11 research outputs found

    Innate modifiers of T cell behaviour during inflammatory disease

    Get PDF
    Neutrophils are the most abundant leukocyte in mammals and represent one of the first lines of defence against invading microorganisms. In recent years, it has become clear that neutrophils are not only responsible for the killing of pathogens, but that they also play an important role in shaping adaptive immune responses. The aim of this project was to determine the role of the neutrophil-derived host defence peptide, cathelicidin, in the generation of Th17 responses during inflammation. Following inoculation with heat-killed Salmonella typhimurium, cathelicidin knockout mice cannot produce IL-17 and show increased IFNγ, whereas other cytokines are produced normally. Ex vivo, I show that cathelicidin is a novel Th17/Tc17 differentiation enhancing factor, which acts directly on both CD4+ and CD8+ T cells to increase their activation status, protect them from death and concentration-dependently upregulate IL-17 production. Gene expression analysis revealed that cathelicidin downregulates the expression of several Th1-related genes and upregulates the aryl hydrocarbon receptor (AHR), a known regulator of Th17 differentiation. The addition of an AHR antagonist to our in vitro cultures abolishes the boost to IL-17 production normally induced by cathelicidin. I provide further evidence that suggests lymph node neutrophils are the cellular source of cathelicidin, which are responsible for amplifying type-17 responses during inflammation. These data contribute to our understanding of how lymph node neutrophils influence developing adaptive immune responses with sophistication and specificity

    The Outcome of Neutrophil-T Cell Contact Differs Depending on Activation Status of Both Cell Types

    Get PDF
    Neutrophils and T cells exist in close proximity in lymph nodes and inflamed tissues duringhealth and disease. They are able to form stable interactions, with profound effects on thephenotype and function of the T cells. However, the outcome of these effects arefrequently contradictory; in some systems neutrophils suppress T cell proliferation, inothers they are activatory or present antigen directly. Published protocols modelling theseinteractions in vitro do not reflect the full range of interactions found in vivo; they do notexamine how activated and naïve T cells differentially respond to neutrophils, or whetherde-granulating or resting neutrophils induce different outcomes. Here, we established aculture protocol to ask these questions with human T cells and autologous neutrophils.We find that resting neutrophils suppress T cell proliferation, activation and cytokineproduction but that de-granulating neutrophils do not, and neutrophil-releasedintracellular contents enhance proliferation. Strikingly, we also demonstrate that T cellsearly in the activation process are susceptible to suppression by neutrophils, while laterstage T cells are not, and naïve T cells do not respond at all. Our protocol therefore allowsnuanced analysis of the outcome of interaction of these cells and may explain thecontradictory results observed previously

    Orchestration of Adaptive T Cell Responses by Neutrophil Granule Contents

    Get PDF
    Neutrophils are the most abundant leukocytes in peripheral blood and respond rapidly to danger, infiltrating tissues within minutes of infectious or sterile injury. Neutrophils were long thought of as simple killers, but now we recognise them as responsive cells able to adapt to inflammation and orchestrate subsequent events with some sophistication. Here, we discuss how these rapid responders release mediators which influence later adaptive T cell immunity through influences on DC priming and directly on the T cells themselves. We consider how the release of granule contents by neutrophils—through NETosis or degranulation—is one way in which the innate immune system directs the phenotype of the adaptive immune response

    High purity isolation of low density neutrophils casts doubt on their exceptionality in health and disease

    Get PDF
    Low density neutrophils (LDNs) are described in a number of inflammatory conditions, cancers and infections and associated with immunopathology, and a mechanistic role in disease. The role of LDNs at homeostasis in healthy individuals has not been investigated. We have developed an isolation protocol that generates high purity LDNs from healthy donors. Healthy LDNs were identical to healthy normal density neutrophils (NDNs), aside from reduced neutrophil extracellular trap formation. CD66b, CD16, CD15, CD10, CD54, CD62L, CXCR2, CD47 and CD11b were expressed at equivalent levels in healthy LDNs and NDNs and underwent apoptosis and ROS production interchangeably. Healthy LDNs had no differential effect on CD4(+) or CD8(+) T cell proliferation or IFNγ production compared with NDNs. LDNs were generated from healthy NDNs in vitro by activation with TNF, LPS or fMLF, suggesting a mechanism of LDN generation in disease however, we show neutrophilia in people with Cystic Fibrosis (CF) was not due to increased LDNs. LDNs are present in the neutrophil pool at homeostasis and have limited functional differences to NDNs. We conclude that increased LDN numbers in disease reflect the specific pathology or inflammatory environment and that neutrophil density alone is inadequate to classify discrete functional populations of neutrophils

    The antimicrobial peptide cathelicidin drives development of experimental autoimmune encephalomyelitis in mice by affecting Th17 differentiation

    Get PDF
    Multiple sclerosis (MS) is a highly prevalent demyelinating autoimmune condition; the mechanisms regulating its severity and progression are unclear. The IL-17-producing Th17 subset of T cells has been widely implicated in MS and in the mouse model, experimental autoimmune encephalomyelitis (EAE). However, the differentiation and regulation of Th17 cells during EAE remain incompletely understood. Although evidence is mounting that the antimicrobial peptide cathelicidin profoundly affects early T cell differentiation, no studies have looked at its role in longer-term T cell responses. Now, we report that cathelicidin drives severe EAE disease. It is released from neutrophils, microglia, and endothelial cells throughout disease; its interaction with T cells potentiates Th17 differentiation in lymph nodes and Th17 to exTh17 plasticity and IFN-γ production in the spinal cord. As a consequence, mice lacking cathelicidin are protected from severe EAE. In addition, we show that cathelicidin is produced by the same cell types in the active brain lesions in human MS disease. We propose that cathelicidin exposure results in highly activated, cytokine-producing T cells, which drive autoimmunity; this is a mechanism through which neutrophils amplify inflammation in the central nervous system

    The neutrophil antimicrobial peptide cathelicidin promotes Th17 differentiation

    Get PDF
    The host defence peptide cathelicidin (LL-37 in humans, mCRAMP in mice) is released from neutrophils by de-granulation, NETosis and necrotic death; it has potent anti-pathogen activity as well as being a broad immunomodulator. Here we report that cathelicidin is a powerful Th17 potentiator which enhances aryl hydrocarbon receptor (AHR) and RORγt expression, in a TGF-β1-dependent manner. In the presence of TGF-β1, cathelicidin enhanced SMAD2/3 and STAT3 phosphorylation, and profoundly suppressed IL-2 and T-bet, directing T cells away from Th1 and into a Th17 phenotype. Strikingly, Th17 but not Th1 cells were protected from apoptosis by cathelicidin. We show that cathelicidin is released by neutrophils in mouse lymph nodes and that cathelicidin-deficient mice display suppressed Th17 responses during inflammation, but not at steady state. We propose that the neutrophil cathelicidin is required for maximal Th17 differentiation, and that this is one method by which early neutrophilia directs subsequent adaptive immune responses

    Interleukin-22 orchestrates a pathological endoplasmic reticulum stress response transcriptional programme in colonic epithelial cells.

    Get PDF
    OBJECTIVE: The functional role of interleukin-22 (IL22) in chronic inflammation is controversial, and mechanistic insights into how it regulates target tissue are lacking. In this study, we evaluated the functional role of IL22 in chronic colitis and probed mechanisms of IL22-mediated regulation of colonic epithelial cells. DESIGN: To investigate the functional role of IL22 in chronic colitis and how it regulates colonic epithelial cells, we employed a three-dimentional mini-gut epithelial organoid system, in vivo disease models and transcriptomic datasets in human IBD. RESULTS: As well as inducing transcriptional modules implicated in antimicrobial responses, IL22 also coordinated an endoplasmic reticulum (ER) stress response transcriptional programme in colonic epithelial cells. In the colon of patients with active colonic Crohn's disease (CD), there was enrichment of IL22-responsive transcriptional modules and ER stress response modules. Strikingly, in an IL22-dependent model of chronic colitis, targeting IL22 alleviated colonic epithelial ER stress and attenuated colitis. Pharmacological modulation of the ER stress response similarly impacted the severity of colitis. In patients with colonic CD, antibody blockade of IL12p40, which simultaneously blocks IL12 and IL23, the key upstream regulator of IL22 production, alleviated the colonic epithelial ER stress response. CONCLUSIONS: Our data challenge perceptions of IL22 as a predominantly beneficial cytokine in IBD and provide novel insights into the molecular mechanisms of IL22-mediated pathogenicity in chronic colitis. Targeting IL22-regulated pathways and alleviating colonic epithelial ER stress may represent promising therapeutic strategies in patients with colitis. TRIAL REGISTRATION NUMBER: NCT02749630

    The antimicrobial peptide cathelicidin drives development of experimental autoimmune encephalomyelitis in mice by affecting Th17 differentiation

    No full text
    Multiple sclerosis (MS) is a highly prevalent demyelinating autoimmune condition; the mechanisms regulating its severity and progression are unclear. The IL-17-producing Th17 subset of T cells has been widely implicated in MS and in the mouse model, experimental autoimmune encephalomyelitis (EAE). However, the differentiation and regulation of Th17 cells during EAE remain incompletely understood. Although evidence is mounting that the antimicrobial peptide cathelicidin profoundly affects early T cell differentiation, no studies have looked at its role in longer-term T cell responses. Now, we report that cathelicidin drives severe EAE disease. It is released from neutrophils, microglia, and endothelial cells throughout disease; its interaction with T cells potentiates Th17 differentiation in lymph nodes and Th17 to exTh17 plasticity and IFN-γ production in the spinal cord. As a consequence, mice lacking cathelicidin are protected from severe EAE. In addition, we show that cathelicidin is produced by the same cell types in the active brain lesions in human MS disease. We propose that cathelicidin exposure results in highly activated, cytokine-producing T cells, which drive autoimmunity; this is a mechanism through which neutrophils amplify inflammation in the central nervous system
    corecore