819 research outputs found

    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

    Elucidating the role of Neuropilin-1 in intra-tumoral regulatory T cell stability

    Get PDF
    Regulatory T cells (Tregs) play an integral role in the adaptive immune system through suppression of self-reactive immune responses in order to prevent autoimmunity and maintain homeostasis. However, they are deleterious in cancer through suppression of the anti-tumor immune response. In fact, we show that deletion of 50% of Tregs results in normal tumor growth. Therefore, it is advantageous to understand the role of Tregs in the tumor microenvironment in order to create targeted cancer therapies. Our lab has shown that the Neuropilin-1 (Nrp1) pathway is required for Treg stability in the tumor microenvironment, but is disposable for maintaining immune homeostasis in the periphery, identifying it as a prime therapeutic target.\ud \ud In order to further understand the role of Nrp1-deficient Tregs intratumorally, we constructed a competitive environment by utilizing Foxp3, which is located on the X chromosome, and as a result of X-inactivation, female Foxp3 Cre-YFP heterozygous mice are cellular heterozygotes. We generated Nrp1 L/L Foxp3 Cre-YFP/+ heterozygous mice comprised of 50% WT Tregs and 50% Nrp1-deficient Tregs. Surprisingly, when given B16 melanoma, heterozygous mice phenocopy Nrp1 L/L Foxp3 Cre-YFP homozygous mice (Figure 1A). This suggests that Nrp1-deficient T regs are playing an active role in shifting the anti-tumor immune response by destabilizing surrounding WT T regs as determined by DNA methylation status (Figure 1B). Neither WT nor Nrp1-deficient Tregs in the tumor from Nrp1 L/L Foxp3 Cre-YFP/+ mice can suppress in a standard microsuppression assay ex vivo, unlike WT Tregs from Foxp3 Cre-YFP mice. Through various co-culture experiments, we revealed that destabilization of WT Tregs is possibly due to a soluble factor derived from Nrp1-deficient Tregs. Our data revealed that Nrp1-deficient Tregs produce large amounts of IFNγ in the tumor microenvironment. Indeed, when treated with IFNγ, WT T regs lose suppressive capacity. In order to uncover potential novel pathways leading to this phenotype, we are performing global transcript studies using RNASeq. Overall, we have shown that Nrp1 is required for intratumoral Treg stability, and in its absence, there is an alteration in the tumor microenvironment, leading to an enhanced anti-tumor immune response. These studies uncover a novel potential target for future cancer immunotherapies that preserves peripheral immune health

    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

    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

    Mucosal Administration of Collagen V Ameliorates the Atherosclerotic Plaque Burden by Inducing Interleukin 35-dependent Tolerance

    Get PDF
    We have shown previously that collagen V (col(V)) autoimmunity is a consistent feature of atherosclerosis in human coronary artery disease and in the Apoe(-/-) mouse model. We have also shown sensitization of Apoe(-/-) mice with col(V) to markedly increase the atherosclerotic burden, providing evidence of a causative role for col(V) autoimmunity in atherosclerotic pathogenesis. Here we sought to determine whether induction of immune tolerance to col(V) might ameliorate atherosclerosis, providing further evidence for a causal role for col(V) autoimmunity in atherogenesis and providing insights into the potential for immunomodulatory therapeutic interventions. Mucosal inoculation successfully induced immune tolerance to col(V) with an accompanying reduction in plaque burden in Ldlr(-/-) mice on a high-cholesterol diet. The results therefore demonstrate that inoculation with col(V) can successfully ameliorate the atherosclerotic burden, suggesting novel approaches for therapeutic interventions. Surprisingly, tolerance and reduced atherosclerotic burden were both dependent on the recently described IL-35 and not on IL-10, the immunosuppressive cytokine usually studied in the context of induced tolerance and amelioration of atherosclerotic symptoms. In addition to the above, using recombinant protein fragments, we were able to localize two epitopes of the α1(V) chain involved in col(V) autoimmunity in atherosclerotic Ldlr(-/-) mice, suggesting future courses of experimentation for the characterization of such epitopes

    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

    Quantifying the contribution of intracranial pressure and arterial blood pressure to spontaneous tympanic membrane displacement

    Get PDF
    Objective: Although previous studies have shown associations between patient symptoms/outcomes and the spontaneous tympanic membrane displacement (spTMD) pulse amplitude, the contribution of the underlying intracranial pressure (ICP) signal to the spTMD pulse remains largely unknown. We have assessed the relative contributions of ICP and arterial blood pressure (ABP) on spTMD at different frequencies in order to determine whether spTMD contains information about the ICP above and beyond that contained in the ABP. Approach: Eleven patients, who all had invasive ICP and ABP measurements in situ, were recruited from our intensive care unit. Their spTMD was recorded and the power spectral densities of the three signals, as well as coherences between the signals, were calculated in the range 0.1–5 Hz. Simple and multiple coherences, coupled with statistical tests using surrogate data, were carried out to quantify the relative contributions of ABP and ICP to spTMD. Main results: Most power of the signals was found to predominate at respiration rate, heart rate, and their harmonics, with little outside of these frequencies. Analysis of the simple coherences found a slight preference for ICP transmission, beyond that from ABP, to the spTMD at lower frequencies (7/11 patients at respiration, 7/10 patients at respiration 1st harmonic) which is reversed at the higher frequencies (2/11 patients at heart rate and its 1st harmonic). Both ICP and ABP were found to independently contribute to the spTMD. The multiple coherence reinforced that ICP is preferentially being transmitted at respiration and respiration 1st harmonic. Significance: Both ABP and ICP contribute independently to the spTMD signal, with most power occurring at clear physiological frequencies—respiration and harmonics and heart rate and harmonics. There is information shared between the ICP and spTMD that is not present in ABP. This analysis has indicated that lower frequencies appear to favour ICP as the driver for spTMD

    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

    The GPCR-gαs-PKA Signaling Axis Promotes T Cell Dysfunction and Cancer Immunotherapy Failure

    Get PDF
    Immune checkpoint blockade (ICB) targeting PD-1 and CTLA-4 has revolutionized cancer treatment. However, many cancers do not respond to ICB, prompting the search for additional strategies to achieve durable responses. G-protein-coupled receptors (GPCRs) are the most intensively studied drug targets but are underexplored in immuno-oncology. Here, we cross-integrated large singe-cell RNA-sequencing datasets from CD8+ T cells covering 19 distinct cancer types and identified an enrichment of Gαs-coupled GPCRs on exhausted CD8+ T cells. These include EP2, EP4, A2AR, β1AR and β2AR, all of which promote T cell dysfunction. We also developed transgenic mice expressing a chemogenetic CD8-restricted Gαs–DREADD to activate CD8-restricted Gαs signaling and show that a Gαs–PKA signaling axis promotes CD8+ T cell dysfunction and immunotherapy failure. These data indicate that Gαs–GPCRs are druggable immune checkpoints that might be targeted to enhance the response to ICB immunotherapies
    • …
    corecore