GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: The FASEB Journal, Wiley, Vol. 34, No. S1 ( 2020-04), p. 1-1
    Type of Medium: Online Resource
    ISSN: 0892-6638 , 1530-6860
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2020
    detail.hit.zdb_id: 1468876-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 197-197
    Abstract: Gastrointestinal (GI) tract involvement is the major cause of morbidity and mortality in acute graft versus host disease (GVHD) and pathological damage is largely attributable to inflammatory cytokine production. Recently, we and others identified GM-CSF as a cytokine that is produced primarily by donor-derived CD4+ T cells and mediates inflammation in the GI tract. However, the precise mechanism by which GM-CSF induces pathological damage and the transcriptional profile of this novel colitogenic CD4+ GM-CSF+ T cell population have not been defined. To address these questions, we employed a well-defined murine model of GVHD [C57BL/6 (H-2b)→Balb/c (H-2d)] and demonstrated that GM-CSF induces inflammation by enhancing the activation of donor-derived dendritic cells in the colon as evidenced by increased expression of costimulatory molecules (i.e. CD80 and CD86) and the production of IL-23. In addition, GM-CSF linked adaptive to innate immunity by promoting indirect alloantigen presentation in the mesenteric lymph nodes which was IL-23 dependent and characterized by an increased number of CD103+ CD11b+ dendritic cells and donor CD4+ T cells with a proinflammatory cytokine phenotype. Unexpectedly, we observed two distinct CD4+ GM-CSF+ populations in the GI tract that were distinguishable by the presence or absence of IFN-γ production by intracellular cytokine staining (i.e. CD4+ GM-CSF+ IFN-γ+ and CD4+ GM-CSF+ IFN-γ-). Notably, CD4+ GM-CSF+ IFN-γ- cells were largely absent from other target organs (e.g. liver, lung), suggesting that this population had a unique role in the biology of GVHD in the GI tract. To determine whether these two populations represented transcriptionally distinct lineages or reflected TH1-biased lineage plasticity, we performed single cell RNA sequencing and immunological profiling on donor-derived sort-purified T cells from the colons of GVHD mice one and three weeks post-transplant using the 10X Genomics platform. After selecting only high quality reads, we recovered 6315 unique barcodes corresponding to individual cells and identified several transcriptionally distinct cell clusters that spatially segregated following Seurat and UMAP analysis. Colonic T cells obtained on days 7 and 21 post transplantation completely separated, indicating that the transcriptional profile of these cells changes dramatically between early and later time points. Detectable transcription of GM-CSF was observed in two distinct populations of CD4+ T cells only at the 21-day timepoint. Notably, only one of these clusters co-expressed IFN-γ, confirming our flow-based results, and indicating that CD4+ GM-CSF+ IFN-γ+ and GM-CSF+ IFN-γ- T cells represented distinct populations. Further analysis revealed that CD4+ GM-CSF+ IFN-γ+ T cells were T-bet+ and differentially expressed high levels of costimulatory molecules (CD137, OX40, and CD81) and PD-1, indicative of an activated T cell phenotype. In contrast, CD4+ GM-CSF+ IFN-γ- T cells were distinguishable by the co-expression of T-bet and Gata-3, which is a TH2-defining transcription factor, as well as by the IL-7R and a series of interferon stimulated genes (IFITM1, IFITM2, and IFITM3), supporting the premise that these cells constitute a discrete TH cell lineage. To further characterize these CD4+ T cell populations, we examined the T cell repertoire (TCR) using a targeted sequencing analysis approach of our barcoded cDNA library. We identified 444 unique clonotypes among CD4+ GM-CSF+ T cells based on sequencing of CDR3 regions of TCR alpha and beta chains. Notably, only 5 clonotypes were shared between CD4+ GM-CSF+ IFN-γ+ and CD4+ GM-CSF+ IFN-γ-T cells, representing 58 of 1154 (~5%) of the total cells in both clusters. Thus, this minimal overlap suggested that these T cells were responding to non-overlapping antigens within the GI tract. Analysis of V beta TCR gene usage revealed that CD4+ GM-CSF+ IFN-γ- cells had a highly-biased repertoire with approximately half of cells utilizing a single V beta gene, Vbeta3. In contrast, CD4+ GM-CSF+ IFN-γ+ T cells had a much more evenly distributed Vbeta receptor profile with no predominant Vbeta usage. Collectively, these studies demonstrate the existence of two transcriptionally distinct CD4+ GM-CSF+ T cell populations that accumulate within the GI tract, possess non-overlapping T cell repertoires, promote indirect alloantigen presentation, and mediate pathological damage during GVHD. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Advanced Science, Wiley, Vol. 10, No. 28 ( 2023-10)
    Abstract: Regulatory T (Treg) cells are inevitable to prevent deleterious immune responses to self and commensal microorganisms. Treg function requires continuous expression of the transcription factor (TF) FOXP3 and is divided into two major subsets: resting (rTregs) and activated (aTregs). Continuous T cell receptor (TCR) signaling plays a vital role in the differentiation of aTregs from their resting state, and in their immune homeostasis. The process by which Tregs differentiate, adapt tissue specificity, and maintain stable phenotypic expression at the transcriptional level is still inconclusivei. In this work, the role of BATF is investigated, which is induced in response to TCR stimulation in naïve T cells and during aTreg differentiation. Mice lacking BATF in Tregs developed multiorgan autoimmune pathology. As a transcriptional regulator, BATF is required for Treg differentiation, homeostasis, and stabilization of FOXP3 expression in different lymphoid and non‐lymphoid tissues. Epigenetically, BATF showed direct regulation of Treg‐specific genes involved in differentiation, maturation, and tissue accumulation. Most importantly, FOXP3 expression and Treg stability require continuous BATF expression in Tregs, as it regulates demethylation and accessibility of the CNS2 region of the Foxp3 locus. Considering its role in Treg stability, BATF should be considered an important therapeutic target in autoimmune disease.
    Type of Medium: Online Resource
    ISSN: 2198-3844 , 2198-3844
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2023
    detail.hit.zdb_id: 2808093-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    Frontiers Media SA ; 2018
    In:  Frontiers in Immunology Vol. 9 ( 2018-12-7)
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 9 ( 2018-12-7)
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2018
    detail.hit.zdb_id: 2606827-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of Experimental Medicine, Rockefeller University Press, Vol. 218, No. 3 ( 2021-03-01)
    Abstract: Tracking how individual naive T cells from a natural TCR repertoire clonally expand, differentiate, and make lineage choices in response to an infection has not previously been possible. Here, using single-cell sequencing technology to identify clones by their unique TCR sequences, we were able to trace the clonal expansion, differentiation trajectory, and lineage commitment of individual virus-specific CD4 T cells during an acute lymphocytic choriomeningitis virus (LCMV) infection. Notably, we found previously unappreciated clonal diversity and cellular heterogeneity among virus-specific helper T cells. Interestingly, although most naive CD4 T cells gave rise to multiple lineages at the clonal level, ∼28% of naive cells exhibited a preferred lineage choice toward either Th1 or TFH cells. Mechanistically, we found that TCR structure, in particular the CDR3 motif of the TCR α chain, skewed lineage decisions toward the TFH cell fate.
    Type of Medium: Online Resource
    ISSN: 0022-1007 , 1540-9538
    RVK:
    Language: English
    Publisher: Rockefeller University Press
    Publication Date: 2021
    detail.hit.zdb_id: 1477240-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Diabetes Association ; 2020
    In:  Diabetes Vol. 69, No. Supplement_1 ( 2020-06-01)
    In: Diabetes, American Diabetes Association, Vol. 69, No. Supplement_1 ( 2020-06-01)
    Abstract: Type 1 diabetes is an autoimmune disease caused by destruction of pancreatic β-cells. During disease progression, β-cells are exposed to proinflammatory cytokines like interleukin-1β (IL-1β) and interferon-γ (IFN-γ), stimulating them to express inducible nitric oxide synthase (iNOS) and produce micromolar levels of nitric oxide. While some cytokine-mediated changes in islet gene expression are known, we do not have a global view of the genes induced by cytokines nor do we know how non-β-cells respond to cytokines. Following 6-hr treatment with IL-1β and IFN-γ, adult C57BL/6 mouse islets were subjected to single-cell RNA-sequencing using the 10x Genomics Chromium platform. iNOS mRNA (Nos2) was induced in 73% of β-cells in response to both cytokines. Unexpectedly, 72% of Δ-cells and 44% of α-cells expressed Nos2 at levels similar to that found in β-cells. This is surprising since β-cells are thought to be the primary islet source of nitric oxide. Of the 241 genes enriched in Nos2-expressing β-cells, 70% were also enriched in Nos2-expressing Δ- and α-cells, suggesting that these cells initially respond to cytokine exposure by activating similar pathways. Among the enriched genes were those indicative of anti-viral defenses, suggesting induction of protective responses. Nos2 expression was negatively correlated with expression of genes encoding heat shock proteins, such as Hsp70, consistent with previous reports that heat shock inhibits cytokine-induced iNOS expression. Finally, we observed IL-1β-induced repression of β-cell identity genes, including Mafa, Pdx1, Slc2a2, and Ucn3. Repression of these identity genes was negatively correlated with Hsp70 expression, suggesting that cellular stress prevents this dedifferentiation. While these studies are ongoing, they suggest that (1) Δ- and α-cells respond to cytokines by upregulating Nos2 mRNA, (2) cytokines repress β-cell identity genes, and (3) both of these outcomes can be inhibited by induction of cellular stress. Disclosure J. Stancill: None. A. Khatun: None. M. Kasmani: None. W. Cui: None. J.A. Corbett: None.
    Type of Medium: Online Resource
    ISSN: 0012-1797 , 1939-327X
    Language: English
    Publisher: American Diabetes Association
    Publication Date: 2020
    detail.hit.zdb_id: 1501252-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Life Science Alliance, LLC ; 2021
    In:  Life Science Alliance Vol. 4, No. 6 ( 2021-06), p. e202000949-
    In: Life Science Alliance, Life Science Alliance, LLC, Vol. 4, No. 6 ( 2021-06), p. e202000949-
    Abstract: Exposure to proinflammatory cytokines is believed to contribute to pancreatic β-cell damage during diabetes development. Although some cytokine-mediated changes in islet gene expression are known, the heterogeneity of the response is not well-understood. After 6-h treatment with IL-1β and IFN-γ alone or together, mouse islets were subjected to single-cell RNA sequencing. Treatment with both cytokines together led to expression of inducible nitric oxide synthase mRNA ( Nos2 ) and antiviral and immune-associated genes in a subset of β-cells. Interestingly, IL-1β alone activated antiviral genes. Subsets of δ- and α-cells expressed Nos2 and exhibited similar gene expression changes as β-cells, including increased expression of antiviral genes and repression of identity genes. Finally, cytokine responsiveness was inversely correlated with expression of genes encoding heat shock proteins. Our findings show that all islet endocrine cell types respond to cytokines, IL-1β induces the expression of protective genes, and cellular stress gene expression is associated with inhibition of cytokine signaling.
    Type of Medium: Online Resource
    ISSN: 2575-1077
    Language: English
    Publisher: Life Science Alliance, LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2948687-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Science Immunology, American Association for the Advancement of Science (AAAS), Vol. 6, No. 64 ( 2021-10-29)
    Abstract: “Stem-like” TCF1 + CD8 + T (T SL ) cells are necessary for long-term maintenance of T cell responses and the efficacy of immunotherapy, but, as tumors contain signals that should drive T cell terminal differentiation, how these cells are maintained in tumors remains unclear. In this study, we found that a small number of TCF1 + tumor-specific CD8 + T cells were present in lung tumors throughout their development. Yet, most intratumoral T cells differentiated as tumors progressed, corresponding with an immunologic shift in the tumor microenvironment (TME) from “hot” (T cell inflamed) to “cold” (non–T cell inflamed). By contrast, most tumor-specific CD8 + T cells in tumor-draining lymph nodes (dLNs) had functions and gene expression signatures similar to T SL from chronic lymphocytic choriomeningitis virus infection, and this population was stable over time despite the changes in the TME. dLN T cells were the developmental precursors of, and were clonally related to, their more differentiated intratumoral counterparts. Our data support the hypothesis that dLN T cells are the developmental precursors of the TCF1 + T cells in tumors that are maintained by continuous migration. Last, CD8 + T cells similar to T SL were also present in LNs from patients with lung adenocarcinoma, suggesting that a similar model may be relevant in human disease. Thus, we propose that the dLN T SL reservoir has a critical function in sustaining antitumor T cells during tumor development and in protecting them from the terminal differentiation that occurs in the TME.
    Type of Medium: Online Resource
    ISSN: 2470-9468
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2021
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Bioinformation, Biomedical Informatics, Vol. 10, No. 6 ( 2014-6-30), p. 384-386
    Type of Medium: Online Resource
    ISSN: 0973-8894 , 0973-2063
    Uniform Title: English
    URL: Issue
    Language: Unknown
    Publisher: Biomedical Informatics
    Publication Date: 2014
    detail.hit.zdb_id: 2203786-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cell Reports, Elsevier BV, Vol. 35, No. 8 ( 2021-05), p. 109160-
    Type of Medium: Online Resource
    ISSN: 2211-1247
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 2649101-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...