Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters

Publication year range
1.
Immunity ; 54(4): 702-720.e17, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33789089

ABSTRACT

Murine regulatory T (Treg) cells in tissues promote tissue homeostasis and regeneration. We sought to identify features that characterize human Treg cells with these functions in healthy tissues. Single-cell chromatin accessibility profiles of murine and human tissue Treg cells defined a conserved, microbiota-independent tissue-repair Treg signature with a prevailing footprint of the transcription factor BATF. This signature, combined with gene expression profiling and TCR fate mapping, identified a population of tissue-like Treg cells in human peripheral blood that expressed BATF, chemokine receptor CCR8 and HLA-DR. Human BATF+CCR8+ Treg cells from normal skin and adipose tissue shared features with nonlymphoid T follicular helper-like (Tfh-like) cells, and induction of a Tfh-like differentiation program in naive human Treg cells partially recapitulated tissue Treg regenerative characteristics, including wound healing potential. Human BATF+CCR8+ Treg cells from healthy tissue share features with tumor-resident Treg cells, highlighting the importance of understanding the context-specific functions of these cells.


Subject(s)
Chromatin/immunology , T-Lymphocytes, Regulatory/immunology , Wound Healing/immunology , Adult , Animals , Basic-Leucine Zipper Transcription Factors/immunology , Cell Differentiation/immunology , Cell Line , Female , Gene Expression Profiling/methods , Gene Expression Regulation/immunology , HaCaT Cells , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Receptors, CCR8/immunology , T Follicular Helper Cells/immunology
2.
Nat Immunol ; 16(1): 85-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25436972

ABSTRACT

Invariant natural killer T cells (iNKT cells) are lipid-sensing innate T cells that are restricted by the antigen-presenting molecule CD1d and express the transcription factor PLZF. iNKT cells accumulate in adipose tissue, where they are anti-inflammatory, but the factors that contribute to their anti-inflammatory nature, as well as their targets in adipose tissue, are unknown. Here we found that iNKT cells in adipose tissue had a unique transcriptional program and produced interleukin 2 (IL-2) and IL-10. Unlike other iNKT cells, they lacked PLZF but expressed the transcription factor E4BP4, which controlled their IL-10 production. The adipose iNKT cells were a tissue-resident population that induced an anti-inflammatory phenotype in macrophages and, through the production of IL-2, controlled the number, proliferation and suppressor function of regulatory T cells (Treg cells) in adipose tissue. Thus, iNKT cells in adipose tissue are unique regulators of immunological homeostasis in this tissue.


Subject(s)
Adipose Tissue/immunology , Kruppel-Like Transcription Factors/biosynthesis , Macrophages/immunology , Natural Killer T-Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , Adipose Tissue/cytology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Cell Growth Processes/immunology , Female , Flow Cytometry , Gene Expression Regulation , Homeostasis/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Macrophages/cytology , Male , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Promyelocytic Leukemia Zinc Finger Protein , Specific Pathogen-Free Organisms , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
3.
Nat Immunol ; 16(7): 708-17, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26054719

ABSTRACT

The transcription factors Batf3 and IRF8 are required for the development of CD8α(+) conventional dendritic cells (cDCs), but the basis for their actions has remained unclear. Here we identified two progenitor cells positive for the transcription factor Zbtb46 that separately generated CD8α(+) cDCs and CD4(+) cDCs and arose directly from the common DC progenitor (CDP). Irf8 expression in CDPs required prior autoactivation of Irf8 that was dependent on the transcription factor PU.1. Specification of the clonogenic progenitor of CD8α(+) cDCs (the pre-CD8 DC) required IRF8 but not Batf3. However, after specification of pre-CD8 DCs, autoactivation of Irf8 became Batf3 dependent at a CD8α(+) cDC-specific enhancer with multiple transcription factor AP1-IRF composite elements (AICEs) within the Irf8 superenhancer. CDPs from Batf3(-/-) mice that were specified toward development into pre-CD8 DCs failed to complete their development into CD8α(+) cDCs due to decay of Irf8 autoactivation and diverted to the CD4(+) cDC lineage.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Dendritic Cells/immunology , Interferon Regulatory Factors/immunology , Repressor Proteins/immunology , Stem Cells/immunology , Animals , Base Sequence , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD24 Antigen/immunology , CD24 Antigen/metabolism , CD8 Antigens/immunology , CD8 Antigens/metabolism , Cells, Cultured , Clone Cells/immunology , Clone Cells/metabolism , Dendritic Cells/metabolism , Flow Cytometry , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Protein Binding , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Homology, Nucleic Acid , Stem Cells/metabolism , Transcriptome/genetics , Transcriptome/immunology
4.
Nat Immunol ; 15(4): 373-83, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24584090

ABSTRACT

The transcription factor BATF is required for the differentiation of interleukin 17 (IL-17)-producing helper T cells (TH17 cells) and follicular helper T cells (TFH cells). Here we identified a fundamental role for BATF in regulating the differentiation of effector of CD8(+) T cells. BATF-deficient CD8(+) T cells showed profound defects in effector population expansion and underwent proliferative and metabolic catastrophe early after encountering antigen. BATF, together with the transcription factors IRF4 and Jun proteins, bound to and promoted early expression of genes encoding lineage-specific transcription-factors (T-bet and Blimp-1) and cytokine receptors while paradoxically repressing genes encoding effector molecules (IFN-γ and granzyme B). Thus, BATF amplifies T cell antigen receptor (TCR)-dependent expression of transcription factors and augments the propagation of inflammatory signals but restrains the expression of genes encoding effector molecules. This checkpoint prevents irreversible commitment to an effector fate until a critical threshold of downstream transcriptional activity has been achieved.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , T-Box Domain Proteins/metabolism , Th17 Cells/immunology , Transcription Factors/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Cell Differentiation/genetics , Cell Growth Processes/genetics , Cells, Cultured , Down-Regulation , Granzymes/genetics , Granzymes/metabolism , Interferon Regulatory Factors/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-jun/metabolism , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Transcriptional Activation/genetics
5.
Immunity ; 47(6): 1129-1141.e5, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29246443

ABSTRACT

During chronic stimulation, CD8+ T cells acquire an exhausted phenotype characterized by expression of inhibitory receptors, down-modulation of effector function, and metabolic impairments. T cell exhaustion protects from excessive immunopathology but limits clearance of virus-infected or tumor cells. We transcriptionally profiled antigen-specific T cells from mice infected with lymphocytic choriomeningitis virus strains that cause acute or chronic disease. T cell exhaustion during chronic infection was driven by high amounts of T cell receptor (TCR)-induced transcription factors IRF4, BATF, and NFATc1. These regulators promoted expression of inhibitory receptors, including PD-1, and mediated impaired cellular metabolism. Furthermore, they repressed the expression of TCF1, a transcription factor required for memory T cell differentiation. Reducing IRF4 expression restored the functional and metabolic properties of antigen-specific T cells and promoted memory-like T cell development. These findings indicate that IRF4 functions as a central node in a TCR-responsive transcriptional circuit that establishes and sustains T cell exhaustion during chronic infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon Regulatory Factors/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation , Gene Expression Regulation , HEK293 Cells , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/immunology , Humans , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Lymphocyte Activation , Lymphocyte Depletion , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/growth & development , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/genetics , Signal Transduction
6.
Immunity ; 47(6): 1100-1113.e6, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29262349

ABSTRACT

Natural killer (NK) cells are present in large populations at the maternal-fetal interface during early pregnancy. However, the role of NK cells in fetal growth is unclear. Here, we have identified a CD49a+Eomes+ subset of NK cells that secreted growth-promoting factors (GPFs), including pleiotrophin and osteoglycin, in both humans and mice. The crosstalk between HLA-G and ILT2 served as a stimulus for GPF-secreting function of this NK cell subset. Decreases in this GPF-secreting NK cell subset impaired fetal development, resulting in fetal growth restriction. The transcription factor Nfil3, but not T-bet, affected the function and the number of this decidual NK cell subset. Adoptive transfer of induced CD49a+Eomes+ NK cells reversed impaired fetal growth and rebuilt an appropriate local microenvironment. These findings reveal properties of NK cells in promoting fetal growth. In addition, this research proposes approaches for therapeutic administration of NK cells in order to reverse restricted nourishments within the uterine microenvironment during early pregnancy.


Subject(s)
Abortion, Habitual/immunology , Adoptive Transfer , Carrier Proteins/metabolism , Cytokines/metabolism , Fetal Development/immunology , Fetal Growth Retardation/prevention & control , Intercellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/transplantation , Abortion, Habitual/genetics , Abortion, Habitual/pathology , Adult , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Carrier Proteins/genetics , Carrier Proteins/immunology , Cellular Microenvironment , Cytokines/genetics , Cytokines/immunology , Decidua/immunology , Decidua/pathology , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/immunology , Fetal Growth Retardation/pathology , Fetus , Gene Expression Regulation, Developmental , HLA-G Antigens/genetics , HLA-G Antigens/immunology , Humans , Integrin alpha1/genetics , Integrin alpha1/immunology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Leukocyte Immunoglobulin-like Receptor B1/genetics , Leukocyte Immunoglobulin-like Receptor B1/immunology , Mice , Mice, Inbred C57BL , Pregnancy , Signal Transduction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology
7.
Immunity ; 44(4): 847-59, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27037189

ABSTRACT

Although antigen recognition mediated by the T cell receptor (TCR) influences many facets of Foxp3(+) regulatory T (Treg) cell biology, including development and function, the cell types that present antigen to Treg cells in vivo remain largely undefined. By tracking a clonal population of Aire-dependent, prostate-specific Treg cells in mice, we demonstrated an essential role for dendritic cells (DCs) in regulating organ-specific Treg cell biology. We have shown that the thymic development of prostate-specific Treg cells required antigen presentation by DCs. Moreover, Batf3-dependent CD8α(+) DCs were dispensable for the development of this clonotype and had negligible impact on the polyclonal Treg cell repertoire. In the periphery, CCR7-dependent migratory DCs coordinated the activation of organ-specific Treg cells in the prostate-draining lymph nodes. Our results demonstrate that the development and peripheral regulation of organ-specific Treg cells are dependent on antigen presentation by DCs, implicating DCs as key mediators of organ-specific immune tolerance.


Subject(s)
Antigen Presentation/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Dendritic Cells/immunology , Prostate/immunology , Repressor Proteins/genetics , Self Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B7-1 Antigen/biosynthesis , B7-1 Antigen/genetics , B7-2 Antigen/biosynthesis , B7-2 Antigen/genetics , Basic-Leucine Zipper Transcription Factors/immunology , CD8 Antigens/metabolism , Cell Differentiation/immunology , Cell Movement/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prostate/cytology , Receptors, Antigen, T-Cell/immunology , Receptors, CCR7/metabolism , Repressor Proteins/immunology , T-Lymphocytes, Regulatory/cytology , Transcription Factors/metabolism , AIRE Protein
8.
Nat Immunol ; 12(5): 450-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21460847

ABSTRACT

The immunoregulatory cytokine interleukin 10 (IL-10) is expressed mainly by T helper type 2 (T(H)2) cells but also by T(H)1 cells during chronic infection. Here we observed plasticity in the expression of IL-10 and IL-13 after chronic T(H)1 stimulation; furthermore, the expression of Il10 and Il13 was regulated by the transcription factor E4BP4. Chronically stimulated E4BP4-deficient (Nfil3(-/-); called 'E4bp4(-/-)' here) T(H)1 cells, regulatory T cells (T(reg) cells) and natural killer T cells (NKT cells) had attenuated expression of IL-10 and IL-13. Enforced expression of E4bp4 initiated the production of IL-10 and IL-13 by conventional T(H)1 cells. E4bp4(-/-) T(H)2 cells showed impairment of IL-10 production with no effect on IL-13. Our results indicate that E4BP4 has multiple functions in controlling the plasticity of IL-13 in T(H)1 cells and IL-10 in T(H)1 cells, T(H)2 cells, T(reg) cells and NKT cells.


Subject(s)
Autoimmune Diseases/immunology , Basic-Leucine Zipper Transcription Factors/immunology , CD4-Positive T-Lymphocytes/immunology , Interleukin-10/immunology , Interleukin-13/immunology , Animals , Electrophoretic Mobility Shift Assay , Flow Cytometry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic , Specific Pathogen-Free Organisms , Transcription, Genetic
10.
Nat Immunol ; 12(6): 536-43, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21572431

ABSTRACT

The transcription factor BATF controls the differentiation of interleukin 17 (IL-17)-producing helper T cells (T(H)17 cells) by regulating expression of the transcription factor RORγt itself and RORγt target genes such as Il17. Here we report the mechanism by which BATF controls in vivo class-switch recombination (CSR). In T cells, BATF directly controlled expression of the transcription factors Bcl-6 and c-Maf, both of which are needed for development of follicular helper T cells (T(FH) cells). Restoring T(FH) cell activity to Batf(-/-) T cells in vivo required coexpression of Bcl-6 and c-Maf. In B cells, BATF directly controlled the expression of both activation-induced cytidine deaminase (AID) and of germline transcripts of the intervening heavy-chain region and constant heavy-chain region (I(H)-C(H)). Thus, BATF functions at multiple hierarchical levels in two cell types to globally regulate switched antibody responses in vivo.


Subject(s)
B-Lymphocytes/immunology , Basic-Leucine Zipper Transcription Factors/immunology , Immunoglobulin Class Switching/immunology , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing , Animals , B-Lymphocytes/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , CD40 Ligand/genetics , CD40 Ligand/immunology , CD40 Ligand/metabolism , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cells, Cultured , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Female , Flow Cytometry , Gene Expression Profiling , Germinal Center/immunology , Germinal Center/metabolism , Immunoglobulin Class Switching/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , Proto-Oncogene Proteins c-bcl-6/metabolism , Recombination, Genetic , T-Lymphocytes/metabolism
11.
Proc Natl Acad Sci U S A ; 117(24): 13740-13749, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32471947

ABSTRACT

Human T cell leukemia virus type 1 (HTLV-1) is the etiologic agent of a T cell neoplasm and several inflammatory diseases. A viral gene, HTLV-1 bZIP factor (HBZ), induces pathogenic Foxp3-expressing T cells and triggers systemic inflammation and T cell lymphoma in transgenic mice, indicating its significance in HTLV-1-associated diseases. Here we show that, unexpectedly, a proinflammatory cytokine, IL-6, counteracts HBZ-mediated pathogenesis. Loss of IL-6 accelerates inflammation and lymphomagenesis in HBZ transgenic mice. IL-6 innately inhibits regulatory T cell differentiation, suggesting that IL-6 functions as a suppressor against HBZ-associated complications. HBZ up-regulates expression of the immunosuppressive cytokine IL-10. IL-10 promotes T cell proliferation only in the presence of HBZ. As a mechanism of growth promotion by IL-10, HBZ interacts with STAT1 and STAT3 and modulates the IL-10/JAK/STAT signaling pathway. These findings suggest that HTLV-1 promotes the proliferation of infected T cells by hijacking the machinery of regulatory T cell differentiation. IL-10 induced by HBZ likely suppresses the host immune response and concurrently promotes the proliferation of HTLV-1 infected T cells.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , HTLV-I Infections/immunology , Human T-lymphotropic virus 1/immunology , Interleukin-6/immunology , Lymphoma/virology , Retroviridae Proteins/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation , Cell Proliferation , HTLV-I Infections/genetics , HTLV-I Infections/pathology , HTLV-I Infections/virology , Host-Pathogen Interactions , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 1/physiology , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Retroviridae Proteins/genetics , T-Lymphocytes, Regulatory/immunology
12.
Plant Cell Environ ; 45(6): 1843-1861, 2022 06.
Article in English | MEDLINE | ID: mdl-35199374

ABSTRACT

Stomatal movement participates in plant immunity by directly affecting the invasion of bacteria, but the genes that regulate stomatal immunity have not been well identified. Here, we characterised the function of the bZIP59 transcription factor from Arabidopsis thaliana, which is constitutively expressed in guard cells. The bzip59 mutant is partially impaired in stomatal closure induced by Pseudomonas syringae pv. tomato strain (Pst) DC3000 and is more susceptible to Pst DC3000 infection. By contrast, the line overexpressing bZIP59 enhances resistance to Pst DC3000 infection. Furthermore, the bzip59 mutant is also partially impaired in stomatal closure induced by flagellin flg22 derived from Pst DC3000, and epistasis analysis revealed that bZIP59 acts upstream of reactive oxygen species (ROS) and nitric oxide (NO) and downstream of salicylic acid signalling in flg22-induced stomatal closure. In addition, the bzip59 mutant showed resistance and sensitivity to Sclerotinia sclerotiorum and Tobacco mosaic virus that do not invade through stomata, respectively. Collectively, our results demonstrate that bZIP59 plays an important role in the stomatal immunity and reveal that the same transcription factor can positively and negatively regulate disease resistance against different pathogens.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Basic-Leucine Zipper Transcription Factors , Solanum lycopersicum , Arabidopsis/genetics , Arabidopsis/immunology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Solanum lycopersicum/metabolism , Plant Diseases/genetics , Plant Diseases/immunology , Plant Diseases/microbiology , Plant Immunity , Plant Stomata/genetics , Plant Stomata/immunology , Pseudomonas syringae/physiology , Transcription Factors/genetics
13.
Immunity ; 39(1): 89-91, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23890066

ABSTRACT

Activation of IgG⁺ memory B cells accounts for much of the antibodies in secondary immune responses. Here, Kometani et al. (2013) demonstrate that reduced amounts of Bach2 in antigen-experienced memory B cells control the robust production of IgG1⁺ plasma cells.


Subject(s)
B-Lymphocytes/immunology , Basic-Leucine Zipper Transcription Factors/immunology , Cell Differentiation/immunology , Plasma Cells/immunology , Animals
14.
Immunity ; 39(1): 136-47, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23850379

ABSTRACT

Memory B cells are essential for generating rapid and robust secondary antibody responses. It has been thought that the unique cytoplasmic domain of IgG causes the prompt activation of antigen-experienced IgG memory B cells. To assess this model, we have generated a mouse containing IgG1 B cells that have never encountered antigen. We found that, upon challenge, antigen-experienced IgG1 memory B cells rapidly differentiated into plasma cells, whereas nonexperienced IgG1 B cells did not, suggesting the importance of the stimulation history. In addition, our results suggest that repression of the Bach2 transcription factor, which results from antigen experience, contributes to predisposition of IgG1 memory B cells to differentiate into plasma cells.


Subject(s)
B-Lymphocytes/immunology , Basic-Leucine Zipper Transcription Factors/immunology , Cell Differentiation/immunology , Plasma Cells/immunology , Animals , B-Lymphocytes/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/genetics , Cells, Cultured , Flow Cytometry , Gene Expression/immunology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunologic Memory/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/immunology , PAX5 Transcription Factor/metabolism , Plasma Cells/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
15.
J Immunol ; 205(4): 901-906, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32669309

ABSTRACT

The basic leucine zipper transcription factor ATF-like 3 (BATF3) is required for the development of conventional type 1 dendritic cells that are essential for cross-presentation and CD8 T cell-mediated immunity against intracellular pathogens and tumors. However, whether BATF3 intrinsically regulates CD8 T cell responses is not well studied. In this article, we report a role for cell-intrinsic Batf3 expression in regulating the establishment of circulating and resident memory T cells after foodborne Listeria monocytogenes infection of mice. Consistent with other studies, Batf3 expression by CD8 T cells was dispensable for the primary response. However, Batf3 -/- T cells underwent increased apoptosis during contraction to contribute to a substantially reduced memory population. Batf3 -/- memory cells had an impaired ability to mount a robust recall response but remained functional. These findings reveal a cell-intrinsic role of Batf3 in regulating CD8 T cell memory development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory/immunology , Repressor Proteins/immunology , Repressor Proteins/metabolism , Animals , Apoptosis/immunology , Cells, Cultured , Cross-Priming/immunology , Female , Immunity, Cellular/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/metabolism , Male , Mice , Mice, Inbred C57BL
16.
J Allergy Clin Immunol ; 147(1): 280-295, 2021 01.
Article in English | MEDLINE | ID: mdl-33069715

ABSTRACT

BACKGROUND: This study group has previously identified IL-9-producing mucosal mast cell (MMC9) as the primary source of IL-9 to drive intestinal mastocytosis and experimental IgE-mediated food allergy. However, the molecular mechanisms that regulate the expansion of MMC9s remain unknown. OBJECTIVES: This study hypothesized that IL-4 regulates MMC9 development and MMC9-dependent experimental IgE-mediated food allergy. METHODS: An epicutaneous sensitization model was used and bone marrow reconstitution experiments were performed to test the requirement of IL-4 receptor α (IL-4Rα) signaling on MMC9s in experimental IgE-mediated food allergy. Flow cytometric, bulk, and single-cell RNA-sequencing analyses on small intestine (SI) MMC9s were performed to illuminate MMC9 transcriptional signature and the effect of IL-4Rα signaling on MMC9 function. A bone marrow-derived MMC9 culture system was used to define IL-4-BATF signaling in MMC9 development. RESULTS: Epicutaneous sensitization- and bone marrow reconstitution-based models of IgE-mediated food allergy revealed an IL-4 signaling-dependent cell-intrinsic effect on SI MMC9 accumulation and food allergy severity. RNA-sequencing analysis of SI-MMC9s identified 410 gene transcripts reciprocally regulated by IL-4 signaling, including Il9 and Batf. Insilico analyses identified a 3491-gene MMC9 transcriptional signature and identified 2 transcriptionally distinct SI MMC9 populations enriched for metabolic or inflammatory programs. Employing an in vitro MMC9-culture model system showed that generation of MMC9-like cells was induced by IL-4 and this was in part dependent on BATF. CONCLUSIONS: IL-4Rα signaling directly modulates MMC9 function and exacerbation of experimental IgE-mediated food allergic reactions. IL-4Rα regulation of MMC9s is in part BATF-dependent and occurs via modulation of metabolic transcriptional programs.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Food Hypersensitivity/immunology , Interleukin-4/immunology , Interleukin-9/immunology , Intestinal Mucosa/immunology , Mast Cells/immunology , Signal Transduction/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Disease Models, Animal , Food Hypersensitivity/genetics , Food Hypersensitivity/pathology , Interleukin-4/genetics , Interleukin-9/genetics , Intestinal Mucosa/pathology , Mast Cells/pathology , Mice , Mice, Knockout , Signal Transduction/genetics
17.
Cancer Sci ; 112(5): 1688-1694, 2021 May.
Article in English | MEDLINE | ID: mdl-33630351

ABSTRACT

Adult T-cell leukemia-lymphoma (ATL) is caused by human T-cell leukemia virus type 1 (HTLV-1) infection. Among HTLV-1 encoded genes, HTLV-1 bZIP factor (HBZ) and tax are critical for the leukemogenesis of ATL. Adult T-cell leukemia-lymphoma needs a long latent period before onset, indicating that both viral genes and alterations (genetic and epigenetic) of the host genome play important roles for leukemogenesis. Viral genes influence genetic and epigenetic changes of the host genome, indicating that the virus is of primary importance in leukemogenesis. HBZ is expressed in all ATL cases, whereas Tax expression is heterogeneous among ATL cases. Different patterns of viral gene expression in tumors are also observed for Epstein-Barr virus. We propose three subtypes of ATL cases based on Tax expression: high, intermittent, and lost expression. HBZ is detected in all ATL cases. Approximately 25% of all ATL cases lost Tax expression at infection of HTLV-1, indicating that HBZ is the only viral gene responsible for leukemogenesis in addition to genetic and epigenetic changes of the host genes in these ATL cases. The host immune responses to Tax are also implicated in the heterogeneity of ATL. Thus, ATL is a heterogeneous disease in terms of its viral gene expression, which is important for pathogenesis of this intractable lymphomatous neoplasm.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Gene Products, tax/genetics , Herpesvirus 4, Human/genetics , Human T-lymphotropic virus 1/genetics , Leukemia-Lymphoma, Adult T-Cell/virology , Retroviridae Proteins/genetics , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , B-Lymphocytes/virology , Basic-Leucine Zipper Transcription Factors/immunology , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Proliferation , Cell Transformation, Viral , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Leukemic , Gene Expression Regulation, Viral , Gene Products, tax/immunology , Gene Products, tax/metabolism , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukemia-Lymphoma, Adult T-Cell/metabolism , Mice , Mice, Transgenic , RNA, Viral/genetics , Retroviridae Proteins/immunology , Retroviridae Proteins/metabolism , T-Lymphocytes, Regulatory/virology , Virus Replication/genetics
18.
Eur J Immunol ; 50(3): 426-438, 2020 03.
Article in English | MEDLINE | ID: mdl-31808546

ABSTRACT

Although CD4+ CD45RA- Foxp3l ° cytokine-secreting T cells (Fr.III cells) have been reported to be increased in systemic lupus erythematosus (SLE), their function and effects on response of B cells are still unclear. Here, we dissect how BACH2 regulates Fr.III cells function and promotes B-cell response in active SLE patients. We measured cytokines and BACH2 expression, and found that Fr.III cells from SLE patients produce much more inflammatory cytokines and were more able to promote B- cell proliferation, IgG, IgA, and TNF-α production than controls in a co-culture system. Fr.III cells expressed high levels of ICOS and CD154, but a low level of Tfr and BACH2, BACH2 expression was negatively correlated with SLE Disease Activity Index. Overexpressed of BACH2 in Fr.III cells, decreased cytokines expression and reduced B-cell response. Furthermore, we identified a reduction of H3K27ac level binding at the BACH2 locus in the SLE Fr.III cells and SLE serum stimulation decreased H3K27ac binding at the BACH2 locus, which could be restored using trichostatin A (TSA). In conclusion, BACH2 was associated with SLE disease activity, regulated the function of Fr.III cells, and promoted B-cells response. Targeting BACH2 may be a new immune intervention therapy of SLE.


Subject(s)
B-Lymphocytes/immunology , Basic-Leucine Zipper Transcription Factors/immunology , Lupus Erythematosus, Systemic/immunology , T-Lymphocyte Subsets/immunology , Adult , Female , Humans , Lymphocyte Activation/immunology , Male , Middle Aged
19.
PLoS Pathog ; 15(6): e1007866, 2019 06.
Article in English | MEDLINE | ID: mdl-31188899

ABSTRACT

The gastric lamina propria of mice that have been experimentally infected with the pathobiont Helicobacter pylori hosts a dense network of myeloid cells that includes BATF3-dependent CD103+ dendritic cells (DCs). We show here that CD103+ DCs are strictly required for gastric Th1 responses to H. pylori and for H. pylori infection control. A similar dependence of type 1 immunity on CD103+ DCs is observed in a Mycobacterium bovis BCG infection model, and in a syngeneic colon cancer model. Strikingly, we find that not only the expansion and/or recruitment of Th1 cells, but also of peripherally induced, neuropilin-negative regulatory T-cells to sites of infection requires BATF3-dependent DCs. A shared feature of the examined models is the strongly reduced production of the chemokines and CXCR3 ligands CXCL9, 10 and 11 in BATF3-deficient mice. The results implicate BATF3-dependent DCs in the recruitment of CXCR3+ effector and regulatory T-cells to target tissues and in their local expansion.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Dendritic Cells/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Mycobacterium bovis/immunology , Repressor Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Tuberculosis/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Cell Line, Tumor , Chemokines, CXC/genetics , Chemokines, CXC/immunology , Dendritic Cells/microbiology , Dendritic Cells/pathology , Helicobacter Infections/genetics , Helicobacter Infections/pathology , Mice , Mice, Knockout , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Repressor Proteins/genetics , T-Lymphocytes, Regulatory/microbiology , T-Lymphocytes, Regulatory/pathology , Tuberculosis/genetics , Tuberculosis/pathology
20.
Cell Tissue Res ; 384(2): 423-434, 2021 May.
Article in English | MEDLINE | ID: mdl-33447883

ABSTRACT

MiR-150-5p is an immune-related miRNA and elevated in the plasma of patients with aplastic anemia (AA), but its role in T cell activation in patients with severe aplastic anemia (SAA) is unclear. This study aims to explore the role of miR-150-5p in T cell activation of SAA. RT-PCR and Western blot were used to detect the expression of mRNA and protein. The cell proportion was detected by flow cytometry. The lentiviruses expressing miR-150-5p inhibitor and Bach2 shRNA were respectively infected to produce stable miR-150-5p or Bach2 knockout cells. Brdu incorporation method was used to detect T cell proliferation. SAA mouse model was induced with cyclophosphamide and busulfan, and intravenous injection of LV inhibitor NC and LV-miR-150-5p inhibitor. The miR-150-5p expression is up-regulated in SAA, which is negatively correlated with Bach2. Inhibition of miR-150-5p reduces the activation of T cells. MiR-150-5p directly targeted 3'UTR of Bach2. Moreover, the expression of miR-150-5p and the activation of T cells were decreased in SAA mouse model. MiR-150-5p promotes T cell activation in SAA by targeting Bach2. Targeting miR-150-5p may be a new molecular therapy for SAA patients.


Subject(s)
Anemia, Aplastic/immunology , Basic-Leucine Zipper Transcription Factors/immunology , MicroRNAs/immunology , T-Lymphocytes/immunology , Anemia, Aplastic/metabolism , Anemia, Aplastic/pathology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Disease Models, Animal , Humans , Lymphocyte Activation , MicroRNAs/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL