Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters

Uruguay Oncology Collection
Publication year range
1.
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
2.
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
3.
Immunity ; 54(9): 2005-2023.e10, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525339

ABSTRACT

Cell fate decisions during early B cell activation determine the outcome of responses to pathogens and vaccines. We examined the early B cell response to T-dependent antigen in mice by single-cell RNA sequencing. Early after immunization, a homogeneous population of activated precursors (APs) gave rise to a transient wave of plasmablasts (PBs), followed a day later by the emergence of germinal center B cells (GCBCs). Most APs rapidly exited the cell cycle, giving rise to non-GC-derived early memory B cells (eMBCs) that retained an AP-like transcriptional profile. Rapid decline of antigen availability controlled these events; provision of excess antigen precluded cell cycle exit and induced a new wave of PBs. Fate mapping revealed a prominent contribution of eMBCs to the MBC pool. Quiescent cells with an MBC phenotype dominated the early response to immunization in primates. A reservoir of APs/eMBCs may enable rapid readjustment of the immune response when failure to contain a threat is manifested by increased antigen availability.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Immunity, Humoral/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Animals , Antigen Presentation/immunology , Cell Differentiation/immunology , Mice , Plasma Cells/immunology , Precursor Cells, B-Lymphoid/immunology
4.
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
5.
Cell ; 157(2): 340-356, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24725403

ABSTRACT

Innate lymphoid cells (ILCs) are a recently recognized group of lymphocytes that have important functions in protecting epithelial barriers against infections and in maintaining organ homeostasis. ILCs have been categorized into three distinct groups, transcriptional circuitry and effector functions of which strikingly resemble the various T helper cell subsets. Here, we identify a common, Id2-expressing progenitor to all interleukin 7 receptor-expressing, "helper-like" ILC lineages, the CHILP. Interestingly, the CHILP differentiated into ILC2 and ILC3 lineages, but not into conventional natural killer (cNK) cells that have been considered an ILC1 subset. Instead, the CHILP gave rise to a peculiar NKp46(+) IL-7Rα(+) ILC lineage that required T-bet for specification and was distinct of cNK cells or other ILC lineages. Such ILC1s coproduced high levels of IFN-γ and TNF and protected against infections with the intracellular parasite Toxoplasma gondii. Our data significantly advance our understanding of ILC differentiation and presents evidence for a new ILC lineage that protects barrier surfaces against intracellular infections.


Subject(s)
Cell Differentiation , Lymphocytes/cytology , Lymphocytes/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , GATA3 Transcription Factor/metabolism , Immunity, Innate , Inhibitor of Differentiation Protein 2/metabolism , Mice , Mice, Inbred C57BL , Receptors, Interleukin-7/metabolism , Stem Cells/cytology , Toxoplasma , Toxoplasmosis/immunology
6.
EMBO J ; 43(12): 2506-2525, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38689024

ABSTRACT

Many microRNAs (miRNAs) are expressed with high spatiotemporal specificity during organismal development, with some being limited to rare cell types, often embedded in complex tissues. Yet, most miRNA profiling efforts remain at the tissue and organ levels. To overcome challenges in accessing the microRNomes from tissue-embedded cells, we had previously developed mime-seq (miRNome by methylation-dependent sequencing), a technique in which cell-specific miRNA methylation in C. elegans and Drosophila enabled chemo-selective sequencing without the need for cell sorting or biochemical purification. Here, we present mime-seq 2.0 for profiling miRNAs from specific mouse cell types. We engineered a chimeric RNA methyltransferase that is tethered to Argonaute protein and efficiently methylates miRNAs at their 3'-terminal 2'-OH in mouse and human cell lines. We also generated a transgenic mouse for conditional expression of this methyltransferase, which can be used to direct methylation of miRNAs in a cell type of choice. We validated the use of this mouse model by profiling miRNAs from B cells and bone marrow plasma cells.


Subject(s)
MicroRNAs , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Mice , Humans , Mice, Transgenic , Methylation , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Sequence Analysis, RNA/methods , Methyltransferases/genetics , Methyltransferases/metabolism , Cell Line , B-Lymphocytes/metabolism
7.
Nat Immunol ; 17(3): 323-30, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26779600

ABSTRACT

Plasma cell differentiation requires silencing of B cell transcription, while it establishes antibody-secretory function and long-term survival. The transcription factors Blimp-1 and IRF4 are essential for the generation of plasma cells; however, their function in mature plasma cells has remained elusive. We found that while IRF4 was essential for the survival of plasma cells, Blimp-1 was dispensable for this. Blimp-1-deficient plasma cells retained their transcriptional identity but lost the ability to secrete antibody. Blimp-1 regulated many components of the unfolded protein response (UPR), including XBP-1 and ATF6. The overlap in the functions of Blimp-1 and XBP-1 was restricted to that response, with Blimp-1 uniquely regulating activity of the kinase mTOR and the size of plasma cells. Thus, Blimp-1 was required for the unique physiological ability of plasma cells that enables the secretion of protective antibody.


Subject(s)
Cell Differentiation/immunology , Immunoglobulins/immunology , Interferon Regulatory Factors/immunology , Plasma Cells/immunology , Transcription Factors/immunology , Unfolded Protein Response/immunology , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/immunology , Animals , Cell Size , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Immunoglobulins/metabolism , Interferon Regulatory Factors/genetics , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Regulatory Factor X Transcription Factors , Sequence Analysis, DNA , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology , Transcription Factors/genetics , Unfolded Protein Response/genetics , X-Box Binding Protein 1
8.
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
9.
Nat Immunol ; 17(10): 1187-96, 2016 10.
Article in English | MEDLINE | ID: mdl-27487330

ABSTRACT

During unresolved infections, some viruses escape immunological control and establish a persistant reservoir in certain cell types, such as human immunodeficiency virus (HIV), which persists in follicular helper T cells (TFH cells), and Epstein-Barr virus (EBV), which persists in B cells. Here we identified a specialized group of cytotoxic T cells (TC cells) that expressed the chemokine receptor CXCR5, selectively entered B cell follicles and eradicated infected TFH cells and B cells. The differentiation of these cells, which we have called 'follicular cytotoxic T cells' (TFC cells), required the transcription factors Bcl6, E2A and TCF-1 but was inhibited by the transcriptional regulators Blimp1, Id2 and Id3. Blimp1 and E2A directly regulated Cxcr5 expression and, together with Bcl6 and TCF-1, formed a transcriptional circuit that guided TFC cell development. The identification of TFC cells has far-reaching implications for the development of strategies to control infections that target B cells and TFH cells and to treat B cell-derived malignancies.


Subject(s)
Arenaviridae Infections/immunology , B-Lymphocytes/immunology , Epstein-Barr Virus Infections/immunology , HIV/immunology , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Germinal Center/pathology , Germinal Center/virology , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
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
11.
EMBO J ; 41(7): e108397, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35156727

ABSTRACT

While PAX5 is an important tumor suppressor gene in B-cell acute lymphoblastic leukemia (B-ALL), it is also involved in oncogenic translocations coding for diverse PAX5 fusion proteins. PAX5-JAK2 encodes a protein consisting of the PAX5 DNA-binding region fused to the constitutively active JAK2 kinase domain. Here, we studied the oncogenic function of the PAX5-JAK2 fusion protein in a mouse model expressing it from the endogenous Pax5 locus, resulting in inactivation of one of the two Pax5 alleles. Pax5Jak2/+ mice rapidly developed an aggressive B-ALL in the absence of another cooperating exogenous gene mutation. The DNA-binding function and kinase activity of Pax5-Jak2 as well as IL-7 signaling contributed to leukemia development. Interestingly, all Pax5Jak2/+ tumors lost the remaining wild-type Pax5 allele, allowing efficient DNA-binding of Pax5-Jak2. While we could not find evidence for a nuclear role of Pax5-Jak2 as an epigenetic regulator, high levels of active phosphorylated STAT5 and increased expression of STAT5 target genes were seen in Pax5Jak2/+ B-ALL tumors, implying that nuclear Pax5-Jak2 phosphorylates STAT5. Together, these data reveal Pax5-Jak2 as an important nuclear driver of leukemogenesis by maintaining phosphorylated STAT5 levels in the nucleus.


Subject(s)
Janus Kinase 2 , Leukemia, B-Cell , PAX5 Transcription Factor , STAT5 Transcription Factor , Animals , Janus Kinase 2/genetics , Leukemia, B-Cell/genetics , Mice , Mutation , PAX5 Transcription Factor/genetics , STAT5 Transcription Factor/genetics , Translocation, Genetic
12.
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
13.
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
14.
Nat Immunol ; 14(10): 1037-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23974957

ABSTRACT

The transcription factor GATA-3 is expressed and required for differentiation and function throughout the T lymphocyte lineage. Despite evidence it may also be expressed in multipotent hematopoietic stem cells (HSCs), any role for GATA-3 in these cells has remained unclear. Here we found GATA-3 was in the cytoplasm in quiescent long-term stem cells from steady-state bone marrow but relocated to the nucleus when HSCs cycled. Relocation depended on signaling via the mitogen-activated protein kinase p38 and was associated with a diminished capacity for long-term reconstitution after transfer into irradiated mice. Deletion of Gata3 enhanced the repopulating capacity and augmented the self-renewal of long-term HSCs in cell-autonomous fashion without affecting the cell cycle. Our observations position GATA-3 as a regulator of the balance between self-renewal and differentiation in HSCs that acts downstream of the p38 signaling pathway.


Subject(s)
GATA3 Transcription Factor/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , GATA3 Transcription Factor/genetics , Gene Deletion , Gene Expression , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Ligands , Mice , Mice, Knockout , Poly I-C/pharmacology , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Immunity ; 44(1): 103-115, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26795246

ABSTRACT

The inhibitor of DNA binding 2 (Id2) is essential for natural killer (NK) cell development with its canonical role being to antagonize E-protein function and alternate lineage fate. Here we have identified a key role for Id2 in regulating interleukin-15 (IL-15) receptor signaling and homeostasis of NK cells by repressing multiple E-protein target genes including Socs3. Id2 deletion in mature NK cells was incompatible with their homeostasis due to impaired IL-15 receptor signaling and metabolic function and this could be rescued by strong IL-15 receptor stimulation or genetic ablation of Socs3. During NK cell maturation, we observed an inverse correlation between E-protein target genes and Id2. These results shift the current paradigm on the role of ID2, indicating that it is required not only to antagonize E-proteins during NK cell commitment, but constantly required to titrate E-protein activity to regulate NK cell fitness and responsiveness to IL-15.


Subject(s)
Cell Differentiation/immunology , Inhibitor of Differentiation Protein 2/immunology , Interleukin-15/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Animals , Cell Lineage/immunology , Cells, Cultured , Female , Flow Cytometry , Male , Mice , Mice, Mutant Strains , Receptors, Interleukin-15/immunology , Receptors, Interleukin-15/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/immunology , Transcription Factors/metabolism
17.
Immunity ; 42(6): 1048-61, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26070482

ABSTRACT

Thymic antigen-presenting cells (APCs) such as dendritic cells and medullary thymic epithelial cells (mTECs) use distinct strategies of self-antigen expression and presentation to mediate central tolerance. The thymus also harbors B cells; whether they also display unique tolerogenic features and how they genealogically relate to peripheral B cells is unclear. Here, we found that Aire is expressed in thymic but not peripheral B cells. Aire expression in thymic B cells coincided with major histocompatibility class II (MHCII) and CD80 upregulation and immunoglobulin class-switching. These features were recapitulated upon immigration of naive peripheral B cells into the thymus, whereby this intrathymic licensing required CD40 signaling in the context of cognate interactions with autoreactive CD4(+) thymocytes. Moreover, a licensing-dependent neo-antigen selectively upregulated in immigrating B cells mediated negative selection through direct presentation. Thus, autoreactivity within the nascent T cell repertoire fuels a feed forward loop that endows thymic B cells with tolerogenic features.


