42 research outputs found

    Hermansky-Pudlak syndrome type 1 causes impaired anti-microbial immunity and inflammation due to dysregulated immunometabolism

    Get PDF
    Hermansky-Pudlak syndrome (HPS) types 1 and 4 are caused by defective vesicle trafficking. The mechanism for Crohn's disease-like inflammation, lung fibrosis, and macrophage lipid accumulation in these patients remains enigmatic. The aim of this study is to understand the cellular basis of inflammation in HPS-1. We performed mass cytometry, proteomic and transcriptomic analyses to investigate peripheral blood cells and serum of HPS-1 patients. Using spatial transcriptomics, granuloma-associated signatures in the tissue of an HPS-1 patient with granulomatous colitis were dissected. In vitro studies were conducted to investigate anti-microbial responses of HPS-1 patient macrophages and cell lines. Monocytes of HPS-1 patients exhibit an inflammatory phenotype associated with dysregulated TNF, IL-1α, OSM in serum, and monocyte-derived macrophages. Inflammatory macrophages accumulate in the intestine and granuloma-associated macrophages in HPS-1 show transcriptional signatures suggestive of a lipid storage and metabolic defect. We show that HPS1 deficiency leads to an altered metabolic program and Rab32-dependent amplified mTOR signaling, facilitated by the accumulation of mTOR on lysosomes. This pathogenic mechanism translates into aberrant bacterial clearance, which can be rescued with mTORC1 inhibition. Rab32-mediated mTOR signaling acts as an immuno-metabolic checkpoint, adding to the evidence that defective bioenergetics can drive hampered anti-microbial activity and contribute to inflammation

    Immunodeficiency, autoimmune thrombocytopenia and enterocolitis caused by autosomal recessive deficiency of PIK3CD-encoded phosphoinositide 3-kinase ÎŽ.

    Get PDF
    Phosphoinositide 3-kinase Ύ (PI3KΎ), a lipid kinase consisting of a catalytic (p110Ύ, encoded by PIK3CD) and a regulatory subunit (p85, encoded by PIK3R1), generates the second messenger phosphatidylinositol (3,4,5)-trisphosphate (PIP3) in the plasma membrane of leukocytes downstream of antigen and cytokine receptors.1 Signaling via PDK1, AKT, mTOR and downstream targets such as FOXO1, contributes to the metabolic and transcriptional changes required for the expansion, differentiation and effector function of lymphocytes. Activating germline mutations in PIK3CD cause the immune dysregulatory disease activated PI3KΎ syndrome (APDS), usually presenting with recurrent sinopulmonary infections in childhood, herpesvirus infections and CD4+ lymphopenia, underscoring the important role of balanced p110Ύ activity in human adaptive immunity. Ablation of p110Ύ in mice leads to aberrant T cell responses and intestinal inflammation. In humans, immune dysregulation including severe colitis is present in many cancer patients who are treated with the p110Ύ-specific inhibitor Idelalisib. Recently, one patient with autosomal recessive deficiency of p85α and two patients with loss-of function mutations in p110Ύ have been described who developed humoral immunodeficiency and colitis

    The role and uses of antibodies in COVID-19 infections: a living review

    Get PDF
    Coronavirus disease 2019 has generated a rapidly evolving field of research, with the global scientific community striving for solutions to the current pandemic. Characterizing humoral responses towards SARS-CoV-2, as well as closely related strains, will help determine whether antibodies are central to infection control, and aid the design of therapeutics and vaccine candidates. This review outlines the major aspects of SARS-CoV-2-specific antibody research to date, with a focus on the various prophylactic and therapeutic uses of antibodies to alleviate disease in addition to the potential of cross-reactive therapies and the implications of long-term immunity

    Somatic mosaicism and common genetic variation contribute to the risk of very-early-onset inflammatory bowel disease

    Get PDF
    Abstract: Very-early-onset inflammatory bowel disease (VEO-IBD) is a heterogeneous phenotype associated with a spectrum of rare Mendelian disorders. Here, we perform whole-exome-sequencing and genome-wide genotyping in 145 patients (median age-at-diagnosis of 3.5 years), in whom no Mendelian disorders were clinically suspected. In five patients we detect a primary immunodeficiency or enteropathy, with clinical consequences (XIAP, CYBA, SH2D1A, PCSK1). We also present a case study of a VEO-IBD patient with a mosaic de novo, pathogenic allele in CYBB. The mutation is present in ~70% of phagocytes and sufficient to result in defective bacterial handling but not life-threatening infections. Finally, we show that VEO-IBD patients have, on average, higher IBD polygenic risk scores than population controls (99 patients and 18,780 controls; P < 4 × 10−10), and replicate this finding in an independent cohort of VEO-IBD cases and controls (117 patients and 2,603 controls; P < 5 × 10−10). This discovery indicates that a polygenic component operates in VEO-IBD pathogenesis

    T cell phenotypes in COVID-19 - a living review

    Get PDF
    COVID-19 is characterized by profound lymphopenia in the peripheral blood, and the remaining T cells display altered phenotypes, characterized by a spectrum of activation and exhaustion. However, antigen-specific T cell responses are emerging as a crucial mechanism for both clearance of the virus and as the most likely route to long-lasting immune memory that would protect against re-infection. Therefore, T cell responses are also of considerable interest in vaccine development. Furthermore, persistent alterations in T cell subset composition and function post-infection have important implications for patients’ long-term immune function. In this review, we examine T cell phenotypes, including those of innate T cells, in both peripheral blood and lungs, and consider how key markers of activation and exhaustion correlate with, and may be able to predict, disease severity. We focus on SARS-CoV-2-specific T cells to elucidate markers that may indicate formation of antigen-specific T cell memory. We also examine peripheral T cell phenotypes in recovery and the likelihood of long-lasting immune disruption. Finally, we discuss T cell phenotypes in the lung as important drivers of both virus clearance and tissue damage. As our knowledge of the adaptive immune response to COVID-19 rapidly evolves, it has become clear that while some areas of the T cell response have been investigated in some detail, others, such as the T cell response in children remain largely unexplored. Therefore, this review will also highlight areas where T cell phenotypes require urgent characterisation

    Anti-microbial activity and inflammation in Hermansky-Pudlak Syndrome type 1

    No full text
    Inflammatory bowel disease (IBD) is caused by a breakdown in tolerance of the immune system to the gut microbiota in genetically predisposed individuals. The mechanisms underlying IBD are complex and multifactorial. Monogenic diseases with intestinal inflammation provide important puzzle pieces in identifying key mechanisms of barrier function. Hermansky-Pudlak Syndrome type 1 (HPS-1) features defective vesicle trafficking and can include complications such as Crohn’s-like intestinal inflammation and pulmonary fibrosis. In this thesis, I pinpoint aberrant immunometabolism in HPS-1 patient macrophages using transcriptomics and functional assays. Following bacterial infection, HPS-1 macrophages feature an augmented TNF/OSM signature, which underlies polygenic IBD. I validate the deregulated immunometabolism in HPS1 knockout HAP1 cells, which have defective cholesterol metabolism and enhanced low density lipoprotein uptake. Using lysosomal immunoprecipitation and proteomics, I identify increased mTOR levels on HPS1 knockout lysosomes, which translates into enhanced mTORC1 signaling. The amplified mTORC1 signaling in HPS1 deficiency is mediated by Rab32. The alterations in metabolism and mTORC1 activation cause impaired anti-bacterial immunity in HPS1 knockout cells and HPS-1 patient macrophages. Mass cytometry of HPS-1 peripheral blood reveals an expansion of inflammatory monocytes, while serum proteomics uncovers a TNF/IL-1α signature in HPS- 1. Suppression of mTORC1 signaling through rapamycin treatment rescues the bacterial clearance defect in HPS-1 patient macrophages, providing a novel therapeutic target for HPS- IBD. These findings have implications beyond HPS, since augmented mTOR signaling and derailed metabolism can trigger inflammation in several diseases, such as lysosomal storage disorders
    corecore