Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Int Immunol ; 36(4): 167-182, 2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38169425

ABSTRACT

Forkhead box P3 (Foxp3)-expressing regulatory T (Treg) cells play essential roles in immune homeostasis but also contribute to establish a favorable environment for tumor growth by suppressing anti-tumor immune responses. It is thus necessary to specifically target tumor-infiltrating Treg cells to minimize effects on immune homeostasis in cancer immunotherapy. However, molecular features that distinguish tumor-infiltrating Treg cells from those in secondary lymphoid organs remain unknown. Here we characterize distinct features of tumor-infiltrating Treg cells by global analyses of the transcriptome and chromatin landscape. They exhibited activated phenotypes with enhanced Foxp3-dependent transcriptional regulation, yet being distinct from activated Treg cells in secondary lymphoid organs. Such differences may be attributed to the extensive clonal expansion of tumor-infiltrating Treg cells. Moreover, we found that TCF7 and LEF1 were specifically downregulated in tumor-infiltrating Treg cells both in mice and humans. These factors and Foxp3 co-occupied Treg suppressive function-related gene loci in secondary lymphoid organ Treg cells, whereas the absence of TCF7 and LEF1 accompanied altered gene expression and chromatin status at these gene loci in tumor-infiltrating Treg cells. Functionally, overexpression of TCF7 and LEF1 in Treg cells inhibited the enhancement of Treg suppressive function upon activation. Our results thus show the downregulation of TCF7 and LEF1 as markers of highly suppressive Treg cells in tumors and suggest that their absence controls the augmentation of Treg suppressive function in tumors. These molecules may be potential targets for novel cancer immunotherapy with minimum effects on immune homeostasis.


Subject(s)
Neoplasms , T-Lymphocytes, Regulatory , Humans , Animals , Mice , Down-Regulation , Forkhead Transcription Factors/metabolism , Chromatin/metabolism , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism
2.
iScience ; 26(10): 107893, 2023 Oct 20.
Article in English | MEDLINE | ID: mdl-37771659

ABSTRACT

Cell differentiation is achieved by acquiring a cell type-specific transcriptional program and epigenetic landscape. While the cell type-specific patterning of enhancers has been shown to precede cell fate decisions, it remains unclear how regulators of these enhancers are induced to initiate cell specification and how they appropriately restrict cells that differentiate. Here, using embryonic stem cell-derived hematopoietic cell differentiation cultures, we show the activation of some hematopoietic enhancers during arterialization of hemogenic endothelium, a prerequisite for hematopoiesis. We further reveal that ZEB2, a factor involved in the transcriptional regulation of arterial endothelial cells, and a hematopoietic regulator MEIS1 are independently required for activating these enhancers. Concomitantly, ZEB2 or MEIS1 deficiency impaired hematopoietic cell development. These results suggest that multiple regulators expressed from an earlier developmental stage non-redundantly contribute to the establishment of hematopoietic enhancer landscape, thereby restricting cell differentiation despite the unrestricted expression of these regulators to hematopoietic cells.

3.
Front Cell Dev Biol ; 11: 1022081, 2023.
Article in English | MEDLINE | ID: mdl-37274738

ABSTRACT

Skeletal muscle has a highly regenerative capacity, but the detailed process is not fully understood. Several in vitro skeletal muscle regeneration models have been developed to elucidate this, all of which rely on specialized culture conditions that limit the accessibility and their application to many general experiments. Here, we established a concise in vitro skeletal muscle regeneration model using mouse primary cells. This model allows evaluation of skeletal muscle regeneration in two-dimensional culture system similar to a typical cell culture, showing a macrophage-dependent regenerative capacity, which is an important process in skeletal muscle regeneration. Based on the concept that this model could assess the contribution of macrophages of various phenotypes to skeletal muscle regeneration, we evaluated the effect of endotoxin pre-stimulation for inducing various changes in gene expression on macrophages and found that the contribution to skeletal muscle regeneration was significantly reduced. The gene expression patterns differed from those of naive macrophages, especially immediately after skeletal muscle injury, suggesting that the difference in responsiveness contributed to the difference in regenerative efficiency. Our findings provide a concise in vitro model that enables the evaluation of the contribution of individual cell types, such as macrophages and muscle stem cells, on skeletal muscle regeneration.

4.
Life Sci Alliance ; 6(3)2023 03.
Article in English | MEDLINE | ID: mdl-36604149

ABSTRACT

Spinal muscular atrophy (SMA) is a congenital neuromuscular disease caused by the mutation or deletion of the survival motor neuron 1 (SMN1) gene. Although the primary cause of progressive muscle atrophy in SMA has classically been considered the degeneration of motor neurons, recent studies have indicated a skeletal muscle-specific pathological phenotype such as impaired mitochondrial function and enhanced cell death. Here, we found that the down-regulation of SMN causes mitochondrial dysfunction and subsequent cell death in in vitro models of skeletal myogenesis with both a murine C2C12 cell line and human induced pluripotent stem cells. During myogenesis, SMN binds to the upstream genomic regions of MYOD1 and microRNA (miR)-1 and miR-206. Accordingly, the loss of SMN down-regulates these miRs, whereas supplementation of the miRs recovers the mitochondrial function, cell survival, and myotube formation of SMN-deficient C2C12, indicating the SMN-miR axis is essential for myogenic metabolic maturation. In addition, the introduction of the miRs into ex vivo muscle stem cells derived from Δ7-SMA mice caused myotube formation and muscle contraction. In conclusion, our data revealed novel transcriptional roles of SMN during myogenesis, providing an alternative muscle-oriented therapeutic strategy for SMA patients.


Subject(s)
Induced Pluripotent Stem Cells , MicroRNAs , Muscular Atrophy, Spinal , Survival of Motor Neuron 1 Protein , Animals , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/metabolism , Muscle Development/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism
5.
J Exp Med ; 220(2)2023 02 06.
Article in English | MEDLINE | ID: mdl-36454183

ABSTRACT

Thymic selection and peripheral activation of conventional T (Tconv) and regulatory T (Treg) cells depend on TCR signaling, whose anomalies are causative of autoimmunity. Here, we expressed in normal mice mutated ZAP-70 molecules with different affinities for the CD3 chains, or wild type ZAP-70 at graded expression levels under tetracycline-inducible control. Both manipulations reduced TCR signaling intensity to various extents and thereby rendered those normally deleted self-reactive thymocytes to become positively selected and form a highly autoimmune TCR repertoire. The signal reduction more profoundly affected Treg development and function because their TCR signaling was further attenuated by Foxp3 that physiologically repressed the expression of TCR-proximal signaling molecules, including ZAP-70, upon TCR stimulation. Consequently, the TCR signaling intensity reduced to a critical range generated pathogenic autoimmune Tconv cells and concurrently impaired Treg development/function, leading to spontaneous occurrence of autoimmune/inflammatory diseases, such as autoimmune arthritis and inflammatory bowel disease. These results provide a general model of how altered TCR signaling evokes autoimmune disease.


Subject(s)
Autoimmune Diseases , Animals , Mice , Autoimmunity , Signal Transduction , T-Lymphocytes, Regulatory , Receptors, Antigen, T-Cell
6.
J Allergy Clin Immunol ; 149(1): 176-188.e7, 2022 01.
Article in English | MEDLINE | ID: mdl-34175136

ABSTRACT

BACKGROUND: Blau syndrome (BS) is an autoinflammatory disease associated with mutations in nucleotide-binding oligomerization domain 2. Although treatments with anti-TNF agents have been reported to be effective, the underlying molecular mechanisms remain unclear. OBJECTIVE: We aimed to elucidate the mechanisms of autoinflammation in patients with BS and to clarify how anti-TNF treatment controls the disease phenotype at the cellular level in clinical samples. METHODS: Macrophages were differentiated from monocytes of 7 BS patients, and global transcriptional profiles of 5 patients were analyzed with or without IFN-γ stimulation. Macrophages were also generated from BS-specific induced pluripotent stem cells (iPSCs), and their transcriptome was examined for comparison. RESULTS: Aberrant inflammatory responses were observed upon IFN-γ stimulation in macrophages from untreated BS patients, but not in those from patients treated with anti-TNF. iPSC-derived macrophages carrying a disease-associated mutation also showed IFN-γ-dependent accelerated inflammatory responses. Comparisons of peripheral blood- and iPSC-derived macrophages revealed the upregulation of nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) targets in unstimulated macrophages as a common feature. CONCLUSIONS: IFN-γ stimulation is one of the key signals driving aberrant inflammatory responses in BS-associated macrophages. However, long-term treatment with anti-TNF agents ameliorates such abnormalities even in the presence of IFN-γ stimulation. Our data thus suggest that preexposure to TNF or functionally similar cytokines inducing NF-κB-driven proinflammatory signaling during macrophage development is a prerequisite for accelerated inflammatory responses upon IFN-γ stimulation in BS.


Subject(s)
Arthritis/immunology , Interferon-gamma/immunology , Macrophages/immunology , Sarcoidosis/immunology , Synovitis/immunology , Tumor Necrosis Factor Inhibitors/pharmacology , Uveitis/immunology , Adult , Arthritis/drug therapy , Arthritis/genetics , Cell Line , Child , Child, Preschool , Female , Humans , Induced Pluripotent Stem Cells/cytology , Male , NF-kappa B/immunology , Sarcoidosis/drug therapy , Sarcoidosis/genetics , Synovitis/drug therapy , Synovitis/genetics , Transcriptome , Tumor Necrosis Factor Inhibitors/therapeutic use , Uveitis/drug therapy , Uveitis/genetics , Young Adult
7.
Immunity ; 54(5): 947-961.e8, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33930308

ABSTRACT

The transcription factor Foxp3 plays crucial roles for Treg cell development and function. Conserved non-coding sequences (CNSs) at the Foxp3 locus control Foxp3 transcription, but how they developmentally contribute to Treg cell lineage specification remains obscure. Here, we show that among Foxp3 CNSs, the promoter-upstream CNS0 and the intergenic CNS3, which bind distinct transcription factors, were activated at early stages of thymocyte differentiation prior to Foxp3 promoter activation, with sequential genomic looping bridging these regions and the promoter. While deletion of either CNS0 or CNS3 partially compromised thymic Treg cell generation, deletion of both completely abrogated the generation and impaired the stability of Foxp3 expression in residual Treg cells. As a result, CNS0 and CNS3 double-deleted mice succumbed to lethal systemic autoimmunity and inflammation. Thus, hierarchical and coordinated activation of Foxp3 CNS0 and CNS3 initiates and stabilizes Foxp3 gene expression, thereby crucially controlling Treg cell development, maintenance, and consequently immunological self-tolerance.


Subject(s)
Enhancer Elements, Genetic/immunology , Forkhead Transcription Factors/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Gene Expression Regulation/immunology , Humans , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/immunology , Self Tolerance/immunology
8.
Immunity ; 52(6): 1119-1132.e4, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32362325

ABSTRACT

The contribution of FOXP3-expressing naturally occurring regulatory T (Treg) cells to common polygenic autoimmune diseases remains ambiguous. Here, we characterized genome-wide epigenetic profiles (CpG methylation and histone modifications) of human Treg and conventional T (Tconv) cells in naive and activated states. We found that single-nucleotide polymorphisms (SNPs) associated with common autoimmune diseases were predominantly enriched in CpG demethylated regions (DRs) specifically present in naive Treg cells but much less enriched in activation-induced DRs common in Tconv and Treg cells. Naive Treg cell-specific DRs were largely included in Treg cell-specific super-enhancers and closely associated with transcription and other epigenetic changes in naive and effector Treg cells. Thus, naive Treg cell-specific CpG hypomethylation had a key role in controlling Treg cell-specific gene transcription and epigenetic modification. The results suggest possible contribution of altered function or development of natural Treg cells to the susceptibility to common autoimmune diseases.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Epigenesis, Genetic , Epigenomics , Genetic Predisposition to Disease , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Biomarkers , Cell Differentiation/genetics , Cell Differentiation/immunology , Computational Biology , CpG Islands , DNA Methylation , Epigenomics/methods , Gene Expression Profiling , Genetic Variation , Humans , Immunophenotyping , Polymorphism, Single Nucleotide , T-Lymphocyte Subsets , T-Lymphocytes, Regulatory/cytology , Transcriptome
9.
Front Immunol ; 10: 2382, 2019.
Article in English | MEDLINE | ID: mdl-31681278

ABSTRACT

Regulatory T (Treg) cells mainly develop within the thymus and arise from CD25+Foxp3- (CD25+ TregP) or CD25-Foxp3+ (Foxp3+ TregP) Treg cell precursors resulting in Treg cells harboring distinct transcriptomic profiles and complementary T cell receptor repertoires. The stable and long-term expression of Foxp3 in Treg cells and their stable suppressive phenotype are controlled by the demethylation of Treg cell-specific epigenetic signature genes including an evolutionarily conserved CpG-rich element within the Foxp3 locus, the Treg-specific demethylated region (TSDR). Here we analyzed the dynamics of the imprinting of the Treg cell-specific epigenetic signature genes in thymic Treg cells. We could demonstrate that CD25+Foxp3+ Treg cells show a progressive demethylation of most signature genes during maturation within the thymus. Interestingly, a partial demethylation of several Treg cell-specific epigenetic signature genes was already observed in Foxp3+ TregP but not in CD25+ TregP. Furthermore, Foxp3+ TregP were very transient in nature and arose at a more mature developmental stage when compared to CD25+ TregP. When the two Treg cell precursors were cultured in presence of IL-2, a factor known to be critical for thymic Treg cell development, we observed a major impact of IL-2 on the demethylation of the TSDR with a more pronounced effect on Foxp3+ TregP. Together, these results suggest that the establishment of the Treg cell-specific hypomethylation pattern is a continuous process throughout thymic Treg cell development and that the two known Treg cell precursors display distinct dynamics for the imprinting of the Treg cell-specific epigenetic signature genes.


Subject(s)
DNA Methylation , Genetic Loci/immunology , Genomic Imprinting/immunology , T-Lymphocytes, Regulatory/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , DNA Methylation/genetics , DNA Methylation/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Mice , Mice, Transgenic , T-Lymphocytes, Regulatory/cytology , Thymocytes/cytology , Thymus Gland/cytology
10.
Sci Immunol ; 4(40)2019 10 25.
Article in English | MEDLINE | ID: mdl-31653719

ABSTRACT

A promising way to restrain hazardous immune responses, such as autoimmune disease and allergy, is to convert disease-mediating T cells into immunosuppressive regulatory T (Treg) cells. Here, we show that chemical inhibition of the cyclin-dependent kinase 8 (CDK8) and CDK19, or knockdown/knockout of the CDK8 or CDK19 gene, is able to induce Foxp3, a key transcription factor controlling Treg cell function, in antigen-stimulated effector/memory as well as naïve CD4+ and CD8+ T cells. The induction was associated with STAT5 activation, independent of TGF-ß action, and not affected by inflammatory cytokines. Furthermore, in vivo administration of a newly developed CDK8/19 inhibitor along with antigen immunization generated functionally stable antigen-specific Foxp3+ Treg cells, which effectively suppressed skin contact hypersensitivity and autoimmune disease in animal models. The results indicate that CDK8/19 is physiologically repressing Foxp3 expression in activated conventional T cells and that its pharmacological inhibition enables conversion of antigen-specific effector/memory T cells into Foxp3+ Treg cells for the treatment of various immunological diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/immunology , Cells, Cultured , Cyclin-Dependent Kinase 8/deficiency , Cyclin-Dependent Kinase 8/immunology , Cyclin-Dependent Kinases/deficiency , Cyclin-Dependent Kinases/immunology , Forkhead Transcription Factors/immunology , Immunologic Memory/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic
11.
Int Immunol ; 31(5): 335-347, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30726915

ABSTRACT

Ten-eleven translocation (TET) proteins regulate DNA methylation and gene expression by converting 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). Although Tet2/Tet3 deficiency has been reported to lead to myeloid cell, B-cell and invariant natural killer T (iNKT) cell malignancy, the effect of TET on regulatory T cells (Tregs) has not been elucidated. We found that Tet2/Tet3 deficiency in Tregs led to lethal hyperproliferation of CD4+Foxp3+ T cells in the spleen and mesenteric lymph nodes after 5 months of age. Additionally, in aged Treg-specific Tet2/Tet3-deficient mice, serum IgG1, IgG3, IgM and IgE levels were markedly elevated. High IL-17 expression was observed in both Foxp3+ and Fopx3- CD4+ T cells, and adoptive transfer of Tet2/Tet3-deficient Tregs into lymphopenic mice inhibited Foxp3 expression and caused conversion into IL-17-producing cells. However, the conserved non-coding DNA sequence-2 (CNS2) region of the Foxp3 gene locus, which has been shown to be particularly important for stable Foxp3 expression, was only partly methylated. We identified novel TET-dependent demethylation sites in the Foxp3 upstream enhancer, which may contribute to stable Foxp3 expression. Together, these data indicate that Tet2 and Tet3 are involved in Treg stability and immune homeostasis in mice.


Subject(s)
DNA-Binding Proteins/immunology , Dioxygenases/immunology , Forkhead Transcription Factors/metabolism , Interleukin-17/biosynthesis , Proto-Oncogene Proteins/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Animals , Cell Proliferation , Interleukin-17/immunology , Mice , Mice, Inbred C57BL
12.
Nat Commun ; 10(1): 549, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30710091

ABSTRACT

The genome organizer, special AT-rich sequence-binding protein-1 (Satb1), plays a pivotal role in the regulation of global gene networks in a cell type-dependent manner and is indispensable for the development of multiple cell types, including mature CD4+ T, CD8+ T, and Foxp3+ regulatory T cells in the thymus. However, it remains unknown how the differentiation and effector program of the Th subsets in the periphery are regulated by Satb1. Here, we demonstrate that Satb1 differentially regulates gene expression profiles in non-pathogenic and pathogenic Th17 cells and promotes the pathogenic effector program of encephalitogenic Th17 cells by regulating GM-CSF via Bhlhe40 and inhibiting PD-1 expression. However, Satb1 is dispensable for the differentiation and non-pathogenic functions of Th17 cells. These results indicate that Satb1 regulates the specific gene expression and function of effector Th17 cells in tissue inflammation.


Subject(s)
Inflammation/immunology , Matrix Attachment Region Binding Proteins/metabolism , Th17 Cells/immunology , Animals , Biomarkers/metabolism , Encephalitis , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Genetic Loci , Inflammation/pathology , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Spinal Cord/metabolism
13.
Curr Opin Immunol ; 49: 64-70, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29065384

ABSTRACT

Treg cells expressing the transcription factor Foxp3 are essential for immunological tolerance and homeostasis. Recent genome-wide studies have revealed that Foxp3+ natural Treg cells possess a number of unique transcriptional and epigenetic features, which appear to be acquired along the course of Treg cell development and maintained throughout their lifespan. These studies also provide novel insights into how genomic variations contribute to genetic susceptibility to human autoimmune diseases by affecting Treg cell development and function.


Subject(s)
Autoimmune Diseases/immunology , Cell Differentiation , Immune Tolerance , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/genetics , Cell Self Renewal , Epigenesis, Genetic , Forkhead Transcription Factors/metabolism , Gene-Environment Interaction , Genetic Predisposition to Disease , Homeostasis , Humans , Polymorphism, Genetic
15.
Proc Natl Acad Sci U S A ; 114(31): E6400-E6409, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28698369

ABSTRACT

T-follicular helper (Tfh) cells differentiate through a multistep process, culminating in germinal center (GC) localized GC-Tfh cells that provide support to GC-B cells. T-follicular regulatory (Tfr) cells have critical roles in the control of Tfh cells and GC formation. Although Tfh-cell differentiation is inhibited by IL-2, regulatory T (Treg) cell differentiation and survival depend on it. Here, we describe a CD25- subpopulation within both murine and human PD1+CXCR5+Foxp3+ Tfr cells. It is preferentially located in the GC and can be clearly differentiated from CD25+ non-GC-Tfr, Tfh, and effector Treg (eTreg) cells by the expression of a wide range of molecules. In comparison to CD25+ Tfr and eTreg cells, CD25- Tfr cells partially down-regulate IL-2-dependent canonical Treg features, but retain suppressive function, while simultaneously up-regulating genes associated with Tfh and GC-Tfh cells. We suggest that, similar to Tfh cells, Tfr cells follow a differentiation pathway generating a mature GC-localized subpopulation, CD25- Tfr cells.


Subject(s)
Germinal Center/cytology , Germinal Center/immunology , Interleukin-2 Receptor alpha Subunit/genetics , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Down-Regulation/immunology , Forkhead Transcription Factors/biosynthesis , Humans , Interleukin-2 Receptor alpha Subunit/biosynthesis , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Receptors, CXCR5/biosynthesis , Repressor Proteins/biosynthesis
16.
Cell Rep ; 19(6): 1176-1188, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28494867

ABSTRACT

T cell receptor (TCR) signaling by MHC class I and II induces thymocytes to acquire cytotoxic and helper fates via the induction of Runx3 and ThPOK transcription factors, respectively. The mechanisms by which TCR signaling is translated into transcriptional programs for each cell fate remain elusive. Here, we show that, in post-selection thymocytes, a genome organizer, SATB1, activates genes for lineage-specifying factors, including ThPOK, Runx3, CD4, CD8, and Treg factor Foxp3, via regulating enhancers in these genes in a locus-specific manner. Indeed, SATB1-deficient thymocytes are partially re-directed into inappropriate T lineages after both MHC class I- and II-mediated selection, and they fail to generate NKT and Treg subsets. Despite its essential role in activating enhancers for the gene encoding ThPOK in TCR-signaled thymocytes, SATB1 becomes dispensable for maintaining ThPOK in CD4+ T cells. Collectively, our findings demonstrate that SATB1 shapes the primary T cell pool by directing lineage-specific transcriptional programs in the thymus.


Subject(s)
Lymphopoiesis , Matrix Attachment Region Binding Proteins/metabolism , T-Lymphocyte Subsets/cytology , Animals , CD4 Antigens/genetics , CD4 Antigens/metabolism , CD8 Antigens/genetics , CD8 Antigens/metabolism , Cell Lineage , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Enhancer Elements, Genetic , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Matrix Attachment Region Binding Proteins/genetics , Mice , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
17.
Nat Commun ; 8: 15366, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28497800

ABSTRACT

The T helper 9 (Th9) cell transcriptional network is formed by an equilibrium of signals induced by cytokines and antigen presentation. Here we show that, within this network, two interferon regulatory factors (IRF), IRF1 and IRF4, display opposing effects on Th9 differentiation. IRF4 dose-dependently promotes, whereas IRF1 inhibits, IL-9 production. Likewise, IRF1 inhibits IL-9 production by human Th9 cells. IRF1 counteracts IRF4-driven Il9 promoter activity, and IRF1 and IRF4 have opposing function on activating histone modifications, thus modulating RNA polymerase II recruitment. IRF1 occupancy correlates with decreased IRF4 abundance, suggesting an IRF1-IRF4-binding competition at the Il9 locus. Furthermore, IRF1 shapes Th9 cells with an interferon/Th1 gene signature. Consistently, IRF1 restricts the IL-9-dependent pathogenicity of Th9 cells in a mouse model of allergic asthma. Thus our study reveals that the molecular ratio between IRF4 and IRF1 balances Th9 fate, thus providing new possibilities for manipulation of Th9 differentiation.


Subject(s)
Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factors/metabolism , Interleukin-9/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Gene Expression Profiling , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factors/genetics , Interleukin-9/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/cytology
19.
Nat Immunol ; 18(2): 173-183, 2017 02.
Article in English | MEDLINE | ID: mdl-27992401

ABSTRACT

Most Foxp3+ regulatory T (Treg) cells develop in the thymus as a functionally mature T cell subpopulation specialized for immune suppression. Their cell fate appears to be determined before Foxp3 expression; yet molecular events that prime Foxp3- Treg precursor cells are largely obscure. We found that Treg cell-specific super-enhancers (Treg-SEs), which were associated with Foxp3 and other Treg cell signature genes, began to be activated in Treg precursor cells. T cell-specific deficiency of the genome organizer Satb1 impaired Treg-SE activation and the subsequent expression of Treg signature genes, causing severe autoimmunity due to Treg cell deficiency. These results suggest that Satb1-dependent Treg-SE activation is crucial for Treg cell lineage specification in the thymus and that its perturbation is causative of autoimmune and other immunological diseases.


Subject(s)
Cell Differentiation/immunology , Forkhead Transcription Factors/metabolism , Matrix Attachment Region Binding Proteins/metabolism , T-Lymphocytes, Regulatory/physiology , Transcriptional Activation/immunology , Animals , Autoimmunity , Cell Lineage , Cells, Cultured , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Immune Tolerance , Male , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , Precursor Cells, T-Lymphoid/physiology
20.
Proc Natl Acad Sci U S A ; 113(17): E2393-402, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27078110

ABSTRACT

High-throughput gene expression data are one of the primary resources for exploring complex intracellular dynamics in modern biology. The integration of large amounts of public data may allow us to examine general dynamical relationships between regulators and target genes. However, obstacles for such analyses are study-specific biases or batch effects in the original data. Here we present Immuno-Navigator, a batch-corrected gene expression and coexpression database for 24 cell types of the mouse immune system. We systematically removed batch effects from the underlying gene expression data and showed that this removal considerably improved the consistency between inferred correlations and prior knowledge. The data revealed widespread cell type-specific correlation of expression. Integrated analysis tools allow users to use this correlation of expression for the generation of hypotheses about biological networks and candidate regulators in specific cell types. We show several applications of Immuno-Navigator as examples. In one application we successfully predicted known regulators of importance in naturally occurring Treg cells from their expression correlation with a set of Treg-specific genes. For one high-scoring gene, integrin ß8 (Itgb8), we confirmed an association between Itgb8 expression in forkhead box P3 (Foxp3)-positive T cells and Treg-specific epigenetic remodeling. Our results also suggest that the regulation of Treg-specific genes within Treg cells is relatively independent of Foxp3 expression, supporting recent results pointing to a Foxp3-independent component in the development of Treg cells.


Subject(s)
Databases, Factual , Gene Expression Regulation , Gene Regulatory Networks , Immune System , T-Lymphocytes, Regulatory , Animals , Forkhead Transcription Factors/genetics , Gene Expression , Genes, Regulator , Genome-Wide Association Study/methods , Hematopoiesis , Immune System/cytology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...