63 research outputs found

    The extent of metalloproteinase-mediated LAG3 cleavage limits the efficacy of PD1 blockade

    Get PDF
    Inhibitory receptors control immune responses preventing exacerbated T cell activation and the onset of autoimmunity; however, they also limit antitumor immunity. Enhanced co-expression of PD1 and LAG3 phenotypically mark functionally exhausted tumor-specific T cells, with dual PD1/LAG3 blockade synergistically limiting tumor growth in murine models. Like PD1, LAG3 expression is induced on activated T cells to negatively regulate T cell activation and proliferation and LAG3 is also required for maximal regulatory T (Treg) cell function. However, LAG3 expression and function is itself regulated by cell surface cleavage of the transmembrane domain connecting peptide by ADAM10 and ADAM17 metalloproteinase-disintegrins. This releases soluble LAG3, of which no biological function has been found to date. To investigate the impact of LAG3 cleavage on T cells within tumors, a non-cleavable LAG3 mouse (LAG3.NC) was generated in which exons 7 and 8 of Lag3, including the connecting peptide, is deleted in Cre-expressing cells. These exons are replaced and modified so that the connecting peptide is absent preventing LAG3 cleavage. LAG3.NC CD4Cre mice (with non-cleavable LAG3 expressed on all CD8+ and CD4+ T cells, including Tregs) and LAG3.NC E8ICre mice (restricted to CD8+ T cells only) exhibit enhanced expression of LAG3 on the respective T cell subsets in B16-F10 or MC38 tumors, co-expressing with PD1. Despite increased LAG3 expression, no difference in B16-F10 or MC38 tumor growth was observed in either LAG3.NC CD4Cre or LAG3.NC E8ICre mice compared with wild-type littermates. Upon therapeutic administration of anti-PD1 antibody (clone G4), MC38 tumor-bearing wild-type mice show significant tumor regression and 40% become tumor-free resulting in long-term survival. LAG3.NC CD4Cre mice were resistant to anti-PD1 therapy and succumb to tumor growth. However, anti-PD1 mediated tumor regression and long-term survival in LAG3.NC E8ICre mice. Analysis of re-stimulated CD8+ TILs isolated from LAG3.NC CD4Cre mice did not show enhanced IFN-gamma and TNF-alpha production following anti-PD1 therapy, which was observed with LAG3.NC E8ICre mice or wild-type littermates. Moreover, reduced proliferation was observed for all T cell subsets in LAG3.NC CD4Cre mice compared with LAG3.NC E8ICre and wild-type littermates following anti-PD1 treatment. As LAG3.NC CD4Cre, but not LAG3.NC E8ICre mice, are resistant to the favorable antitumor immune effects induced by anti-PD1, this suggests that enhanced LAG3 expression on CD4+ T cells or Tregs may act as a barrier to effective anti-PD1 immunotherapy. LAG3.NC mice crossed with Cre that restricts non-cleavable LAG3 to Tregs (Foxp3yfpiCre) or CD4+ T cells (ThPOKCre) are currently under analysis

    Remodulation of the tumor microenvironment by regulatory T cells

    Get PDF
    The tumor microenvironment is a complex system, which is composed of various types of non-tumor cells including stromal fibroblasts, the blood and lymphatic vascular networks, the extracellular matrix and notably, the infiltrating immune cells. Regulatory T cells (Treg cells) play a pivotal role in tumor malignant progression and contribute to the resistance of tumors to traditional anti-cancer therapies; however, the elimination of Treg cells is not a clinically viable approach, given their crucial role in maintaining immune homeostasis and preventing autoimmunity. Our laboratory has recently reported that Treg-restricted genetic disruption of Neuropilin-1 (Nrp1) selectively induced destabilization of Treg cells within the tumor microenvironment, leading to tumor clearance without inducing autoimmunity. Interestingly, despite of their dramatic tumor-suppressive function, Nrp1-deficient Treg cells are present in tumor in a comparable manner (number and kinetics) to their wild type counterparts, which provides an invaluable research tool to interrogate the function of intratumoral Treg cells without physically removing them and inducing systemic autoimmunity. In this study we aim to systematically investigate the cellular and molecular mediators as well as the underlying mechanisms of Treg-cell function within the tumor microenvironment using a systems biology approach. With the unique Treg-restrictive Nrp1-deficient mice tumor models combined with a multifaceted approach that consists of flow cytometry based immunophenotyping and large-scale transcriptomic profiling, our results indicated that Treg cells act as an early key regulator of tumor immune infiltration and actively induce the global remodulation of the tumor immune transcriptome. Computational deconvolution analysis of the gene profiling data derived from mixed populations further predicted a list of critically modified targets that are regulated at the single cell level. The functional validation of these targets may provide mechanism(s) by which Treg cells interplay with the tumor microenvironment to potentiate tumor growth. This may lead to the development of novel and selective cancer immunotherapies

    Regulatory T Cells: Barriers of Immune Infiltration Into the Tumor Microenvironment

    Get PDF
    Regulatory T cells (Tregs) are key immunosuppressive cells that promote tumor growth by hindering the effector immune response. Tregs utilize multiple suppressive mechanisms to inhibit pro-inflammatory responses within the tumor microenvironment (TME) by inhibition of effector function and immune cell migration, secretion of inhibitory cytokines, metabolic disruption and promotion of metastasis. In turn, Tregs are being targeted in the clinic either alone or in combination with other immunotherapies, in efforts to overcome the immunosuppressive TME and increase anti-tumor effects. However, it is now appreciated that Tregs not only suppress cells intratumorally via direct engagement, but also serve as key interactors in the peritumor, stroma, vasculature and lymphatics to limit anti-tumor immune responses prior to tumor infiltration. We will review the suppressive mechanisms that Tregs utilize to alter immune and non-immune cells outside and within the TME and discuss how these mechanisms collectively allow Tregs to create and promote a physical and biological barrier, resulting in an immune-excluded or limited tumor microenvironment

    Central Nervous System Destruction Mediated by Glutamic Acid Decarboxylase-Specific CD4+ T Cells

    Get PDF
    High titers of autoantibodies against glutamic acid decarboxylase 65 (GAD65) are commonly observed in patients suffering from type 1 diabetes (T1D) as well as Stiff Person syndrome (SPS), a disorder that affects the central nervous system, and a variant of SPS, progressive encephalomyelitis with rigidity and myoclonus (PERM). While there is a considerable amount of data focusing on the role of GAD65-specific CD4+ T cells in T1D, little is known about their role in SPS. Here we show that mice possessing a monoclonal GAD65-specific CD4+ T cell population (4B5, PA19.9G11 or PA17.9G7) develop a lethal encephalomyelitis-like disease in the absence of any other T cells or B cells. GAD65-reactive CD4+ T cells were found throughout the CNS in direct concordance with GAD65 expression and activated microglia: proximal to the circumventricular organs at the interface between the brain parenchyma and the blood brain barrier. In the presence of B cells, high titer anti-GAD65 autoantibodies were generated but these had no effect on the incidence or severity of disease. In addition, GAD65-specific CD4+ T cells isolated from the brain were activated and produced IFN-γ. These findings suggest that GAD65-reactive CD4+ T cells alone mediate a lethal encephalomyelitis-like disease that may serve as a useful model to study GAD65-mediated diseases of the CNS

    Senior Recital

    Full text link
    Program listing performers and works performe

    Role of LAG-3 in Regulatory T Cells

    Get PDF
    AbstractRegulatory T cells (Tregs) limit autoimmunity but also attenuate the magnitude of antipathogen and antitumor immunity. Understanding the mechanism of Treg function and therapeutic manipulation of Tregs in vivo requires identification of Treg-selective receptors. A comparative analysis of gene expression arrays from antigen-specific CD4+ T cells differentiating to either an effector/memory or a regulatory phenotype revealed Treg-selective expression of LAG-3, a CD4-related molecule that binds MHC class II. Antibodies to LAG-3 inhibit suppression by induced Tregs both in vitro and in vivo. Natural CD4+CD25+ Tregs express LAG-3 upon activation, which is significantly enhanced in the presence of effector cells, whereas CD4+CD25+ Tregs from LAG-3−/− mice exhibit reduced regulatory activity. Lastly, ectopic expression of LAG-3 on CD4+ T cells significantly reduces their proliferative capacity and confers on them suppressor activity toward effector T cells. We propose that LAG-3 marks regulatory T cell populations and contributes to their suppressor activity

    Organs to Cells and Cells to Organoids: The Evolution of in vitro Central Nervous System Modelling

    Get PDF
    With 100 billion neurons and 100 trillion synapses, the human brain is not just the most complex organ in the human body, but has also been described as “the most complex thing in the universe.” The limited availability of human living brain tissue for the study of neurogenesis, neural processes and neurological disorders has resulted in more than a century-long strive from researchers worldwide to model the central nervous system (CNS) and dissect both its striking physiology and enigmatic pathophysiology. The invaluable knowledge gained with the use of animal models and post mortem human tissue remains limited to cross-species similarities and structural features, respectively. The advent of human induced pluripotent stem cell (hiPSC) and 3-D organoid technologies has revolutionised the approach to the study of human brain and CNS in vitro, presenting great potential for disease modelling and translational adoption in drug screening and regenerative medicine, also contributing beneficially to clinical research. We have surveyed more than 100 years of research in CNS modelling and provide in this review an historical excursus of its evolution, from early neural tissue explants and organotypic cultures, to 2-D patient-derived cell monolayers, to the latest development of 3-D cerebral organoids. We have generated a comprehensive summary of CNS modelling techniques and approaches, protocol refinements throughout the course of decades and developments in the study of specific neuropathologies. Current limitations and caveats such as clonal variation, developmental stage, validation of pluripotency and chromosomal stability, functional assessment, reproducibility, accuracy and scalability of these models are also discussed

    The James Webb Space Telescope Mission

    Full text link
    Twenty-six years ago a small committee report, building on earlier studies, expounded a compelling and poetic vision for the future of astronomy, calling for an infrared-optimized space telescope with an aperture of at least 4m4m. With the support of their governments in the US, Europe, and Canada, 20,000 people realized that vision as the 6.5m6.5m James Webb Space Telescope. A generation of astronomers will celebrate their accomplishments for the life of the mission, potentially as long as 20 years, and beyond. This report and the scientific discoveries that follow are extended thank-you notes to the 20,000 team members. The telescope is working perfectly, with much better image quality than expected. In this and accompanying papers, we give a brief history, describe the observatory, outline its objectives and current observing program, and discuss the inventions and people who made it possible. We cite detailed reports on the design and the measured performance on orbit.Comment: Accepted by PASP for the special issue on The James Webb Space Telescope Overview, 29 pages, 4 figure
    corecore