Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cells ; 11(1)2022 01 02.
Article in English | MEDLINE | ID: mdl-35011703

ABSTRACT

High eosinophil (EOS) counts are a key feature of eosinophilic asthma. EOS notably affect asthmatic response by generating several lipid mediators. Mice have been utilized in hopes of defining new pharmacological targets to treat asthma. However, many pinpointed targets in mice did not translate into clinics, underscoring that key differences exist between the two species. In this study, we compared the ability of human (h) and mouse (m) EOS to biosynthesize key bioactive lipids derived from arachidonic acid (AA), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). hEOS were isolated from the blood of healthy subjects and mild asthmatics, while mEOSs were differentiated from the bone marrow. EOSs were treated with fatty acids and lipid mediator biosynthesis assessed by LC-MS/MS. We found that hEOS biosynthesized leukotriene (LT) C4 and LTB4 in a 5:1 ratio while mEOS almost exclusively biosynthesized LTB4. The biosynthesis of the 15-lipoxygenase (LO) metabolites 15-HETE and 12-HETE also differed, with a 15-HETE:12-HETE ratio of 6.3 for hEOS and 0.727 for mEOS. EOS biosynthesized some specialized pro-resolving mediators, and the levels from mEOS were 9-times higher than those of hEOS. In contrast, hEOS produced important amounts of the endocannabinoid 2-arachidonoyl-glycerol (2-AG) and its congeners from EPA and DHA, a biosynthetic pathway that was up to ~100-fold less prominent in mEOS. Our data show that hEOS and mEOS biosynthesize the same lipid mediators but in different amounts. Compared to asthmatics, mouse models likely have an amplified involvement of LTB4 and specialized pro-resolving mediators and a diminished impact of the endocannabinoid 2-arachidonoyl-glycerol and its congeners.


Subject(s)
Arachidonic Acids/metabolism , Chromatography, Liquid/methods , Docosahexaenoic Acids/metabolism , Eicosanoids/metabolism , Endocannabinoids/metabolism , Eosinophils/metabolism , Glycerides/metabolism , Tandem Mass Spectrometry/methods , Animals , Humans , Mice
2.
PLoS One ; 16(11): e0260636, 2021.
Article in English | MEDLINE | ID: mdl-34847189

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths. While the recent use of immune checkpoint inhibitors significantly improves patient outcomes, responsiveness remains restricted to a small proportion of patients. Conventional dendritic cells (DCs) play a major role in anticancer immunity. In mice, two subpopulations of DCs are found in the lung: DC2s (CD11b+Sirpα+) and DC1s (CD103+XCR1+), the latest specializing in the promotion of anticancer immune responses. However, the impact of lung cancer on DC populations and the consequent influence on the anticancer immune response remain poorly understood. To address this, DC populations were studied in murine models of Lewis Lung Carcinoma (LLC) and melanoma-induced lung metastasis (B16F10). We report that direct exposure to live or dead cancer cells impacts the capacity of DCs to differentiate into CD103+ DC1s, leading to profound alterations in CD103+ DC1 proportions in the lung. In addition, we observed the accumulation of CD103loCD11b+ DCs, which express DC2 markers IRF4 and Sirpα, high levels of T-cell inhibitory molecules PD-L1/2 and the regulatory molecule CD200. Finally, DC1s were injected in combination with an immune checkpoint inhibitor (anti-PD-1) in the B16F10 model of resistance to the anti-PD-1 immune checkpoint therapy; the co-injection restored sensitivity to immunotherapy. Thus, we demonstrate that lung tumor development leads to the accumulation of CD103loCD11b+ DCs with a regulatory potential combined with a reduced proportion of highly-specialized antitumor CD103+ DC1s, which could promote cancer growth. Additionally, promoting an anticancer DC signature could be an interesting therapeutic avenue to increase the efficacy of existing immune checkpoint inhibitors.


Subject(s)
Carcinoma, Lewis Lung , Dendritic Cells , Immune Checkpoint Inhibitors , Lung Neoplasms , Melanoma, Experimental , Neoplasm Proteins/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Neoplasm Metastasis
3.
Front Cell Infect Microbiol ; 11: 617481, 2021.
Article in English | MEDLINE | ID: mdl-34295830

ABSTRACT

Lung dendritic cells (DCs) are divided into two major populations, which include CD103+XCR1+ cDC1s and CD11b+Sirpα+ cDC2s. The maintenance of their relative proportions is dynamic and lung inflammation, such as caused by exposure to lipopolysaccharide (LPS), a component of the outer membrane of Gram-negative bacteria, can have a significant impact on the local cDC signature. Alterations in the lung cDC signature could modify the capacity of the immune system to respond to various pathogens. We consequently aimed to assess the impact of the Gram-negative bacteria Pseudomonas aeruginosa on lung cDC1 and cDC2 populations, and to identify the mechanisms leading to alterations in cDC populations. We observed that exposure to P. aeruginosa decreased the proportions of CD103+XCR1+ cDC1s, while increasing that of CD11b+ DCs. We identified two potential mechanisms involved in this modulation of lung cDC populations. First, we observed an increase in bone marrow pre-DC IRF4 expression suggesting a higher propensity of pre-DCs to differentiate towards the cDC2 lineage. This observation was combined with a reduced capacity of lung XCR1+ DC1s to express CD103. In vitro, we demonstrated that GM-CSF-induced CD103 expression on cDCs depends on GM-CSF receptor internalization and RUNX1 activity. Furthermore, we observed that cDCs stimulation with LPS or P. aeruginosa reduced the proportions of intracellular GM-CSF receptor and decreased RUNX1 mRNA expression. Altogether, these results suggest that alterations in GM-CSF receptor intracellular localization and RUNX1 signaling could be involved in the reduced CD103 expression on cDC1 in response to P. aeruginosa. To verify whether the capacity of cDCs to express CD103 following P. aeruginosa exposure impacts the immune response, WT and Cd103-/- mice were exposed to P. aeruginosa. Lack of CD103 expression led to an increase in the number of neutrophils in the airways, suggesting that lack of CD103 expression on cDC1s could favor the innate immune response to this bacterium.


Subject(s)
Dendritic Cells , Pseudomonas aeruginosa , Animals , Lipopolysaccharides , Lung , Mice , Mice, Inbred C57BL , Signal Transduction
4.
Am J Respir Cell Mol Biol ; 63(2): 209-218, 2020 08.
Article in English | MEDLINE | ID: mdl-32289229

ABSTRACT

In a proportion of patients with hypersensitivity pneumonitis, the biological and environmental factors that sustain inflammation are ill defined, resulting in no effective treatment option. Bioaerosols found in occupational settings are complex and often include Toll-like receptor ligands, such as endotoxins. How Toll-like receptor ligands contribute to the persistence of hypersensitivity pneumonitis, however, remains poorly understood. In a previous study, we found that an S1P1 (sphingosine-1-phosphate receptor 1) agonist prevented the reactivation of antigen-driven B-cell responses in the lung. Here, we assessed the impact of endotoxins on B-cell activation in preexisting hypersensitivity pneumonitis and the role of S1P1 in this phenomenon. The impact of endotoxins on pre-established hypersensitivity pneumonitis was studied in vivo. S1P1 levels were tracked on B cells in the course of the disease using S1P1-eGFP knockin mice, and the role of S1P1 on B-cell functions was assessed using pharmacological tools. S1P1 was found on B cells in experimental hypersensitivity pneumonitis. Endotoxin exposure enhanced neutrophil accumulation in the BAL of mice with experimental hypersensitivity pneumonitis. This was associated with enhanced CD69 cell-surface expression on lymphocytes in the BAL. In isolated B cells, endotoxins increased cell-surface levels of costimulatory molecules and CD69, which was prevented by an S1P1 agonist. S1P1 modulators also reduced TNF production by B cells and their capacity to trigger T-cell cooperation ex vivo. An S1P1 ligand directly inhibited endotoxin-induced B-cell activation.


Subject(s)
Alveolitis, Extrinsic Allergic/immunology , B-Lymphocytes/immunology , Endotoxins/immunology , Sphingosine-1-Phosphate Receptors/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Female , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , Mice , Neutrophils/immunology
5.
J Muscle Res Cell Motil ; 40(3-4): 309-318, 2019 12.
Article in English | MEDLINE | ID: mdl-31222587

ABSTRACT

Chronic obstructive pulmonary disease (COPD) can sometimes be associated with skeletal muscle atrophy. Hypoxemic episodes, which occur during disease exacerbation and daily physical activity, are frequent in COPD patients. However, the link between hypoxemia and muscle atrophy remains unclear, along with mechanisms of muscle hypoxic stress response. Myogenic progenitors (MPs) and fibro/adipogenic progenitors (FAPs) express CD34 and participate to muscle mass maintenance. Although there is evidence linking CD34 expression and muscle repair, the link between CD34 expression, muscle wasting and the hypoxic stress observed in COPD has never been studied. Using a 2-day model of exposure to hypoxic conditions, we investigated the impact of hypoxia on skeletal muscle wasting and function, and elucidated the importance of CD34 expression in that response. A 2-day exposure to hypoxic conditions induces muscle atrophy, which was significantly worse in Cd34-/- mice compared to wild type (WT). Moreover, the lack of CD34 expression negatively impacts the maximal strength of the extensor digitorum longus muscle in response to hypoxia. Following exposure to hypoxic conditions, FAPs (which support MPs differentiation and myogenesis) are significantly lower in Cd34-/- mice compared to WT animals while the expression of myogenic regulatory factors and degradation factors (Atrogin) are similar. CD34 expression is important in the maintenance of muscle mass and function in response to hypoxic stress. These results highlight a new potential role for CD34 in muscle mass maintenance in hypoxic stress such as observed in COPD.


Subject(s)
Antigens, CD34/metabolism , Muscle, Skeletal/metabolism , Animals , Cell Hypoxia/physiology , Humans , Mice
6.
Eur J Immunol ; 49(4): 638-652, 2019 04.
Article in English | MEDLINE | ID: mdl-30707446

ABSTRACT

Conventional DCs are a heterogeneous population that bridge the innate and adaptive immune systems. The lung DC population comprises CD103+ XCR1+ DC1s and CD11b+ DC2s; their various combined functions cover the whole spectrum of immune responses needed to maintain homeostasis. Here, we report that in vivo exposure to LPS leads to profound alterations in the proportions of CD103+ XCR1+ DCs in the lung. Using ex vivo LPS and TNF stimulations of murine lung and spleen-isolated DCs, we show that this is partly due to a direct downregulation of the GM-CSF-induced DC CD103 expression. Furthermore, we demonstrate that LPS-induced systemic inflammation alters the transcriptional signature of DC precursors toward a lower capacity to differentiate into XCR1+ DCs. Also, we report that TNF prevents the capacity of pre-DCs to express CD103 upon maturation. Overall, our results indicate that exposure to LPS directly impacts the capacity of pre-DCs to differentiate into XCR1+ DCs, in addition to lowering their capacity to express CD103. This leads to decreased proportions of CD103+ XCR1+ DCs in the lung, favoring CD11b+ DCs, which likely plays a role in the break in homeostasis following LPS exposure, and in determining the nature of the immune response to LPS.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Integrin alpha Chains/metabolism , Lipopolysaccharides/immunology , Lung/immunology , Lung/metabolism , Animals , Antigens, CD/genetics , Biomarkers , Bone Marrow/immunology , Bone Marrow/metabolism , Cells, Cultured , Dendritic Cells/drug effects , Gene Expression , Immunophenotyping , Inflammation Mediators/metabolism , Integrin alpha Chains/genetics , Lung/pathology , Mice , Pneumonia/etiology , Pneumonia/metabolism , Pneumonia/pathology , Signal Transduction/drug effects , Tumor Necrosis Factors/pharmacology
7.
PLoS One ; 12(6): e0179678, 2017.
Article in English | MEDLINE | ID: mdl-28628641

ABSTRACT

BACKGROUND: Pulmonary dendritic cells drive lung responses to foreign antigens, including Saccharopolyspora rectivirgula, a causative agent of hypersensitivity pneumonitis. While the airway inflammatory mechanisms involved in hypersensitivity pneumonitis are well described, the mechanisms leading to the break in homeostasis and hypersensitivity pneumonitis onset are not well-described, and could involve CD103+ dendritic cells, which are found at baseline and during inflammatory responses in the lung. However, recent demonstration of the ability of CD103+ dendritic cells to induce inflammatory responses starkly contrasts with their classically described role as regulatory cells. These discrepancies may be attributable to the lack of current information on the importance of CD103 expression and modulation on these cells during inflammatory episodes. METHODS: To verify the importance of CD103 expression in the regulation of hypersensitivity pneumonitis, wild-type and Cd103-/- mice were exposed intranasally to S. rectivirgula and airway inflammation was quantified. Surface expression of CD103 in response to S. rectivirgula exposure was studied and cell transfers were used to determine the relative importance of CD103 expression on dendritic cells and T cells in regulating the inflammation in hypersensitivity pneumonitis. RESULTS: Cd103-/- mice developed an exacerbated inflammatory response as early as 18h following S. rectivirgula exposure. CD103 expression on dendritic cells was downregulated quickly following S. rectivirgula exposure, and cell transfers demonstrated that CD103 expression on dendritic cells specifically (and not T cells) regulates the onset and severity of this response. CONCLUSION: All in all, we demonstrate that CD103 expression by dendritic cells, but not T cells, is crucial for homeostasis maintenance and the regulation of the TH17 airway inflammatory response in hypersensitivity pneumonitis.


Subject(s)
Alveolitis, Extrinsic Allergic/pathology , Antigens, CD/metabolism , Dendritic Cells/metabolism , Integrin alpha Chains/metabolism , Alveolitis, Extrinsic Allergic/immunology , Alveolitis, Extrinsic Allergic/microbiology , Animals , Antigens, Bacterial/immunology , Antigens, CD/genetics , Bronchoalveolar Lavage Fluid/cytology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/immunology , Disease Models, Animal , Down-Regulation , Immunoglobulin G/blood , Immunoglobulin G/immunology , Integrin alpha Chains/deficiency , Integrin alpha Chains/genetics , Leukocytes/cytology , Lung/cytology , Lung/immunology , Lung/pathology , Mice , Mice, Knockout , Saccharopolyspora/metabolism , Saccharopolyspora/pathogenicity , Severity of Illness Index , Spleen/cytology , Spleen/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
8.
Physiol Rep ; 5(7)2017 Apr.
Article in English | MEDLINE | ID: mdl-28364028

ABSTRACT

Despite improved awareness of work-related diseases and preventive measures, many workers are still at high risk of developing occupational hypersensitivity airway diseases. This stems from a lack of knowledge of bioaerosol composition and their potential effects on human health. Recently, archaea species were identified in bioaerosols, raising the possibility that they play a major role in exposure-related pathology. Specifically, Methanosphaera stadtmanae (MSS) and Methanobrevibacter smithii (MBS) are found in high concentrations in agricultural environments and respiratory exposure to crude extract demonstrates immunomodulatory activity in mice. Nevertheless, our knowledge of the specific impact of methanogens exposure on airway immunity and their potential to induce airway hypersensitivity responses in workers remains scant. Analysis of the lung mucosal response to methanogen crude extracts in mice demonstrated that MSS and MBS predominantly induced TH17 airway inflammation, typical of a type IV hypersensitivity response. Furthermore, the response to MSS was associated with antigen-specific IgG1 and IgG2a production. However, despite the presence of eosinophils after MSS exposure, only a weak TH2 response and no airway hyperresponsiveness were observed. Finally, using eosinophil and mast cell-deficient mice, we confirmed that these cells are dispensable for the TH17 response to MSS, although eosinophils likely contribute to the exacerbation of inflammatory processes induced by MSS crude extract exposure. We conclude that, as MSS induces a clear type IV hypersensitivity lung response, it has the potential to be harmful to workers frequently exposed to this methanogen, and that preventive measures should be taken to avoid chronic hypersensitivity disease development in workers.


Subject(s)
Hypersensitivity/microbiology , Inflammation/microbiology , Lung/microbiology , Methanobacteriaceae , Respiratory Hypersensitivity/microbiology , Animals , Disease Models, Animal , Immunoglobulin G/analysis , Mice
9.
Respir Res ; 16: 7, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25645346

ABSTRACT

BACKGROUND: In vivo phosphorylation of sphingosine analogs with their ensuing binding and activation of their cell-surface sphingosine-1-phosphate receptors is regarded as the main immunomodulatory mechanism of this new class of drugs. Prophylactic treatment with sphingosine analogs interferes with experimental asthma by impeding the migration of dendritic cells to draining lymph nodes. However, whether these drugs can also alleviate allergic airway inflammation after its onset remains to be determined. Herein, we investigated to which extent and by which mechanisms the sphingosine analog AAL-R interferes with key features of asthma in a murine model during ongoing allergic inflammation induced by Dermatophagoides pteronyssinus. METHODS: BALB/c mice were exposed to either D. pteronyssinus or saline, intranasally, once-daily for 10 consecutive days. Mice were treated intratracheally with either AAL-R, its pre-phosphorylated form AFD-R, or the vehicle before every allergen challenge over the last four days, i.e. after the onset of allergic airway inflammation. On day 11, airway responsiveness to methacholine was measured; inflammatory cells and cytokines were quantified in the airways; and the numbers and/or viability of T cells, B cells and dendritic cells were assessed in the lungs and draining lymph nodes. RESULTS: AAL-R decreased airway hyperresponsiveness induced by D. pteronyssinus by nearly 70%. This was associated with a strong reduction of IL-5 and IL-13 levels in the airways and with a decreased eosinophilic response. Notably, the lung CD4(+) T cells were almost entirely eliminated by AAL-R, which concurred with enhanced apoptosis/necrosis in that cell population. This inhibition occurred in the absence of dendritic cell number modulation in draining lymph nodes. On the other hand, the pre-phosphorylated form AFD-R, which preferentially acts on cell-surface sphingosine-1-phosphate receptors, was relatively impotent at enhancing cell death, which led to a less efficient control of T cell and eosinophil responses in the lungs. CONCLUSION: Airway delivery of the non-phosphorylated sphingosine analog, but not its pre-phosphorylated counterpart, is highly efficient at controlling the local T cell response after the onset of allergic airway inflammation. The mechanism appears to involve local induction of lymphocyte apoptosis/necrosis, while mildly affecting dendritic cell and T cell accumulation in draining lymph nodes.


Subject(s)
Anti-Allergic Agents/pharmacology , Asthma/prevention & control , Bronchial Hyperreactivity/prevention & control , Dermatophagoides pteronyssinus , Lung/drug effects , Pneumonia/prevention & control , Sphingosine/pharmacology , Animals , Apoptosis/drug effects , Asthma/immunology , Asthma/metabolism , Asthma/physiopathology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/physiopathology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Interleukin-13/metabolism , Interleukin-5/metabolism , Lung/immunology , Lung/metabolism , Lung/physiopathology , Mice, Inbred C57BL , Necrosis , Phosphorylation , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/physiopathology , Sphingosine/analogs & derivatives , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors
10.
Am J Physiol Lung Cell Mol Physiol ; 308(8): L816-26, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25681437

ABSTRACT

Although CD103(+) cells recently emerged as key regulatory cells in the gut, the role of CD103 ubiquitous expression in the lung and development of allergic airway disease has never been studied. To answer this important question, we evaluated the response of Cd103(-/-) mice in two separate well-described mouse models of asthma (ovalbumin and house dust mite extract). Pulmonary inflammation was assessed by analysis of bronchoalveolar lavage content, histology, and cytokine response. CD103 expression was analyzed on lung dendritic cells and T cell subsets by flow cytometry. Cd103(-/-) mice exposed to antigens developed exacerbated lung inflammation, characterized by increased eosinophilic infiltration, severe tissue inflammation, and altered cytokine response. In wild-type mice exposed to house dust mite, CD103(+) dendritic cells are increased in the lung and an important subset of CD4(+) T cells, CD8(+) T cells, and T regulatory cells express CD103. Importantly, Cd103(-/-) mice presented a deficiency in the resolution phase of inflammation, which supports an important role for this molecule in the control of inflammation severity. These results suggest an important role for CD103 in the control of airway inflammation in asthma.


Subject(s)
Antigens, CD/metabolism , Asthma/metabolism , Integrin alpha Chains/metabolism , Lung/metabolism , Animals , Antigens, CD/genetics , Asthma/immunology , Cytokines/biosynthesis , Dendritic Cells/immunology , Gene Expression , Inflammation/metabolism , Integrin alpha Chains/genetics , Lung/immunology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...