77 research outputs found

    Interleukin 23 in IBD Pathogenesis

    Get PDF
    Interleukin-23 (IL-23) is a cytokine that belongs to the IL-12 cytokine family that is produced mainly by antigen-presenting cells. IL-23 receptor is expressed by various innate and adaptive immune cells, including group 3 innate lymphoid cells (ILC3), neutrophils, Ī³Ī“ T cells, Th17 and natural killer T (NKT) cells. IL-23 regulates various functions of the responding cells critical for host protective responses but is also implicated in many chronic inflammatory diseases including inflammatory bowel diseases (IBD). IL-23 receptor signaling components and downstream effector cytokines IL-17A/F, interferon-gamma (IFN-Ī³), IL-22, granulocyte macrophage colonyā€“stimulating factor (GMCSF) have been shown to impact IBD-like disease development in various animal models; therapeutic approaches targeting the IL-23 pathway in IBD are in clinical trials. In this chapter, we attempt to review the literature on IL-23ā€“mediated IBD pathogenesis. We did this by gathering the current information about the individual IL-23ā€“producing and IL-23ā€“responsive cells as to how they contribute to IBD pathology through various inflammatory mediators

    Schnurri-3 (KRC) Interacts with c-Jun to Regulate the IL-2 Gene in T Cells

    Get PDF
    The activator protein 1 (AP-1) transcription factor is a key participant in the control of T cell proliferation, cytokine production, and effector function. In the immune system, AP-1 activity is highest in T cells, suggesting that a subset of T cellā€“specific coactivator proteins exist to selectively potentiate AP-1 function. Here, we describe that the expression of Schnurri-3, also known as Īŗ recognition component (KRC), is induced upon T cell receptor signaling in T cells and functions to regulate the expression of the interleukin 2 (IL-2) gene. Overexpression of KRC in transformed and primary T cells leads to increased IL-2 production, whereas dominant-negative KRC, or loss of KRC protein in KRC-null mice, results in diminished IL-2 production. KRC physically associates with the c-Jun transcription factor and serves as a coactivator to augment AP-1ā€“dependent IL-2 gene transcription

    Ets-1 is a negative regulator of Th17 differentiation

    Get PDF
    IL-17 is a proinflammatory cytokine that plays a role in the clearance of extracellular bacteria and contributes to the pathology of many autoimmune and allergic conditions. IL-17 is produced mainly by a newly characterized subset of T helper (Th) cells termed Th17. Although the role of Th17 cells in the pathology of autoimmune diseases is well established, the transcription factors regulating the differentiation of Th17 cells remain poorly characterized. We report that Ets-1ā€“deficient Th cells differentiated more efficiently to Th17 cells than wild-type cells. This was attributed to both low IL-2 production and increased resistance to the inhibitory effect of IL-2 on Th17 differentiation. The resistance to IL-2 suppression was caused by a defect downstream of STAT5 phosphorylation, but was not caused by a difference in the level of RORĪ³t. Furthermore, Ets-1ā€“deficient mice contained an abnormally high level of IL-17 transcripts in their lungs and exhibited increased mucus production by airway epithelial cells in an IL-17ā€“dependent manner. Based on these observations, we report that Ets-1 is a negative regulator of Th17 differentiation

    Adaptive Autoimmunity and Foxp3-Based Immunoregulation in Zebrafish

    Get PDF
    Background: Jawed vertebrates generate their immune-receptor repertoire by a recombinatorial mechanism that has the potential to produce harmful autoreactive lymphocytes. In mammals, peripheral tolerance to self-antigens is enforced by Foxp3+ regulatory T cells. Recombinatorial mechanisms also operate in teleosts, but active immunoregulation is thought to be a late incorporation to the vertebrate lineage. Methods/Principal Findings: Here we report the characterization of adaptive autoimmunity and Foxp3-based immunoregulation in the zebrafish. We found that zebrafish immunization with an homogenate of zebrafish central nervous system (zCNS) triggered CNS inflammation and specific antibodies. We cloned the zebrafish ortholog for mammalian Foxp3 (zFoxp3) which induced a regulatory phenotype on mouse T cells and controlled IL-17 production in zebrafish embryos. Conclusions/Significance: Our findings demonstrate the acquisition of active mechanisms of self-tolerance early in vertebrate evolution, suggesting that active regulatory mechanisms accompany the development of the molecular potential for adaptive autoimmunity. Moreover, they identify the zebrafish as a tool to study the molecular pathways controlling adaptive immunity

    Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid

    Get PDF
    To maintain immune homeostasis, the intestinal immune system has evolved redundant regulatory strategies. In this regard, the gut is home to a large number of regulatory T (T reg) cells, including the Foxp3+ T reg cell. Therefore, we hypothesized that the gut environment preferentially supports extrathymic T reg cell development. We show that peripheral conversion of CD4+ T cells to T reg cells occurs primarily in gut-associated lymphoid tissue (GALT) after oral exposure to antigen and in a lymphopenic environment. Dendritic cells (DCs) purified from the lamina propria (Lp; LpDCs) of the small intestine were found to promote a high level of T reg cell conversion relative to lymphoid organā€“derived DCs. This enhanced conversion by LpDCs was dependent on TGF-Ī² and retinoic acid (RA), which is a vitamin A metabolite highly expressed in GALT. Together, these data demonstrate that the intestinal immune system has evolved a self-contained strategy to promote T reg cell neoconversion

    The TYK2-P1104A Autoimmune Protective Variant Limits Coordinate Signals Required to Generate Specialized T Cell Subsets

    Get PDF
    TYK2 is a JAK family member that functions downstream of multiple cytokine receptors. Genome wide association studies have linked a SNP (rs34536443) within TYK2 encoding a Proline to Alanine substitution at amino acid 1104, to protection from multiple autoimmune diseases including systemic lupus erythematosus (SLE) and multiple sclerosis (MS). The protective role of this SNP in autoimmune pathogenesis, however, remains incompletely understood. Here we found that T follicular helper (Tfh) cells, switched memory B cells, and IFNAR signaling were decreased in healthy individuals that expressed the protective variant TYK2A1104 (TYK2P). To study this variant in vivo, we developed a knock-in murine model of this allele. Murine Tyk2P expressing T cells homozygous for the protective allele, but not cells heterozygous for this change, manifest decreased IL-12 receptor signaling, important for Tfh lineage commitment. Further, homozygous Tyk2P T cells exhibited diminished in vitro Th1 skewing. Surprisingly, despite these signaling changes, in vivo formation of Tfh and GC B cells was unaffected in two models of T cell dependent immune responses and in two alternative SLE models. TYK2 is also activated downstream of IL-23 receptor engagement. Here, we found that Tyk2P expressing T cells had reduced IL-23 dependent signaling as well as a diminished ability to skew toward Th17 in vitro. Consistent with these findings, homozygous, but not heterozygous, Tyk2P mice were fully protected in a murine model of MS. Homozygous Tyk2P mice had fewer infiltrating CD4+ T cells within the CNS. Most strikingly, homozygous mice had a decreased proportion of IL-17+/IFNĪ³+, double positive, pathogenic CD4+ T cells in both the draining lymph nodes (LN) and CNS. Thus, in an autoimmune model, such as EAE, impacted by both altered Th1 and Th17 signaling, the Tyk2P allele can effectively shield animals from disease. Taken together, our findings suggest that TYK2P diminishes IL-12, IL-23, and IFN I signaling and that its protective effect is most likely manifest in the setting of autoimmune triggers that concurrently dysregulate at least two of these important signaling cascades

    Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression

    Get PDF
    The study of T regulatory cells (T reg cells) has been limited by the lack of specific surface markers and an inability to define mechanisms of suppression. We show that the expression of CD39/ENTPD1 in concert with CD73/ecto-5ā€²-nucleotidase distinguishes CD4+/CD25+/Foxp3+ T reg cells from other T cells. These ectoenzymes generate pericellular adenosine from extracellular nucleotides. The coordinated expression of CD39/CD73 on T reg cells and the adenosine A2A receptor on activated T effector cells generates immunosuppressive loops, indicating roles in the inhibitory function of T reg cells. Consequently, T reg cells from Cd39-null mice show impaired suppressive properties in vitro and fail to block allograft rejection in vivo. We conclude that CD39 and CD73 are surface markers of T reg cells that impart a specific biochemical signature characterized by adenosine generation that has functional relevance for cellular immunoregulation

    CD4+CCR6+ TĀ cells, but not Ī³Ī“ TĀ cells, are important for the IL-23R-dependent progression of antigen-induced inflammatory arthritis in mice

    Get PDF
    IL-23 plays an important role in the development of arthritis and the IL-23 receptor (IL-23R) is expressed on different types of TĀ cells. However, it is not fully clear which IL-23R+ TĀ cells are critical in driving TĀ cell-mediated synovitis. We demonstrate, using knock-in IL-23R-GFP reporter (IL-23RGFP/+) mice, that CD4+CCR6+ TĀ cells and Ī³Ī“ TĀ cells, but not CD8+ TĀ cells, express the IL-23R(GFP). During early arthritis, IL-23R(GFP)+CD4+CCR6+ TĀ cells, but not IL-23R(GFP)+ Ī³Ī“ TĀ cells, were present in the inflamed joints. IL-23RGFP/+ mice were bred as homozygotes to obtain IL-23RGFP/GFP (IL-23R deficient/IL-23Rāˆ’/āˆ’) mice, which express GFP under the IL-23R promotor. Arthritis progression and joint damage were significantly milder in IL-23Rāˆ’/āˆ’ mice, which revealed less IL-17A+ cells in their lymphoid tissues. Surprisingly, IL-23Rāˆ’/āˆ’ mice had increased numbers of IL-23R(GFP)+CD4+CCR6+ and CCR7+CD4+CCR6+ TĀ cells in their spleen compared to WT, and IL-23 suppressed CCR7 expression in vitro. However, IL-23R(GFP)+CD4+CCR6+ TĀ cells were present in the synovium of IL-23Rāˆ’/āˆ’ mice at day 4. Finally, adoptive transfer experiments revealed that CD4+CCR6+ TĀ cells and not Ī³Ī“ TĀ cells drive arthritis progression. These data suggest that IL-23R-dependent TĀ cell-mediated synovitis is dependent on CD4+CCR6+ TĀ cells and not on Ī³Ī“ TĀ cells

    Fingolimod Alters Tissue Distribution and Cytokine Production of Human and Murine Innate Lymphoid Cells

    Get PDF
    Sphingosine-1 phosphate receptor 1 (S1PR1) is expressed by lymphocytes and regulates their egress from secondary lymphoid organs. Innate lymphoid cell (ILC) family has been expanded with the discovery of group 1, 2 and 3 ILCs, namely ILC1, ILC2 and ILC3. ILC3 and ILC1 have remarkable similarity to CD4+ helper T cell lineage members Th17 and Th1, respectively, which are important in the pathology of multiple sclerosis (MS). Whether human ILC subsets express S1PR1 or respond to its ligands have not been studied. In this study, we used peripheral blood/cord blood and tonsil lymphocytes as a source of human ILCs. We show that human ILCs express S1PR1 mRNA and protein and migrate toward S1P receptor ligands. Comparison of peripheral blood ILC numbers between fingolimod-receiving and treatment-free MS patients revealed that, in vivo, ILCs respond to fingolimod, an S1PR1 agonist, resulting in ILC-penia in circulation. Similarly, murine ILCs responded to fingolimod by exiting blood and accumulating in the secondary lymph nodes. Importantly, ex vivo exposure of ILC3 and ILC1 to fingolimod or SEW2871, another S1PR1 antagonist, reduced production of ILC3- and ILC1- associated cytokines GM-CSF, IL-22, IL-17, and IFN-Ī³, respectively. Surprisingly, despite reduced number of lamina propria-resident ILC3s in the long-term fingolimod-treated mice, ILC3-associated IL-22, IL-17A, GM-CSF and antimicrobial peptides were high in the gut compared to controls, suggesting that its long term use may not compromise mucosal barrier function. To our knowledge, this is the first study to investigate the impact of fingolimod on human ILC subsets in vivo and ex vivo, and provides insight into the impact of long term fingolimod use on ILC populations

    Interplay between pathogenic Th17 and regulatory T cells

    No full text
    Autoimmune diseases arise from a break in tolerance toward selfā€antigens and are characterised by the development of pathogenic T cell populations infiltrating the target organ. Regulatory T cells play an important role in maintaining selfā€tolerance through their inhibitory functions on effector T cells. The usage of ex vivoā€generated regulatory T cells (Treg) has been regarded as a potentially attractive therapeutic approach for autoimmune diseases. However, the dynamics of Treg in autoimmunity are not well understood. Here, we summarise our published findings on the interplay between Treg and Th17 effector cells in vivo during experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. We have developed Foxp3gfp ā€œknockā€inā€ mice and myelin oligodendrocyte glycoprotein (MOG)35ā€“55/IAb (MHC class II)ā€tetramers to track autoantigenā€specific Treg in vivo during EAE. On immunisation with MOG, Treg cells were detected in the central nervous system (CNS) as early as day 10. However, at the onset of disease, the accumulation of MOGā€specific effector T cells (Tā€eff) largely prevailed. Subsequently, during remission Tā€eff rapidly contracted whereas a highly suppressive Treg population persisted in the CNS. The interplay between effector Th17 and Treg extend beyond their functions in vivo as we and others have identified the factors responsible for Th17 differentiation. While TGFā€Ī² is a critical differentiation factor for Treg cells, IL6 completely inhibits the generation of Treg cells induced by TGFā€Ī². Instead, IL6 and TGFā€Ī² together induce the differentiation of pathogenic Th17 cells. Our data demonstrate a dichotomy in the generation of Th17 cells that induce autoimmunity and Treg cells that inhibit autoimmune tissue injury
    • ā€¦
    corecore