.


Subject(s)
Cell Transformation, Neoplastic/genetics , Disease Progression , Genomics/methods , Lymphoma, Follicular/genetics , Lymphoma, Follicular/physiopathology , Base Sequence , CREB-Binding Protein/genetics , Cluster Analysis , Cohort Studies , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein , Exome/genetics , High-Throughput Nucleotide Sequencing , Histones/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Molecular Sequence Annotation , Molecular Sequence Data , Mutagenesis , Mutation/genetics , Myeloid Differentiation Factor 88/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Phylogeny , Polycomb Repressive Complex 2/genetics , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA , Trans-Activators/genetics , Tumor Necrosis Factor alpha-Induced Protein 3
12.
FEBS Lett ; 587(6): 711-6, 2013 Mar 18.
Article in English | MEDLINE | ID: mdl-23395800

ABSTRACT

Early B-cell factor 1 (Ebf1) is a transcription factor whose inactivation in all cells results in high bone mass because of an increase in bone formation. This observation suggests Ebf1 may be an inhibitor of osteoblast differentiation. To test this contention, we analyzed Ebf1 pattern of expression and function in osteoblasts ex vivo and in vivo through osteoblast-specific inactivation in the mouse. We show here that in vivo deletion of Ebf1 in osteoblast progenitors does not affect osteoblast differentiation or bone formation accrual post-natally. These observations indicate that the phenotype described in Ebf1(-/)(-) mice is not osteoblast-autonomous.


Subject(s)
Bone and Bones/metabolism , Gene Expression Regulation, Developmental , Osteoblasts/metabolism , Osteogenesis/genetics , Trans-Activators/genetics , Animals , Bone and Bones/cytology , Cell Differentiation , Embryo, Mammalian , Gene Deletion , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL