Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Elife ; 122024 Apr 03.
Article in English | MEDLINE | ID: mdl-38567749

ABSTRACT

Vitamin D possesses immunomodulatory functions and vitamin D deficiency has been associated with the rise in chronic inflammatory diseases, including asthma (Litonjua and Weiss, 2007). Vitamin D supplementation studies do not provide insight into the molecular genetic mechanisms of vitamin D-mediated immunoregulation. Here, we provide evidence for vitamin D regulation of two human chromosomal loci, Chr17q12-21.1 and Chr17q21.2, reliably associated with autoimmune and chronic inflammatory diseases. We demonstrate increased vitamin D receptor (Vdr) expression in mouse lung CD4+ Th2 cells, differential expression of Chr17q12-21.1 and Chr17q21.2 genes in Th2 cells based on vitamin D status and identify the IL-2/Stat5 pathway as a target of vitamin D signaling. Vitamin D deficiency caused severe lung inflammation after allergen challenge in mice that was prevented by long-term prenatal vitamin D supplementation. Mechanistically, vitamin D induced the expression of the Ikzf3-encoded protein Aiolos to suppress IL-2 signaling and ameliorate cytokine production in Th2 cells. These translational findings demonstrate mechanisms for the immune protective effect of vitamin D in allergic lung inflammation with a strong molecular genetic link to the regulation of both Chr17q12-21.1 and Chr17q21.2 genes and suggest further functional studies and interventional strategies for long-term prevention of asthma and other autoimmune disorders.


Subject(s)
Asthma , Pneumonia , Vitamin D Deficiency , Mice , Animals , Humans , Vitamin D/pharmacology , Interleukin-2 , Inflammation , Th2 Cells , Vitamin D Deficiency/metabolism , Vitamins
2.
Proc Natl Acad Sci U S A ; 120(2): e2206480120, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36595677

ABSTRACT

The resolution of infection is an active process with specific molecular and cellular mechanisms that temper inflammation and enhance pathogen clearance. Here, the specialized pro-resolving mediator (SPM) Maresin 1 (MaR1) inhibited respiratory syncytial virus (RSV)-induced inflammation. inlerleukin-13 production from type 2 innate lymphoid cells (ILC) and CD4 T helper type 2 cells was decreased by exogenous MaR1. In addition, MaR1 increased amphiregulin production and decreased RSV viral transcripts to promote resolution. MaR1 also promoted interferon-ß production in mouse lung tissues and also in pediatric lung slices. MaR1 significantly inhibited the RSV-triggered aberrant inflammatory phenotype in FoxP3-expressing Tregs. The receptor for MaR1, leucine-rich repeat-containing G protein-coupled receptor 6 (LGR6), was constitutively expressed on Tregs. Following RSV infection, mice lacking Lgr6 had exacerbated type 2 immune responses with an increased viral burden and blunted responses to MaR1. Together, these findings have uncovered a multi-pronged protective signaling axis for MaR1-Lgr6, improving Tregs's suppressive function and upregulating host antiviral genes resulting in decreased viral burden and pathogen-mediated inflammation, ultimately promoting restoration of airway mucosal homeostasis.


Subject(s)
Pneumonia, Viral , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Mice , Animals , Immunity, Innate , Lymphocytes , Inflammation , Docosahexaenoic Acids/pharmacology , Receptors, G-Protein-Coupled
3.
iScience ; 25(10): 105185, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36238896

ABSTRACT

Lipid phosphate phosphatases are a family of enzymes with diverse cellular metabolic functions. Phospholipid phosphatase 6 (PLPP6) is a regulator of cellular polyisoprenyl phosphates; however, its in vivo functions remain to be determined. Here, mouse PLPP6 was characterized to possess similar catalytic properties as the human enzyme. Plpp6 knockout mice (Plpp6 -/- ) were generated and displayed decreased airway allergen sensitization, pointing to a role for PLPP6 in the early events of lung allergic responses. Dendritic cell (DC) responses were investigated and endocytosis of allergen via macropinocytosis was decreased in Plpp6 -/- DCs that had lower cholesterol content. When reversed by cholesterol loading, the DC macropinocytosis defect is corrected. Adoptive transfer of Plpp6 -/- DCs to wild-type mice during sensitization was sufficient to decrease allergen-induced responses. Together, our findings have identified PLPP6 as a pivotal regulator of DC cholesterol content and macropinocytosis, cellular mechanisms that are important for pathologic responses in allergen-induced lung inflammation.

4.
Front Immunol ; 12: 704427, 2021.
Article in English | MEDLINE | ID: mdl-34489955

ABSTRACT

Viral pneumonias are a major cause of morbidity and mortality, owing in part to dysregulated excessive lung inflammation, and therapies to modulate host responses to viral lung injury are urgently needed. Protectin conjugates in tissue regeneration 1 (PCTR1) and protectin D1 (PD1) are specialized pro-resolving mediators (SPMs) whose roles in viral pneumonia are of interest. In a mouse model of Respiratory Syncytial Virus (RSV) pneumonia, intranasal PCTR1 and PD1 each decreased RSV genomic viral load in lung tissue when given after RSV infection. Concurrent with enhanced viral clearance, PCTR1 administration post-infection, decreased eosinophils, neutrophils, and NK cells, including NKG2D+ activated NK cells, in the lung. Intranasal PD1 administration post-infection decreased lung eosinophils and Il-13 expression. PCTR1 increased lung expression of cathelicidin anti-microbial peptide and decreased interferon-gamma production by lung CD4+ T cells. PCTR1 and PD1 each increased interferon-lambda expression in human bronchial epithelial cells in vitro and attenuated RSV-induced suppression of interferon-lambda in mouse lung in vivo. Liquid chromatography coupled with tandem mass spectrometry of RSV-infected and untreated mouse lungs demonstrated endogenous PCTR1 and PD1 that decreased early in the time course while cysteinyl-leukotrienes (cys-LTs) increased during early infection. As RSV infection resolved, PCTR1 and PD1 increased and cys-LTs decreased to pre-infection levels. Together, these results indicate that PCTR1 and PD1 are each regulated during RSV pneumonia, with overlapping and distinct mechanisms for PCTR1 and PD1 during the resolution of viral infection and its associated inflammation.


Subject(s)
Docosahexaenoic Acids/pharmacology , Lung/immunology , Pneumonia, Viral , Respiratory Syncytial Virus Infections , Respiratory Syncytial Viruses/immunology , Administration, Intranasal , Animals , Inflammation/immunology , Inflammation/prevention & control , Lung/virology , Male , Mice , Pneumonia, Viral/immunology , Pneumonia, Viral/prevention & control , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Viral Load/drug effects
5.
Immunity ; 51(6): 1102-1118.e7, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31757673

ABSTRACT

Young children are more susceptible to developing allergic asthma than adults. As neural innervation of the peripheral tissue continues to develop after birth, neurons may modulate tissue inflammation in an age-related manner. Here we showed that sympathetic nerves underwent a dopaminergic-to-adrenergic transition during post-natal development of the lung in mice and humans. Dopamine signaled through a specific dopamine receptor (DRD4) to promote T helper 2 (Th2) cell differentiation. The dopamine-DRD4 pathway acted synergistically with the cytokine IL-4 by upregulating IL-2-STAT5 signaling and reducing inhibitory histone trimethylation at Th2 gene loci. In murine models of allergen exposure, the dopamine-DRD4 pathway augmented Th2 inflammation in the lungs of young mice. However, this pathway operated marginally after sympathetic nerves became adrenergic in the adult lung. Taken together, the communication between dopaminergic nerves and CD4+ T cells provides an age-related mechanism underlying the susceptibility to allergic inflammation in the early lung.


Subject(s)
Adrenergic Neurons/cytology , Asthma/pathology , Dopamine/metabolism , Dopaminergic Neurons/cytology , Lung/pathology , Th2 Cells/immunology , Adolescent , Adult , Age Factors , Aged , Animals , Asthma/immunology , Cells, Cultured , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Interleukin-2/metabolism , Interleukin-4/immunology , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neurogenesis/physiology , Receptors, Dopamine D4/metabolism , STAT5 Transcription Factor/metabolism , Sympathetic Nervous System/cytology
6.
Nat Biomed Eng ; 3(12): 961-973, 2019 12.
Article in English | MEDLINE | ID: mdl-31712645

ABSTRACT

Dysregulated leukocyte responses underlie the pathobiology of sepsis, which is a leading cause of death. However, measures of leukocyte function are not routinely available in clinical care. Here we report the development and testing of an inertial microfluidic system for the label-free isolation and downstream functional assessment of leukocytes from 50 µl of peripheral blood. We used the system to assess leukocyte phenotype and function in serial samples from 18 hospitalized patients with sepsis and 10 healthy subjects. The sepsis samples had significantly higher levels of CD16dim and CD16- neutrophils and CD16+ 'intermediate' monocytes, as well as significantly lower levels of neutrophil-elastase release, O2- production and phagolysosome formation. Repeated sampling of sepsis patients over 7 days showed that leukocyte activation (measured by isodielectric separation) and leukocyte phenotype and function were significantly more predictive of the clinical course than complete-blood-count parameters. We conclude that the serial assessment of leukocyte function in microlitre blood volumes is feasible and that it provides significantly more prognostic information than leukocyte counting.


Subject(s)
Leukocytes , Microfluidic Analytical Techniques/methods , Sepsis/blood , Sepsis/diagnosis , Severity of Illness Index , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Female , GPI-Linked Proteins , Humans , Leukocyte Count , Leukocyte Elastase/blood , Male , Microfluidic Analytical Techniques/instrumentation , Middle Aged , Monocytes , Neutrophils , Phenotype , Receptors, IgG , Young Adult
7.
Allergol Int ; 68(2): 143-149, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30573389

ABSTRACT

Asthma is a highly prevalent heterogeneous inflammatory disorder of the airways. Not all patients respond to anti-inflammatory treatment with corticosteroids, leading to significant morbidity in severe asthma. Much attention has been paid to defining the cellular and molecular mechanisms of type 2 inflammation that are operative in asthma. Development of targeted therapies for pathologic type 2 inflammation is opening a new approach to asthma treatment; however, not all asthmatics have type 2 airway inflammation, especially those with severe corticosteroid-refractory asthma. Much less is known about non-type 2 immunological mechanisms in asthma. In health, inflammation triggers resolution mechanisms that control immune (type 1 and type 2) responses and enable the restoration of tissue homeostasis. The resolution response is comprised of cellular and molecular events, including production of specialized pro-resolving mediators (SPMs). SPMs halt leukocyte recruitment, promote macrophage efferocytosis, and restore epithelial barrier integrity, all of which are critical to resolution of inflammation in the lungs. Here, we review recent insights into the disruption of these homeostatic mechanisms and their contributions to non-type 2 inflammation in severe asthma immunopathogenesis.


Subject(s)
Asthma/immunology , Inflammation/immunology , Neutrophils/immunology , Animals , Humans , Inflammation Mediators/immunology , Killer Cells, Natural/immunology , Neutrophils/cytology , Severity of Illness Index
8.
Sci Immunol ; 3(26)2018 08 03.
Article in English | MEDLINE | ID: mdl-30076281

ABSTRACT

Severe asthma is a debilitating and treatment refractory disease. As many as half of these patients have complex neutrophil-predominant lung inflammation that is distinct from milder asthma with type 2 eosinophilic inflammation. New insights into severe asthma pathogenesis are needed. Concomitant exposure of mice to an aeroallergen and endotoxin during sensitization resulted in complex neutrophilic immune responses to allergen alone during later airway challenge. Unlike allergen alone, sensitization with allergen and endotoxin led to NETosis. In addition to neutrophil extracellular traps (NETs), enucleated neutrophil cytoplasts were evident in the lungs. Surprisingly, allergen-driven airway neutrophilia was decreased in peptidyl arginine deiminase 4-deficient mice with defective NETosis but not by deoxyribonuclease treatment, implicating the cytoplasts for the non-type 2 immune responses to allergen. Neutrophil cytoplasts were also present in mediastinal lymph nodes, and the cytoplasts activated lung dendritic cells in vitro to trigger antigen-specific interleukin-17 (IL-17) production from naïve CD4+ T cells. Bronchoalveolar lavage fluid from patients with severe asthma and high neutrophil counts had detectable NETs and cytoplasts that were positively correlated with IL-17 levels. Together, these translational findings have identified neutrophil cytoplast formation in asthmatic lung inflammation and linked the cytoplasts to T helper 17-mediated neutrophilic inflammation in severe asthma.

9.
Physiol Rev ; 98(3): 1335-1370, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29717929

ABSTRACT

Airborne pathogens and environmental stimuli evoke immune responses in the lung. It is critical to health that these responses be controlled to prevent tissue damage and the compromise of organ function. Resolution of inflammation is a dynamic process that is coordinated by biochemical and cellular mechanisms. Recently, specialized proresolving mediators (SPMs) have been identified in resolution exudates. These molecules orchestrate anti-inflammatory and proresolving actions that are cell type specific. In this review, we highlight SPM biosynthesis, the influence of SPMs on the innate and adaptive immune responses in the lung, as well as recent insights from SPMs on inflammatory disease pathophysiology. Uncovering these mediators and cellular mechanisms for resolution is providing new windows into physiology and disease pathogenesis.


Subject(s)
Fatty Acids, Omega-3/metabolism , Lipoxins/metabolism , Lung Diseases/immunology , Adaptive Immunity , Animals , Humans , Immunity, Innate , Lung Diseases/metabolism
10.
J Immunol ; 200(8): 2757-2766, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29523657

ABSTRACT

Specialized proresolving mediators (SPMs) decrease NF-κB activity to prevent excessive tissue damage and promote the resolution of acute inflammation. Mechanisms for NF-κB regulation by SPMs remain to be determined. In this study, after LPS challenge, the SPMs 15-epi-lipoxin A4 (15-epi-LXA4), resolvin D1, resolvin D2, resolvin D3, and 17-epi-resolvin D1 were produced in vivo in murine lungs. In LPS-activated human bronchial epithelial cells, select SPMs increased expression of the NF-κB regulators A20 and single Ig IL-1R-related molecule (SIGIRR). Of interest, 15-epi-LXA4 induced A20 and SIGIRR in an lipoxin A4 receptor/formyl peptide receptor 2 (ALX/FPR2) receptor-dependent manner in epithelial cells and in murine pneumonia. This SPM regulated NF-κB-induced cytokines to decrease pathogen-mediated inflammation. In addition to dampening lung inflammation, surprisingly, 15-epi-LXA4 also enhanced pathogen clearance with increased antimicrobial peptide expression. Taken together, to our knowledge these results are the first to identify endogenous agonists for A20 and SIGIRR expression to regulate NF-κB activity and to establish mechanisms for NF-κB regulation by SPMs for pneumonia resolution.


Subject(s)
Docosahexaenoic Acids/immunology , Fatty Acids, Unsaturated/immunology , Inflammation Mediators/immunology , Lipoxins/immunology , NF-kappa B/immunology , Pneumonia, Bacterial/immunology , Animals , Cell Line , Docosahexaenoic Acids/metabolism , Fatty Acids, Unsaturated/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation Mediators/metabolism , Lipoxins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Bacterial/metabolism , Receptors, Interleukin-1/agonists , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism
11.
Am J Respir Cell Mol Biol ; 58(3): 341-351, 2018 03.
Article in English | MEDLINE | ID: mdl-28957638

ABSTRACT

Ozone and obesity both increase IL-17A in the lungs. In mice, obesity augments the airway hyperresponsiveness and neutrophil recruitment induced by acute ozone exposure. Therefore, we examined the role of IL-17A in obesity-related increases in the response to ozone observed in obese mice. Lean wild-type and obese db/db mice were pretreated with IL-17A-blocking or isotype antibodies, exposed to air or ozone (2 ppm for 3 h), and evaluated 24 hours later. Microarray analysis of lung tissue gene expression was used to examine the mechanistic basis for effects of anti-IL-17A. Compared with lean mice, ozone-exposed obese mice had greater concentrations of BAL IL-17A and greater numbers of pulmonary IL-17A+ cells. Ozone-induced increases in BAL IL-23 and CCL20, cytokines important for IL-17A+ cell recruitment and activation, were also greater in obese mice. Anti-IL-17A treatment reduced ozone-induced airway hyperresponsiveness toward levels observed in lean mice. Anti-IL-17A treatment also reduced BAL neutrophils in both lean and obese mice, possibly because of reductions in CXCL1. Microarray analysis identified gastrin-releasing peptide (GRP) receptor (Grpr) among those genes that were both elevated in the lungs of obese mice after ozone exposure and reduced after anti-IL-17A treatment. Furthermore, ozone exposure increased BAL GRP to a greater extent in obese than in lean mice, and GRP-neutralizing antibody treatment reduced obesity-related increases in ozone-induced airway hyperresponsiveness and neutrophil recruitment. Our data indicate that IL-17A contributes to augmented responses to ozone in db/db mice. Furthermore, IL-17A appears to act at least in part by inducing expression of Grpr.


Subject(s)
Gastrin-Releasing Peptide/immunology , Interleukin-17/immunology , Obesity/pathology , Ozone/toxicity , Receptors, Bombesin/metabolism , Respiratory Hypersensitivity/immunology , Animals , Antibodies, Blocking/pharmacology , Chemokine CCL20/immunology , Chemokine CXCL1/immunology , Female , Interleukin-23 Subunit p19/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/immunology , Receptors, Bombesin/genetics
12.
Sci Immunol ; 2(9)2017 Mar 10.
Article in English | MEDLINE | ID: mdl-28783702

ABSTRACT

Severe asthma is typically characterized by chronic airway inflammation that is refractory to corticosteroids and associated with excess morbidity. Patients were recruited into the National Heart, Lung, and Blood Institute-sponsored Severe Asthma Research Program and comprehensively phenotyped by bronchoscopy. Bronchoalveolar lavage (BAL) cells were analyzed by flow cytometry. Compared with healthy individuals (n = 21), patients with asthma (n = 53) had fewer BAL natural killer (NK) cells. Patients with severe asthma (n = 29) had a marked increase in the ratios of CD4+ T cells to NK cells and neutrophils to NK cells. BAL NK cells in severe asthma were skewed toward the cytotoxic CD56dim subset, with significantly increased BAL fluid levels of the cytotoxic mediator granzyme A. The numbers of BAL CD56dim NK cells and CCR6-CCR4- T helper 1-enriched CD4+ T cells correlated inversely with lung function [forced expiratory volume in 1 s (FEV1) % predicted] in asthma. Relative to cells from healthy controls, peripheral blood NK cells from asthmatic patients had impaired killing of K562 myeloid target cells despite releasing more cytotoxic mediators. Ex vivo exposure to dexamethasone markedly decreased blood NK cell lysis of target cells and cytotoxic mediator release. NK cells expressed airway lipoxin A4/formyl peptide receptor 2 receptors, and in contrast to dexamethasone, lipoxin A4-exposed NK cells had preserved functional responses. Together, our findings indicate that the immunology of the severe asthma airway is characterized by decreased NK cell cytotoxicity with increased numbers of target leukocytes, which is exacerbated by corticosteroids that further disable NK cell function. These failed resolution mechanisms likely contribute to persistent airway inflammation in severe asthma.

13.
Environ Health Perspect ; 125(2): 246-253, 2017 02.
Article in English | MEDLINE | ID: mdl-27472835

ABSTRACT

BACKGROUND: Ozone increases IL-33 in the lungs, and obesity augments the pulmonary effects of acute ozone exposure. OBJECTIVES: We assessed the role of IL-33 in the augmented effects of ozone observed in obese mice. METHODS: Lean wildtype and obese db/db mice were pretreated with antibodies blocking the IL-33 receptor, ST2, and then exposed to ozone (2 ppm for 3 hr). Airway responsiveness was assessed, bronchoalveolar lavage (BAL) was performed, and lung cells harvested for flow cytometry 24 hr later. Effects of ozone were also assessed in obese and lean mice deficient in γδ T cells and their wildtype controls. RESULTS AND DISCUSSION: Ozone caused greater increases in BAL IL-33, neutrophils, and airway responsiveness in obese than lean mice. Anti-ST2 reduced ozone-induced airway hyperresponsiveness and inflammation in obese mice but had no effect in lean mice. Obesity also augmented ozone-induced increases in BAL CXCL1 and IL-6, and in BAL type 2 cytokines, whereas anti-ST2 treatment reduced these cytokines. In obese mice, ozone increased lung IL-13+ innate lymphoid cells type 2 (ILC2) and IL-13+ γδ T cells. Ozone increased ST2+ γδ T cells, indicating that these cells can be targets of IL-33, and γδ T cell deficiency reduced obesity-related increases in the response to ozone, including increases in type 2 cytokines. CONCLUSIONS: Our data indicate that IL-33 contributes to augmented responses to ozone in obese mice. Obesity and ozone also interacted to promote type 2 cytokine production in γδ T cells and ILC2 in the lungs, which may contribute to the observed effects of IL-33. Citation: Mathews JA, Krishnamoorthy N, Kasahara DI, Cho Y, Wurmbrand AP, Ribeiro L, Smith D, Umetsu D, Levy BD, Shore SA. 2017. IL-33 drives augmented responses to ozone in obese mice. Environ Health Perspect 125:246-253; http://dx.doi.org/10.1289/EHP272.


Subject(s)
Air Pollutants/toxicity , Interleukin-13/metabolism , Ozone/toxicity , Animals , Bronchoalveolar Lavage Fluid , Mice , Toxicity Tests
14.
Am J Respir Cell Mol Biol ; 54(5): 656-63, 2016 05.
Article in English | MEDLINE | ID: mdl-26550921

ABSTRACT

Human precision-cut lung slices (hPCLSs) provide a unique ex vivo model for translational research. However, the limited and unpredictable availability of human lung tissue greatly impedes their use. Here, we demonstrate that cryopreservation of hPCLSs facilitates banking of live human lung tissue for routine use. Our results show that cryopreservation had little effect on overall cell viability and vital functions of immune cells, including phagocytes and T lymphocytes. In addition, airway contraction and relaxation in response to specific agonists and antagonists, respectively, were unchanged after cryopreservation. At the subcellular level, cryopreserved hPCLSs maintained Ca(2+)-dependent regulatory mechanisms for the control of airway smooth muscle cell contractility. To exemplify the use of cryopreserved hPCLSs in smooth muscle research, we provide evidence that bitter-taste receptor (TAS2R) agonists relax airways by blocking Ca(2+) oscillations in airway smooth muscle cells. In conclusion, the banking of cryopreserved hPCLSs provides a robust bioassay for translational research of lung physiology and disease.


Subject(s)
Biological Assay/methods , Bronchodilator Agents/pharmacology , Cryopreservation , Lung/cytology , Receptors, G-Protein-Coupled/agonists , Taste/drug effects , Tissue Banks , Calcium Signaling/drug effects , Cell Survival/drug effects , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phagocytes/drug effects , Phagocytes/metabolism , Receptors, G-Protein-Coupled/metabolism
15.
J Immunol ; 194(3): 863-7, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25539814

ABSTRACT

Asthma is a chronic inflammatory disease that fails to resolve. Recently, a key role for type 2 innate lymphoid cells (ILC2s) was linked to asthma pathogenesis; however, mechanisms for ILC2 regulation remain to be determined. In this study, metabololipidomics of murine lungs identified temporal changes in endogenous maresin 1 (MaR1) during self-limited allergic inflammation. Exogenous MaR1 reduced lung inflammation and ILC2 expression of IL-5 and IL-13 and increased amphiregulin. MaR1 augmented de novo generation of regulatory T cells (Tregs), which interacted with ILC2s to markedly suppress cytokine production in a TGF-ß-dependent manner. Ab-mediated depletion of Tregs interrupted MaR1 control of ILC2 expression of IL-13 in vivo. Together, the findings uncover Tregs as potent regulators of ILC2 activation; MaR1 targets Tregs and ILC2s to restrain allergic lung inflammation, suggesting MaR1 as the basis for a new proresolving therapeutic approach to asthma and other chronic inflammatory diseases.


Subject(s)
Docosahexaenoic Acids/metabolism , Immunity, Innate , Lymphocyte Activation , Pneumonia/immunology , Pneumonia/metabolism , T-Lymphocyte Subsets , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Adoptive Transfer , Animals , Cytokines/biosynthesis , Disease Models, Animal , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immunomodulation , Mice , Mice, Transgenic , Pneumonia/therapy
16.
Proc Natl Acad Sci U S A ; 111(46): 16526-31, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25369934

ABSTRACT

Unregulated acute inflammation can lead to collateral tissue injury in vital organs, such as the lung during the acute respiratory distress syndrome. In response to tissue injury, circulating platelet-neutrophil aggregates form to augment neutrophil tissue entry. These early cellular events in acute inflammation are pivotal to timely resolution by mechanisms that remain to be elucidated. Here, we identified a previously undescribed biosynthetic route during human platelet-neutrophil interactions for the proresolving mediator maresin 1 (MaR1; 7R,14S-dihydroxy-docosa-4Z,8E,10E,12Z,16Z,19Z-hexaenoic acid). Docosahexaenoic acid was converted by platelet 12-lipoxygenase to 13S,14S-epoxy-maresin, which was further transformed by neutrophils to MaR1. In a murine model of acute respiratory distress syndrome, lipid mediator metabololipidomics uncovered MaR1 generation in vivo in a temporally regulated manner. Early MaR1 production was dependent on platelet-neutrophil interactions, and intravascular MaR1 was organ-protective, leading to decreased lung neutrophils, edema, tissue hypoxia, and prophlogistic mediators. Together, these findings identify a transcellular route for intravascular maresin 1 biosynthesis via platelet-neutrophil interactions that regulates the extent of lung inflammation.


Subject(s)
Blood Platelets/metabolism , Docosahexaenoic Acids/biosynthesis , Inflammation/metabolism , Lung/metabolism , Neutrophils/metabolism , Animals , Arachidonate 12-Lipoxygenase/metabolism , Blood Platelets/drug effects , Bronchoalveolar Lavage Fluid/cytology , Cell Hypoxia , Chemotaxis, Leukocyte , Disease Models, Animal , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/physiology , Docosahexaenoic Acids/therapeutic use , Heterografts , Humans , Hydrochloric Acid/toxicity , Inflammation/pathology , Lung/drug effects , Lung/pathology , Male , Metabolomics , Mice , Mice, Inbred BALB C , Molecular Structure , Platelet Activating Factor/pharmacology , Platelet Transfusion , Recombinant Proteins/metabolism , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Thrombin/pharmacology
17.
J Immunol ; 192(12): 5471-5475, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24829419

ABSTRACT

We reported previously that c-kit ligation by membrane-bound stem cell factor (mSCF) boosts IL-6 production in dendritic cells (DCs) and a Th17-immune response. However, Th17 establishment also requires heterodimeric IL-23, but the mechanisms that regulate IL-23 gene expression in DCs are not fully understood. We show that IL-23p19 gene expression in lung DCs is dependent on mSCF, which is regulated by the metalloproteinase MMP-9. Th1-inducing conditions enhanced MMP-9 activity, causing cleavage of mSCF, whereas the opposite was true for Th17-promoting conditions. In MMP-9(-/-) mice, a Th1-inducing condition could maintain mSCF and enhance IL-23p19 in DCs, promoting IL-17-producing CD4(+) T cells in the lung. Conversely, mSCF cleavage from bone marrow DCs in vitro by rMMP-9 led to reduced IL-23p19 expression under Th17-inducing conditions, with dampening of intracellular AKT phosphorylation. Collectively, these results show that the c-kit/mSCF/MMP-9 axis regulates IL-23 gene expression in DCs to control IL-17 production in the lung.


Subject(s)
Dendritic Cells/immunology , Gene Expression Regulation/immunology , Interleukin-17/immunology , Interleukin-23 Subunit p19/immunology , Lung/immunology , Matrix Metalloproteinase 9/immunology , Stem Cell Factor/immunology , Animals , Cell Membrane/genetics , Cell Membrane/immunology , Cell Membrane/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression Regulation/genetics , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-23 Subunit p19/biosynthesis , Interleukin-23 Subunit p19/genetics , Lung/cytology , Lung/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Phosphorylation/genetics , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
18.
Curr Opin Allergy Clin Immunol ; 14(1): 7-12, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24300419

ABSTRACT

PURPOSE OF REVIEW: Binding of the receptor tyrosine kinase, c-kit, to its ligand, stem cell factor (SCF), mediates numerous biological functions. Important roles for c-kit in hematopoiesis, melanogenesis, erythropoiesis, spermatogenesis, and carcinogenesis are well documented. Similarly, activation of mast cells and eosinophils by c-kit ligation has long been known to result in degranulation with concomitant release of pro-inflammatory mediators including cytokines. This review will highlight a recently discovered function of c-kit in regulating the adaptive immune responses with relevance to allergic diseases. RECENT FINDINGS: Recent studies in a number of laboratories including our own highlight the previously unappreciated functions for c-kit in immunological processes. Increased expression of c-kit and its ligand, SCF, on dendritic cells by Th2/Th17-inducing stimuli leads to c-kit activation and immune skewing toward these subsets and away from Th1 responses. Treatment of dendritic cells with inhibitors of c-kit activation such as imatinib mesylate (Gleevec) induces breach of T-cell tolerance, skewing of responses toward Th1, and activation of natural killer cells. SUMMARY: Taken together, these observations suggest that the c-kit/SCF axis may be a useful target for redirecting deleterious immune responses in various disease settings, including allergic diseases that are often associated with Th2 and Th17 responses.


Subject(s)
Dendritic Cells/immunology , Hypersensitivity/metabolism , Killer Cells, Natural/immunology , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/metabolism , Th17 Cells/immunology , Th2 Cells/immunology , Adaptive Immunity , Animals , Benzamides/pharmacology , Cytokines/immunology , Humans , Imatinib Mesylate , Lymphocyte Activation/drug effects , Piperazines/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/immunology , Pyrimidines/pharmacology , Respiratory System/immunology , Signal Transduction , Th1-Th2 Balance/drug effects
19.
J Immunol ; 191(1): 25-9, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23733880

ABSTRACT

Dendritic cell (DC)-T cell interactions that underlie inducible/adaptive regulatory T cell generation and airway tolerance are not well understood. In this study, we show that mice lacking CD11c(hi) lung DCs, but containing plasmacytoid DCs (pDCs), fail tolerization with inhaled Ag and cannot support Foxp3 induction in vivo in naive CD4(+) T cells. CD103(+) DCs from tolerized mice efficiently induced Foxp3 in cocultured naive CD4(+) T cells but pDCs and lung macrophages failed to do so. CD103(+) DCs, but not pDCs or lung macrophages, upregulated the expression of retinaldehyde dehydrogenase 2 (aldh1a2), which is key for the production of retinoic acid, a cofactor for TGF-ß for Foxp3 induction. Batf3(-/-) mice, selectively lacking CD103(+) DCs, failed tolerization by inhaled Ag. Collectively, our data show that pulmonary tolerance is dependent on CD103(+) DCs, correlating with their ability to upregulate aldh1a2, which can promote Foxp3 expression in T cells.


Subject(s)
Aldehyde Dehydrogenase/biosynthesis , Antigens, CD/biosynthesis , CD11c Antigen/physiology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Forkhead Transcription Factors/biosynthesis , Integrin alpha Chains/biosynthesis , Lung/immunology , Up-Regulation/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Dendritic Cells/enzymology , Dendritic Cells/metabolism , Immune Tolerance/immunology , Lung/enzymology , Lung/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
20.
Nat Med ; 18(10): 1525-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22961107

ABSTRACT

Immune tolerance is instituted early in life, during which time regulatory T (T(reg)) cells have an important role. Recurrent infections with respiratory syncytial virus (RSV) in early life increase the risk for asthma in adult life. Repeated infection of infant mice tolerized to ovalbumin (OVA) through their mother's milk with RSV induced allergic airway disease in response to OVA sensitization and challenge, including airway inflammation, hyper-reactivity and higher OVA-specific IgE, as compared to uninfected tolerized control mice. Virus infection induced GATA-3 expression and T helper type 2 (T(H)2) cytokine production in forkhead box P3 (FOXP3)(+) T(reg) cells and compromised the suppressive function of pulmonary T(reg) cells in a manner that was dependent on interleukin-4 receptor α (IL-4Rα) expression in the host. Thus, by promoting a T(H)2-type inflammatory response in the lung, RSV induced a T(H)2-like effector phenotype in T(reg) cells and attenuated tolerance to an unrelated antigen (allergen). Our findings highlight a mechanism by which viral infection targets a host-protective mechanism in early life and increases susceptibility to allergic disease.


Subject(s)
Asthma/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Asthma/virology , Forkhead Transcription Factors/biosynthesis , GATA3 Transcription Factor/biosynthesis , Immune Tolerance , Immunoglobulin E/blood , Interleukin-4 Receptor alpha Subunit/immunology , Interleukin-4 Receptor alpha Subunit/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/immunology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...