Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27.292
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 39: 719-757, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33646859

ABSTRACT

The enigmatic eosinophil has emerged as an exciting component of the immune system, involved in a plethora of homeostatic and inflammatory responses. Substantial progress has been achieved through experimental systems manipulating eosinophils in vivo, initially in mice and more recently in humans. Researchers using eosinophil knockout mice have identified a contributory role for eosinophils in basal and inflammatory processes and protective immunity. Primarily fueled by the purported proinflammatory role of eosinophils in eosinophil-associated diseases, a series of anti-eosinophil therapeutics have emerged as a new class of drugs. These agents, which dramatically deplete eosinophils, provide a valuable opportunity to characterize the consequences of eosinophil knockout humans. Herein, we comparatively describe mouse and human eosinophil knockouts. We put forth the view that human eosinophils negatively contribute to a variety of diseases and, unlike mouse eosinophils, do not yet have an identified role in physiological health; thus, clarifying all roles of eosinophils remains an ongoing pursuit.


Subject(s)
Eosinophils , Pharmaceutical Preparations , Animals , Biological Therapy , Humans , Mice , Mice, Knockout
2.
Nat Immunol ; 25(8): 1367-1382, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38992254

ABSTRACT

Upregulation of diverse self-antigens that constitute components of the inflammatory response overlaps spatially and temporally with the emergence of pathogen-derived foreign antigens. Therefore, discrimination between these inflammation-associated self-antigens and pathogen-derived molecules represents a unique challenge for the adaptive immune system. Here, we demonstrate that CD8+ T cell tolerance to T cell-derived inflammation-associated self-antigens is efficiently induced in the thymus and supported by redundancy in cell types expressing these molecules. In addition to thymic epithelial cells, this included thymic eosinophils and innate-like T cells, a population that expressed molecules characteristic for all major activated T cell subsets. We show that direct T cell-to-T cell antigen presentation by minute numbers of innate-like T cells was sufficient to eliminate autoreactive CD8+ thymocytes. Tolerance to such effector molecules was of critical importance, as its breach caused by decreased thymic abundance of a single model inflammation-associated self-antigen resulted in autoimmune elimination of an entire class of effector T cells.


Subject(s)
Antigen Presentation , Autoantigens , CD8-Positive T-Lymphocytes , Inflammation , Thymocytes , Thymus Gland , Animals , Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Mice , Thymus Gland/immunology , Inflammation/immunology , Antigen Presentation/immunology , Thymocytes/immunology , Thymocytes/metabolism , Mice, Inbred C57BL , Immunity, Innate , Autoimmunity/immunology , Immune Tolerance/immunology , Mice, Transgenic , Mice, Knockout , Lymphocyte Activation/immunology , Eosinophils/immunology
3.
Nat Immunol ; 25(6): 1059-1072, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38802511

ABSTRACT

Asthma, the most prevalent respiratory disease, affects more than 300 million people and causes more than 250,000 deaths annually. Type 2-high asthma is characterized by interleukin (IL)-5-driven eosinophilia, along with airway inflammation and remodeling caused by IL-4 and IL-13. Here we utilize IL-5 as the targeting domain and deplete BCOR and ZC3H12A to engineer long-lived chimeric antigen receptor (CAR) T cells that can eradicate eosinophils. We call these cells immortal-like and functional IL-5 CAR T cells (5TIF) cells. 5TIF cells were further modified to secrete an IL-4 mutein that blocks IL-4 and IL-13 signaling, designated as 5TIF4 cells. In asthma models, a single infusion of 5TIF4 cells in fully immunocompetent mice, without any conditioning regimen, led to sustained repression of lung inflammation and alleviation of asthmatic symptoms. These data show that asthma, a common chronic disease, can be pushed into long-term remission with a single dose of long-lived CAR T cells.


Subject(s)
Asthma , Receptors, Chimeric Antigen , Animals , Asthma/immunology , Asthma/therapy , Mice , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , Interleukin-5/immunology , Interleukin-5/metabolism , Disease Models, Animal , Humans , Interleukin-4/immunology , Interleukin-4/metabolism , Mice, Inbred C57BL , Eosinophils/immunology , Female , Interleukin-13/metabolism , Interleukin-13/immunology
4.
Cell ; 184(23): 5715-5727.e12, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34717799

ABSTRACT

The enteric nervous system (ENS) controls several intestinal functions including motility and nutrient handling, which can be disrupted by infection-induced neuropathies or neuronal cell death. We investigated possible tolerance mechanisms preventing neuronal loss and disruption in gut motility after pathogen exposure. We found that following enteric infections, muscularis macrophages (MMs) acquire a tissue-protective phenotype that prevents neuronal loss, dysmotility, and maintains energy balance during subsequent challenge with unrelated pathogens. Bacteria-induced neuroprotection relied on activation of gut-projecting sympathetic neurons and signaling via ß2-adrenergic receptors (ß2AR) on MMs. In contrast, helminth-mediated neuroprotection was dependent on T cells and systemic production of interleukin (IL)-4 and IL-13 by eosinophils, which induced arginase-expressing MMs that prevented neuronal loss from an unrelated infection located in a different intestinal region. Collectively, these data suggest that distinct enteric pathogens trigger a state of disease or tissue tolerance that preserves ENS number and functionality.


Subject(s)
Enteric Nervous System/microbiology , Enteric Nervous System/parasitology , Infections/microbiology , Infections/parasitology , Neurons/pathology , Neuroprotection , Organ Specificity , Yersinia pseudotuberculosis/physiology , Animals , Eosinophils/metabolism , Hematopoietic Stem Cells/metabolism , Immunity , Infections/immunology , Interleukin-13/metabolism , Interleukin-4/metabolism , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Strongyloides/physiology , Strongyloidiasis/genetics , Strongyloidiasis/immunology , Strongyloidiasis/parasitology , Transcriptome/genetics , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis Infections/microbiology
5.
Nat Immunol ; 23(10): 1484-1494, 2022 10.
Article in English | MEDLINE | ID: mdl-36138182

ABSTRACT

The heterogeneous cellular microenvironment of human airway chronic inflammatory diseases, including chronic rhinosinusitis (CRS) and asthma, is still poorly understood. Here, we performed single-cell RNA sequencing (scRNA-seq) on the nasal mucosa of healthy individuals and patients with three subtypes of CRS and identified disease-specific cell subsets and molecules that specifically contribute to the pathogenesis of CRS subtypes. As such, ALOX15+ macrophages contributed to the type 2 immunity-driven pathogenesis of one subtype of CRS, eosinophilic CRS with nasal polyps (eCRSwNP), by secreting chemokines that recruited eosinophils, monocytes and T helper 2 (TH2) cells. An inhibitor of ALOX15 reduced the release of proinflammatory chemokines in human macrophages and inhibited the overactivation of type 2 immunity in a mouse model of eosinophilic rhinosinusitis. Our findings advance the understanding of the heterogeneous immune microenvironment and the pathogenesis of CRS subtypes and identify potential therapeutic approaches for the treatment of CRS and potentially other type 2 immunity-mediated diseases.


Subject(s)
Nasal Polyps , Rhinitis , Sinusitis , Animals , Chronic Disease , Eosinophils , Humans , Mice , Nasal Mucosa
6.
Nat Immunol ; 23(9): 1309-1316, 2022 09.
Article in English | MEDLINE | ID: mdl-36002647

ABSTRACT

Eosinophils are important effector cells and therapeutic targets in allergic diseases. Emerging data indicate that eosinophils infiltrate a variety of solid tumor types and have pleiotropic activities by at least two non-mutually exclusive mechanisms: direct interactions with tumor cells, and intricate cross-talk with lymphocytes. In light of the immune checkpoint inhibition revolution in cancer therapy, we review eosinophil-lymphocyte interactions in the tumor microenvironment. We also analyze potential interactions between eosinophils and lymphocyte subsets, including T cells, natural killer cells and innate lymphoid cells. We provide perspectives on the consequences of these interactions and how eosinophils are accessory cells that can affect the response to various forms of T cell-mediated immunotherapies and might be therapeutically targeted to improve cancer immunotherapy.


Subject(s)
Neoplasms , Tumor Microenvironment , Eosinophils , Humans , Immune Checkpoint Inhibitors , Immunity, Innate , Immunotherapy , Killer Cells, Natural/pathology
7.
Nat Immunol ; 22(10): 1210-1217, 2021 10.
Article in English | MEDLINE | ID: mdl-34545250

ABSTRACT

When helper T (TH) cell polarization was initially described three decades ago, the TH cell universe grew dramatically. New subsets were described based on their expression of few specific cytokines. Beyond TH1 and TH2 cells, this led to the coining of various TH17 and regulatory (Treg) cell subsets as well as TH22, TH25, follicular helper (TFH), TH3, TH5 and TH9 cells. High-dimensional single-cell analysis revealed that a categorization of TH cells into a single-cytokine-based nomenclature fails to capture the complexity and diversity of TH cells. Similar to the simple nomenclature used to describe innate lymphoid cells (ILCs), we propose that TH cell polarization should be categorized in terms of the help they provide to phagocytes (type 1), to B cells, eosinophils and mast cells (type 2) and to non-immune tissue cells, including the stroma and epithelium (type 3). Studying TH cells based on their helper function and the cells they help, rather than phenotypic features such as individual analyzed cytokines or transcription factors, better captures TH cell plasticity and conversion as well as the breadth of immune responses in vivo.


Subject(s)
Cytokines/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , B-Lymphocytes/immunology , Cell Plasticity/immunology , Eosinophils/immunology , Epithelium/immunology , Humans , Immunity, Innate/immunology , Lymphocytes/immunology , Phagocytes/immunology
8.
Nat Immunol ; 22(7): 851-864, 2021 07.
Article in English | MEDLINE | ID: mdl-34099918

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are essential to maintain tissue homeostasis. In cancer, ILC2s can harbor both pro-tumorigenic and anti-tumorigenic functions, but we know little about their underlying mechanisms or whether they could be clinically relevant or targeted to improve patient outcomes. Here, we found that high ILC2 infiltration in human melanoma was associated with a good clinical prognosis. ILC2s are critical producers of the cytokine granulocyte-macrophage colony-stimulating factor, which coordinates the recruitment and activation of eosinophils to enhance antitumor responses. Tumor-infiltrating ILC2s expressed programmed cell death protein-1, which limited their intratumoral accumulation, proliferation and antitumor effector functions. This inhibition could be overcome in vivo by combining interleukin-33-driven ILC2 activation with programmed cell death protein-1 blockade to significantly increase antitumor responses. Together, our results identified ILC2s as a critical immune cell type involved in melanoma immunity and revealed a potential synergistic approach to harness ILC2 function for antitumor immunotherapies.


Subject(s)
Antibodies/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Interleukin-33/pharmacology , Lymphocytes/drug effects , Melanoma, Experimental/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Cell Line, Tumor , Chemotaxis, Leukocyte/drug effects , Cytotoxicity, Immunologic/drug effects , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/metabolism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/metabolism
9.
Immunity ; 57(7): 1549-1566.e8, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38776917

ABSTRACT

The activities, ontogeny, and mechanisms of lineage expansion of eosinophils are less well resolved than those of other immune cells, despite the use of biological therapies targeting the eosinophilia-promoting cytokine interleukin (IL)-5 or its receptor, IL-5Rα. We combined single-cell proteomics and transcriptomics and generated transgenic IL-5Rα reporter mice to revisit eosinophilopoiesis. We reconciled human and murine eosinophilopoiesis and provided extensive cell-surface immunophenotyping and transcriptomes at different stages along the continuum of eosinophil maturation. We used these resources to show that IL-5 promoted eosinophil-lineage expansion via transit amplification, while its deletion or neutralization did not compromise eosinophil maturation. Informed from our resources, we also showed that interferon response factor-8, considered an essential promoter of myelopoiesis, was not intrinsically required for eosinophilopoiesis. This work hence provides resources, methods, and insights for understanding eosinophil ontogeny, the effects of current precision therapeutics, and the regulation of eosinophil development and numbers in health and disease.


Subject(s)
Cell Lineage , Eosinophils , Interleukin-5 , Mice, Transgenic , Proteomics , Single-Cell Analysis , Transcriptome , Eosinophils/immunology , Eosinophils/metabolism , Animals , Interleukin-5/metabolism , Interleukin-5/genetics , Humans , Mice , Proteomics/methods , Single-Cell Analysis/methods , Cell Differentiation/immunology , Mice, Inbred C57BL , Gene Expression Profiling/methods , Interleukin-5 Receptor alpha Subunit/metabolism , Interleukin-5 Receptor alpha Subunit/genetics , Myelopoiesis/genetics , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Mice, Knockout
10.
Nat Immunol ; 21(9): 998-1009, 2020 09.
Article in English | MEDLINE | ID: mdl-32747815

ABSTRACT

Metastasis constitutes the primary cause of cancer-related deaths, with the lung being a commonly affected organ. We found that activation of lung-resident group 2 innate lymphoid cells (ILC2s) orchestrated suppression of natural killer (NK) cell-mediated innate antitumor immunity, leading to increased lung metastases and mortality. Using multiple models of lung metastasis, we show that interleukin (IL)-33-dependent ILC2 activation in the lung is involved centrally in promoting tumor burden. ILC2-driven innate type 2 inflammation is accompanied by profound local suppression of interferon-γ production and cytotoxic function of lung NK cells. ILC2-dependent suppression of NK cells is elaborated via an innate regulatory mechanism, which is reliant on IL-5-induced lung eosinophilia, ultimately limiting the metabolic fitness of NK cells. Therapeutic targeting of IL-33 or IL-5 reversed NK cell suppression and alleviated cancer burden. Thus, we reveal an important function of IL-33 and ILC2s in promoting tumor metastasis via their capacity to suppress innate type 1 immunity.


Subject(s)
Eosinophils/immunology , Killer Cells, Natural/immunology , Lung Neoplasms/immunology , Lung/immunology , Lymphocytes/immunology , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Immune Tolerance , Immunity, Innate , Interleukin-33/metabolism , Interleukin-5/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis , Th2 Cells/immunology
11.
Immunity ; 56(7): 1548-1560.e5, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37279752

ABSTRACT

Cryptococcus neoformans is the leading cause of fungal meningitis and is characterized by pathogenic eosinophil accumulation in the context of type-2 inflammation. The chemoattractant receptor GPR35 is expressed by granulocytes and promotes their migration to the inflammatory mediator 5-hydroxyindoleacetic acid (5-HIAA), a serotonin metabolite. Given the inflammatory nature of cryptococcal infection, we examined the role of GPR35 in the circuitry underlying cell recruitment to the lung. GPR35 deficiency dampened eosinophil recruitment and fungal growth, whereas overexpression promoted eosinophil homing to airways and fungal replication. Activated platelets and mast cells were the sources of GPR35 ligand activity and pharmacological inhibition of serotonin conversion to 5-HIAA, or genetic deficiency in 5-HIAA production by platelets and mast cells resulted in more efficient clearance of Cryptococcus. Thus, the 5-HIAA-GPR35 axis is an eosinophil chemoattractant receptor system that modulates the clearance of a lethal fungal pathogen, with implications for the use of serotonin metabolism inhibitors in the treatment of fungal infections.


Subject(s)
Cryptococcosis , Invasive Fungal Infections , Humans , Eosinophils , Hydroxyindoleacetic Acid , Mast Cells , Blood Platelets , Ligands , Receptors, Formyl Peptide , Serotonin , Cryptococcosis/microbiology , Cryptococcosis/pathology , Receptors, G-Protein-Coupled/genetics
12.
Nat Immunol ; 20(3): 257-264, 2019 03.
Article in English | MEDLINE | ID: mdl-30778250

ABSTRACT

Post-translational modification of chemokines mediated by the dipeptidyl peptidase DPP4 (CD26) has been shown to negatively regulate lymphocyte trafficking, and its inhibition enhances T cell migration and tumor immunity by preserving functional chemokine CXCL10. By extending those initial findings to pre-clinical models of hepatocellular carcinoma and breast cancer, we discovered a distinct mechanism by which inhibition of DPP4 improves anti-tumor responses. Administration of the DPP4 inhibitor sitagliptin resulted in higher concentrations of the chemokine CCL11 and increased migration of eosinophils into solid tumors. Enhanced tumor control was preserved in mice lacking lymphocytes and was ablated after depletion of eosinophils or treatment with degranulation inhibitors. We further demonstrated that tumor-cell expression of the alarmin IL-33 was necessary and sufficient for eosinophil-mediated anti-tumor responses and that this mechanism contributed to the efficacy of checkpoint-inhibitor therapy. These findings provide insight into IL-33- and eosinophil-mediated tumor control, revealed when endogenous mechanisms of DPP4 immunoregulation are inhibited.


Subject(s)
Dipeptidyl Peptidase 4/immunology , Eosinophils/immunology , Interleukin-33/immunology , Neoplasms, Experimental/immunology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/immunology , Chemokine CCL11/immunology , Chemokine CCL11/metabolism , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Disease Models, Animal , Eosinophils/drug effects , Eosinophils/metabolism , Humans , Interleukin-33/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/prevention & control , Sitagliptin Phosphate/pharmacology
13.
Immunity ; 55(7): 1148-1150, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35830822

ABSTRACT

A plethora of studies have established the importance of eosinophils in protective immunity against infections and in allergy. In this issue of Immunity, Ignacio et al. (2022) define a vital for eosinophils in coordinating a microbiota-epithelial-immune axis that maintains intestinal homeostasis.


Subject(s)
Intestines , Microbiota , Eosinophils , Homeostasis
14.
Immunity ; 55(7): 1250-1267.e12, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35709757

ABSTRACT

The intestine harbors a large population of resident eosinophils, yet the function of intestinal eosinophils has not been explored. Flow cytometry and whole-mount imaging identified eosinophils residing in the lamina propria along the length of the intestine prior to postnatal microbial colonization. Microscopy, transcriptomic analysis, and mass spectrometry of intestinal tissue revealed villus blunting, altered extracellular matrix, decreased epithelial cell turnover, increased gastrointestinal motility, and decreased lipid absorption in eosinophil-deficient mice. Mechanistically, intestinal epithelial cells released IL-33 in a microbiota-dependent manner, which led to eosinophil activation. The colonization of germ-free mice demonstrated that eosinophil activation in response to microbes regulated villous size alterations, macrophage maturation, epithelial barrier integrity, and intestinal transit. Collectively, our findings demonstrate a critical role for eosinophils in facilitating the mutualistic interactions between the host and microbiota and provide a rationale for the functional significance of their early life recruitment in the small intestine.


Subject(s)
Communicable Diseases , Microbiota , Animals , Eosinophils , Homeostasis , Intestinal Mucosa , Intestine, Small , Mice
15.
Cell ; 157(6): 1249-1250, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24906141

ABSTRACT

Subcutaneous white adipose tissue can be induced to undergo "browning" and acquire thermogenic capacity in response to physiological stimuli such as cold exposure or exercise. In this issue of Cell, Qiu et al. and Rao et al. demonstrate that pink-staining eosinophils and alternatively activated macrophages play key roles in an immune cascade mediating this metabolic switch.


Subject(s)
Adipose Tissue, Brown/metabolism , Eosinophils/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Macrophages/metabolism , Nerve Growth Factors/metabolism , Signal Transduction , Animals , Male
16.
Cell ; 157(6): 1292-1308, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24906148

ABSTRACT

Beige fat, which expresses the thermogenic protein UCP1, provides a defense against cold and obesity. Although a cold environment is the physiologic stimulus for inducing beige fat in mice and humans, the events that lead from the sensing of cold to the development of beige fat remain poorly understood. Here, we identify the efferent beige fat thermogenic circuit, consisting of eosinophils, type 2 cytokines interleukin (IL)-4/13, and alternatively activated macrophages. Genetic loss of eosinophils or IL-4/13 signaling impairs cold-induced biogenesis of beige fat. Mechanistically, macrophages recruited to cold-stressed subcutaneous white adipose tissue (scWAT) undergo alternative activation to induce tyrosine hydroxylase expression and catecholamine production, factors required for browning of scWAT. Conversely, administration of IL-4 to thermoneutral mice increases beige fat mass and thermogenic capacity to ameliorate pre-established obesity. Together, our findings have uncovered the efferent circuit controlling biogenesis of beige fat and provide support for its targeting to treat obesity.


Subject(s)
Adipose Tissue, Brown/metabolism , Eosinophils/metabolism , Interleukin-13/metabolism , Interleukin-4/metabolism , Macrophages/metabolism , Signal Transduction , Adipocytes, Brown/metabolism , Animals , Catecholamines/metabolism , Cold Temperature , Interleukin-13/genetics , Interleukin-4/genetics , Male , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Obesity/metabolism , Receptors, CCR2/metabolism , STAT6 Transcription Factor/metabolism , Thermogenesis
17.
Nature ; 615(7950): 151-157, 2023 03.
Article in English | MEDLINE | ID: mdl-36509106

ABSTRACT

In the past decade, single-cell transcriptomics has helped to uncover new cell types and states and led to the construction of a cellular compendium of health and disease. Despite this progress, some difficult-to-sequence cells remain absent from tissue atlases. Eosinophils-elusive granulocytes that are implicated in a plethora of human pathologies1-5-are among these uncharted cell types. The heterogeneity of eosinophils and the gene programs that underpin their pleiotropic functions remain poorly understood. Here we provide a comprehensive single-cell transcriptomic profiling of mouse eosinophils. We identify an active and a basal population of intestinal eosinophils, which differ in their transcriptome, surface proteome and spatial localization. By means of a genome-wide CRISPR inhibition screen and functional assays, we reveal a mechanism by which interleukin-33 (IL-33) and interferon-γ (IFNγ) induce the accumulation of active eosinophils in the inflamed colon. Active eosinophils are endowed with bactericidal and T cell regulatory activity, and express the co-stimulatory molecules CD80 and PD-L1. Notably, active eosinophils are enriched in the lamina propria of a small cohort of patients with inflammatory bowel disease, and are closely associated with CD4+ T cells. Our findings provide insights into the biology of eosinophils and highlight the crucial contribution of this cell type to intestinal homeostasis, immune regulation and host defence. Furthermore, we lay a framework for the characterization of eosinophils in human gastrointestinal diseases.


Subject(s)
Colitis , Eosinophils , Immunity , Intestines , Animals , Humans , Mice , Colitis/immunology , Colitis/pathology , Eosinophils/classification , Eosinophils/cytology , Eosinophils/immunology , Eosinophils/metabolism , Inflammatory Bowel Diseases/immunology , Single-Cell Gene Expression Analysis , Transcriptome , Proteome , Interleukin-33 , Interferon-gamma , T-Lymphocytes , B7-1 Antigen/metabolism , Intestines/immunology , Intestines/pathology
18.
Nat Immunol ; 17(4): 356-63, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27002843

ABSTRACT

Innate cells are responsible for the rapid recognition of infection and mediate essential mechanisms of pathogen elimination, and also facilitate adaptive immune responses. We review here the numerous intricate interactions among innate cells that initiate protective immunity. The efficient eradication of pathogens depends on the coordinated actions of multiple cells, including innate cells and epithelial cells. Rather than acting as isolated effector cells, innate cells are in constant communication with other responding cells of the immune system, locally and distally. These interactions are critically important for the efficient control of primary infections as well for the development of 'trained' innate cells that facilitate the rapid elimination of homologous or heterologous infections.


Subject(s)
Adaptive Immunity/immunology , Cytokines/immunology , Immunity, Innate/immunology , Infections/immunology , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Animals , Basophils/immunology , Eosinophils/immunology , Humans , Macrophages/immunology , Mast Cells/immunology , Monocytes/immunology , Neutrophils/immunology
19.
Nat Immunol ; 17(6): 636-45, 2016 06.
Article in English | MEDLINE | ID: mdl-27111145

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) secrete type 2 cytokines, which protect against parasites but can also contribute to a variety of inflammatory airway diseases. We report here that interleukin 1ß (IL-1ß) directly activated human ILC2s and that IL-12 induced the conversion of these activated ILC2s into interferon-γ (IFN-γ)-producing ILC1s, which was reversed by IL-4. The plasticity of ILCs was manifested in diseased tissues of patients with severe chronic obstructive pulmonary disease (COPD) or chronic rhinosinusitis with nasal polyps (CRSwNP), which displayed IL-12 or IL-4 signatures and the accumulation of ILC1s or ILC2s, respectively. Eosinophils were a major cellular source of IL-4, which revealed cross-talk between IL-5-producing ILC2s and IL-4-producing eosinophils. We propose that IL-12 and IL-4 govern ILC2 functional identity and that their imbalance results in the perpetuation of type 1 or type 2 inflammation.


Subject(s)
Cell Plasticity , Eosinophils/immunology , Immunity, Innate , Interleukin-12/metabolism , Interleukin-1beta/metabolism , Interleukin-4/metabolism , Lymphocytes/immunology , Nasal Polyps/immunology , Pneumonia/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Rhinitis/immunology , Sinusitis/immunology , Animals , Cell Differentiation , Cells, Cultured , Female , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Mice , Mice, SCID , Th1 Cells/immunology , Th1-Th2 Balance , Th2 Cells/immunology
20.
Immunity ; 50(2): 378-389.e5, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30784579

ABSTRACT

Currently, we lack an understanding of the individual and combinatorial roles for chemokine receptors in the inflammatory process. We report studies on mice with a compound deletion of Ccr1, Ccr2, Ccr3, and Ccr5, which together control monocytic and eosinophilic recruitment to resting and inflamed sites. Analysis of resting tissues from these mice, and mice deficient in each individual receptor, provides clear evidence for redundant use of these receptors in establishing tissue-resident monocytic cell populations. In contrast, analysis of cellular recruitment to inflamed sites provides evidence of specificity of receptor use for distinct leukocyte subtypes and no indication of comprehensive redundancy. We find no evidence of involvement of any of these receptors in the recruitment of neutrophils or lymphocytes to resting or acutely inflamed tissues. Our data shed important light on combinatorial inflammatory chemokine receptor function and highlight Ccr2 as the primary driver of myelomonocytic cell recruitment in acutely inflamed contexts.


Subject(s)
Eosinophils/immunology , Inflammation/immunology , Monocytes/immunology , Receptors, CCR/immunology , Animals , Chemokines/immunology , Chemokines/metabolism , Eosinophils/metabolism , Gene Expression Profiling/methods , Inflammation/genetics , Inflammation/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Receptors, CCR/genetics , Receptors, CCR/metabolism , Receptors, CCR1/immunology , Receptors, CCR1/metabolism , Receptors, CCR2/immunology , Receptors, CCR2/metabolism , Receptors, CCR3/immunology , Receptors, CCR3/metabolism , Receptors, CCR5/immunology , Receptors, CCR5/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL