26 research outputs found

    Transcriptional Control of Regulatoy T cells

    Get PDF
    Regulatory T cells (Treg cells) are crucial mediators of peripheral self-tolerance, and their specific ablation causes catastrophic autoimmunity. The protein Foxp3 has been described as the key transcription factor delineating cells of this lineage. In this thesis, we investigate different levels of transcriptional control in Foxp3-expressing Treg cells: At first, we identified CpG rich regions along the Foxp3 gene and investigated them for their epigenetic profile. We were able to validate already-described Treg-specific demethylated regions, but we also identified new Treg-specific demethylation patterns along the Foxp3 gene. These patterns are initiated during thymic Treg development and persist in circulation, and remain intact also in DNA-methyltransferase deficient mice. Next, we explored the epigenetic control of Treg cells on a broader scale. We isolated tissue-resident Treg cells from fat, skin, liver, and lymph nodes and subjected them to whole-genome tagmentation-based methylation analysis in correlation with RNA expression profiling. This enables us to investigate tissue-specific epigenetic patterns that drive and manifest Treg cell adaption to local tissues. Additionally, we observed that the Foxp3 core promoter is completely demethylated also in Foxp3 non-expressing cell types. Therefore, we identified binding partners to the Foxp3 gene promoter in a novel procedure called inverted Chromatin IP. We validated the suppressive nature of these target proteins via luciferase-based screens, tested their differential expression profile in different cell types, and investigated the effect of their virus-mediated overexpression on in-vitro Treg cell induction. Finally, we investigated the role of one specific transcriptional regulator in Treg cells: Rbpj, commonly known as an important co-factor of Notch signaling. While we did not find evidence that Notch signaling was active in bona fide Treg cells, we still measured an upregulation of Rbpj mRNA in Treg cells compared to their conventional T-cell (Tconv) counterparts in many different tissues. Upon Treg-specific Rbpj gene deletion, we observed a steady increase in Treg frequency and number in several anatomical locations, finally leading to autoimmune pathology despite the presence of otherwise functionally-intact Treg cells. We identified the IL7-receptor, an important growth-promoting cytokine receptor, to be specifically upregulated in Rbpj-deficient Treg cells, whereas Dtx-1, a paramount anergy-promoting factor, was strongly downregulated. Furthermore, gene expression profiling and antibody staining revealed that both Treg and Tconv cells gained TH subset lineage profiles, indicating an inability of Treg cells to control (auto) immune responses in-vivo. Our data suggest a novel, Notch-independent function of Rbpj to specifically regulate Treg cell proliferation and functionality

    Stepwise acquisition of unique epigenetic signatures during differentiation of tissue Treg cells

    Get PDF
    Regulatory T cells in non-lymphoid tissues are not only critical for maintaining self-tolerance, but are also important for promoting organ homeostasis and tissue repair. It is proposed that the generation of tissue Treg cells is a stepwise, multi-site process, accompanied by extensive epigenome remodeling, finally leading to the acquisition of unique tissue-specific epigenetic signatures. This process is initiated in the thymus, where Treg cells acquire core phenotypic and functional properties, followed by a priming step in secondary lymphoid organs that permits Treg cells to exit the lymphoid organs and seed into non-lymphoid tissues. There, a final specialization process takes place in response to unique microenvironmental cues in the respective tissue. In this review, we will summarize recent findings on this multi-site tissue Treg cell differentiation and highlight the importance of epigenetic remodeling during these stepwise events

    Guidelines for the use of flow cytometry and cell sorting in immunological studies (third edition)

    Get PDF
    The third edition of Flow Cytometry Guidelines provides the key aspects to consider when performing flow cytometry experiments and includes comprehensive sections describing phenotypes and functional assays of all major human and murine immune cell subsets. Notably, the Guidelines contain helpful tables highlighting phenotypes and key differences between human and murine cells. Another useful feature of this edition is the flow cytometry analysis of clinical samples with examples of flow cytometry applications in the context of autoimmune diseases, cancers as well as acute and chronic infectious diseases. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid. All sections are written and peer‐reviewed by leading flow cytometry experts and immunologists, making this edition an essential and state‐of‐the‐art handbook for basic and clinical researchers.DFG, 389687267, Kompartimentalisierung, Aufrechterhaltung und Reaktivierung humaner Gedächtnis-T-Lymphozyten aus Knochenmark und peripherem BlutDFG, 80750187, SFB 841: Leberentzündungen: Infektion, Immunregulation und KonsequenzenEC/H2020/800924/EU/International Cancer Research Fellowships - 2/iCARE-2DFG, 252623821, Die Rolle von follikulären T-Helferzellen in T-Helferzell-Differenzierung, Funktion und PlastizitätDFG, 390873048, EXC 2151: ImmunoSensation2 - the immune sensory syste

    Premature Expression of Foxp3 in Double-Negative Thymocytes

    No full text
    <div><p>Peripheral immune regulation depends on the generation of thymic-derived regulatory T (tT<sub>reg</sub>) cells to maintain self-tolerance and to counterbalance overshooting immune responses. The expression of the T<sub>reg</sub> lineage defining transcription factor Foxp3 in developing tT<sub>reg</sub> cells depends on TCR signaling during the thymic selection process of these T cells. In this study, we surprisingly identify Foxp3<sup>+</sup> immature thymocytes at the double-negative (DN) stage in transcription factor 7 (Tcf7)-deficient mice. These Foxp3<sup>+</sup> cells did not express a TCR (β or γδ chains), CD3 or CD5 and therefore these cells were true DN cells. Further investigation of this phenomenon in a transgenic TCR model showed that Foxp3-expressing DN cells could not respond to TCR stimulation <i>in vivo</i>. These data suggest that Foxp3 expression in these DN cells occurred independently of TCR signaling. Interestingly, these Foxp3<sup>+</sup> DN cells were located in a transition state between DN1 and DN2 (CD4<sup>-</sup>CD8<sup>-</sup>CD3<sup>-</sup>TCR<sup>-</sup>CD44<sup>high</sup>CD25<sup>low</sup>). Our results indicate that Tcf7 is involved in preventing the premature expression of Foxp3 in DN thymocytes.</p></div

    DataSheet_1_Using combined single-cell gene expression, TCR sequencing and cell surface protein barcoding to characterize and track CD4+ T cell clones from murine tissues.pdf

    No full text
    Single-cell gene expression analysis using sequencing (scRNA-seq) has gained increased attention in the past decades for studying cellular transcriptional programs and their heterogeneity in an unbiased manner, and novel protocols allow the simultaneous measurement of gene expression, T-cell receptor clonality and cell surface protein expression. In this article, we describe the methods to isolate scRNA/TCR-seq-compatible CD4+ T cells from murine tissues, such as skin, spleen, and lymph nodes. We describe the processing of cells and quality control parameters during library preparation, protocols for multiplexing of samples, and strategies for sequencing. Moreover, we describe a step-by-step bioinformatic analysis pipeline from sequencing data generated using these protocols. This includes quality control, preprocessing of sequencing data and demultiplexing of individual samples. We perform quantification of gene expression and extraction of T-cell receptor alpha and beta chain sequences, followed by quality control and doublet detection, and methods for harmonization and integration of datasets. Next, we describe the identification of highly variable genes and dimensionality reduction, clustering and pseudotemporal ordering of data, and we demonstrate how to visualize the results with interactive and reproducible dashboards. We will combine different analytic R-based frameworks such as Bioconductor and Seurat, illustrating how these can be interoperable to optimally analyze scRNA/TCR-seq data of CD4+ T cells from murine tissues.</p

    DataSheet_1_Inflammatory perturbations in early life long-lastingly shape the transcriptome and TCR repertoire of the first wave of regulatory T cells.pdf

    No full text
    The first wave of Foxp3+ regulatory T cells (Tregs) generated in neonates is critical for the life-long prevention of autoimmunity. Although it is widely accepted that neonates are highly susceptible to infections, the impact of neonatal infections on this first wave of Tregs is completely unknown. Here, we challenged newborn Treg fate-mapping mice (Foxp3eGFPCreERT2xROSA26STOP-eYFP) with the Toll-like receptor (TLR) agonists LPS and poly I:C to mimic inflammatory perturbations upon neonatal bacterial or viral infections, respectively, and subsequently administrated tamoxifen during the first 8 days of life to selectively label the first wave of Tregs. Neonatally-tagged Tregs preferentially accumulated in non-lymphoid tissues (NLTs) when compared to secondary lymphoid organs (SLOs) irrespective of the treatment. One week post challenge, no differences in the frequency and phenotypes of neonatally-tagged Tregs were observed between challenged mice and untreated controls. However, upon aging, a decreased frequency of neonatally-tagged Tregs in both NLTs and SLOs was detected in challenged mice when compared to untreated controls. This decrease became significant 12 weeks post challenge, with no signs of altered Foxp3 stability. Remarkably, this late decrease in the frequency of neonatally-tagged Tregs only occurred when newborns were challenged, as treating 8-days-old mice with TLR agonists did not result in long-lasting alterations of the first wave of Tregs. Combined single-cell T cell receptor (TCR)-seq and RNA-seq revealed that neonatal inflammatory perturbations drastically diminished TCR diversity and long-lastingly altered the transcriptome of neonatally-tagged Tregs, exemplified by lower expression of Tigit, Foxp3, and Il2ra. Together, our data demonstrate that a single, transient encounter with a pathogen in early life can have long-lasting consequences for the first wave of Tregs, which might affect immunological tolerance, prevention of autoimmunity, and other non-canonical functions of tissue-resident Tregs in adulthood.</p

    Analysis of Foxp3<sup>+</sup> DN cells in TEa-Tcf7-deficient mice.

    No full text
    <p>(A) Representative plots showing TCRVβ6 and TCRVα2 expression on CD4SP thymocytes from TEa-Tcf7<sup>+/+</sup> and TEa-Tcf7<sup>-/-</sup> mice in the presence or absence of cognate antigen (Ag). The Tg TCR population is divided into TCR<sup>high</sup> and TCR<sup>low</sup> populations. (B-C) Quantification of the percentage of total (B) or TCR<sup>high</sup> (C) TCRVβ6<sup>+</sup>TCRVα2<sup>+</sup> cells among CD4SP thymocytes (n = 8). (D) Representative plots showing Foxp3 expression in DN TCRVβ6<sup>+</sup>TCRVα2<sup>+</sup> thymocytes from TEa-Tcf7<sup>+/+</sup> and TEa-Tcf7<sup>-/-</sup> mice in the absence of Ag. (E) Quantification of Foxp3<sup>+</sup> DN TCRVβ6<sup>+</sup>TCRVα2<sup>+</sup> thymocytes from TEa-Tcf7<sup>+/+</sup> and TEa-Tcf7<sup>-/-</sup> mice in the presence or absence of Ag (n = 8). (F-G) Representative plots showing TCRVβ6 and TCRVα2 expression on DN Foxp3<sup>+</sup> (F) or CD4SP Foxp3<sup>+</sup> (G) thymocytes from TEa-Tcf7<sup>+/+</sup> and TEa-Tcf7<sup>-/-</sup> mice in the presence or absence of Ag. Cells are pre-gated on TCRVβ6<sup>+</sup>TCRVα2<sup>+</sup>. Each dot represents one individual animal and mean is shown for all quantified data. Numbers show percentages of cells within the indicated box. NS, not significant, *** P < 0.001, **** P < 0.0001 (unpaired t-test).</p

    Foxp3 expression at the DN cell stage in Tcf7-deficient mice.

    No full text
    <p>(A) Representative plots and quantification of Foxp3 staining in CD4<sup>-</sup>CD8<sup>-</sup> (DN) thymocytes from Tcf7<sup>+/+</sup> and Tcf7<sup>-/-</sup> mice (n = 8). (B) Left panels: Representative plots showing Foxp3 and intracellular (IC) TCRβ staining in DN thymocytes from Tcf7<sup>+/+</sup> and Tcf7<sup>-/-</sup> mice. Middle panels: TCRγδ and CD3 staining on DN Foxp3<sup>+</sup>TCRβ<sup>-</sup> cells (gate R1). Right panel: Quantification of DN Foxp3<sup>+</sup>TCRβ<sup>-</sup>TCRγδ<sup>-</sup>CD3<sup>-</sup> cells (gate R2) depicted as the percentage of total DN cells (n = 6). (C) Left panel: Representative histograms showing CD5 staining on Foxp3<sup>+</sup> DN, Foxp3<sup>+</sup> DP, and Foxp3<sup>+</sup> CD4SP cells from Tcf7<sup>-/-</sup> mice. Right panel: Quantification of CD5 geometric mean from DN, DP, and CD4SP Foxp3<sup>+</sup> or Foxp3<sup>-</sup> populations (n = 3). Mean + SD are shown for all quantified data. Numbers show percentages of cells within the indicated box. Each symbol represents an individual animal. ** P < 0.01 (unpaired t-test).</p

    Brown adipose tissue harbors a distinct sub-population of regulatory T cells.

    No full text
    Regulatory T (Treg) cells are critical determinants of both immune responses and metabolic control. Here we show that systemic ablation of Treg cells compromised the adaptation of whole-body energy expenditure to cold exposure, correlating with impairment in thermogenic marker gene expression and massive invasion of pro-inflammatory macrophages in brown adipose tissue (BAT). Indeed, BAT harbored a unique sub-set of Treg cells characterized by a unique gene signature. As these Treg cells respond to BAT activation upon cold exposure, this study defines a BAT-specific Treg sub-set with direct implications for the regulation of energy homeostasis in response to environmental stress
    corecore