Subject(s)
B-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/metabolism , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , Antigen Presentation/genetics , Autoantigens/immunology , B7-1 Antigen/genetics , B7-1 Antigen/metabolism , CD40 Antigens/genetics , Cell Differentiation/genetics , Cells, Cultured , Central Tolerance/genetics , Clonal Selection, Antigen-Mediated/genetics , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Humans , Immunoglobulin Class Switching/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Signal Transduction , Transcription Factors/genetics , AIRE Protein
18.
Nature ; 563(7732): 564-568, 2018 11.
Article in English | MEDLINE | ID: mdl-30405245

ABSTRACT

Genetic regulators and environmental stimuli modulate T cell activation in autoimmunity and cancer. The enzyme co-factor tetrahydrobiopterin (BH4) is involved in the production of monoamine neurotransmitters, the generation of nitric oxide, and pain1,2. Here we uncover a link between these processes, identifying a fundamental role for BH4 in T cell biology. We find that genetic inactivation of GTP cyclohydrolase 1 (GCH1, the rate-limiting enzyme in the synthesis of BH4) and inhibition of sepiapterin reductase (the terminal enzyme in the synthetic pathway for BH4) severely impair the proliferation of mature mouse and human T cells. BH4 production in activated T cells is linked to alterations in iron metabolism and mitochondrial bioenergetics. In vivo blockade of BH4 synthesis abrogates T-cell-mediated autoimmunity and allergic inflammation, and enhancing BH4 levels through GCH1 overexpression augments responses by CD4- and CD8-expressing T cells, increasing their antitumour activity in vivo. Administration of BH4 to mice markedly reduces tumour growth and expands the population of intratumoral effector T cells. Kynurenine-a tryptophan metabolite that blocks antitumour immunity-inhibits T cell proliferation in a manner that can be rescued by BH4. Finally, we report the development of a potent SPR antagonist for possible clinical use. Our data uncover GCH1, SPR and their downstream metabolite BH4 as critical regulators of T cell biology that can be readily manipulated to either block autoimmunity or enhance anticancer immunity.


Subject(s)
Autoimmune Diseases/immunology , Biopterins/analogs & derivatives , Neoplasms/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Administration, Oral , Alcohol Oxidoreductases/antagonists & inhibitors , Alcohol Oxidoreductases/metabolism , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Biopterins/biosynthesis , Biopterins/metabolism , Biopterins/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Coenzymes/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , GTP Cyclohydrolase/genetics , GTP Cyclohydrolase/metabolism , Humans , Hypersensitivity/immunology , Iron/metabolism , Kynurenine/metabolism , Kynurenine/pharmacology , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
19.
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
20.
EMBO J ; 38(19): e101233, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31414712

ABSTRACT

Tissues in multicellular organisms are populated by resident macrophages, which perform both generic and tissue-specific functions. The latter are induced by signals from the microenvironment and rely on unique tissue-specific molecular programs requiring the combinatorial action of tissue-specific and broadly expressed transcriptional regulators. Here, we identify the transcription factors Bhlhe40 and Bhlhe41 as novel regulators of alveolar macrophages (AMs)-a population that provides the first line of immune defense and executes homeostatic functions in lung alveoli. In the absence of these factors, AMs exhibited decreased proliferation that resulted in a severe disadvantage of knockout AMs in a competitive setting. Gene expression analyses revealed a broad cell-intrinsic footprint of Bhlhe40/Bhlhe41 deficiency manifested by a downregulation of AM signature genes and induction of signature genes of other macrophage lineages. Genome-wide characterization of Bhlhe40 DNA binding suggested that these transcription factors directly repress the expression of lineage-inappropriate genes in AMs. Taken together, these results identify Bhlhe40 and Bhlhe41 as key regulators of AM self-renewal and guardians of their identity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Profiling/methods , Homeodomain Proteins/genetics , Macrophages, Alveolar/cytology , Acetylation , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Proliferation , Cell Self Renewal , Cell Survival , Down-Regulation , Gene Knockdown Techniques , Histones/metabolism , Homeodomain Proteins/metabolism , Macrophages, Alveolar/metabolism , Mice , Organ Specificity , Phenotype , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL