Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 35
Filtrer
1.
Neurosci Biobehav Rev ; 162: 105724, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38762130

RÉSUMÉ

Alzheimer's disease (AD) is prevalent around the world, yet our understanding of the disease is still very limited. Recent work suggests that the cornerstone of AD may include the inflammation that accompanies it. Failure of a normal pro-inflammatory immune response to resolve may lead to persistent central inflammation that contributes to unsuccessful clearance of amyloid-beta plaques as they form, neuronal death, and ultimately cognitive decline. Individual metabolic, and dietary (lipid) profiles can differentially regulate this inflammatory process with aging, obesity, poor diet, early life stress and other inflammatory factors contributing to a greater risk of developing AD. Here, we integrate evidence for the interface between these factors, and how they contribute to a pro-inflammatory brain milieu. In particular, we discuss the importance of appropriate polyunsaturated fatty acids (PUFA) in the diet for the metabolism of specialised pro-resolving mediators (SPMs); raising the possibility for dietary strategies to improve AD outlook.


Sujet(s)
Vieillissement , Maladie d'Alzheimer , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/physiopathologie , Humains , Vieillissement/physiologie , Vieillissement/métabolisme , Animaux , Maladies neuro-inflammatoires/immunologie , Maladies neuro-inflammatoires/métabolisme , Inflammation/métabolisme , Encéphale/métabolisme , Encéphale/physiopathologie
2.
Respir Res ; 25(1): 161, 2024 Apr 13.
Article de Anglais | MEDLINE | ID: mdl-38614991

RÉSUMÉ

BACKGROUND: Longitudinal studies have identified childhood asthma as a risk factor for obstructive pulmonary disease (COPD) and asthma-COPD overlap (ACO) where persistent airflow limitation can develop more aggressively. However, a causal link between childhood asthma and COPD/ACO remains to be established. Our study aimed to model the natural history of childhood asthma and COPD and to investigate the cellular/molecular mechanisms that drive disease progression. METHODS: Allergic airways disease was established in three-week-old young C57BL/6 mice using house dust mite (HDM) extract. Mice were subsequently exposed to cigarette smoke (CS) and HDM for 8 weeks. Airspace enlargement (emphysema) was measured by the mean linear intercept method. Flow cytometry was utilised to phenotype lung immune cells. Bulk RNA-sequencing was performed on lung tissue. Volatile organic compounds (VOCs) in bronchoalveolar lavage-fluid were analysed to screen for disease-specific biomarkers. RESULTS: Chronic CS exposure induced emphysema that was significantly augmented by HDM challenge. Increased emphysematous changes were associated with more abundant immune cell lung infiltration consisting of neutrophils, interstitial macrophages, eosinophils and lymphocytes. Transcriptomic analyses identified a gene signature where disease-specific changes induced by HDM or CS alone were conserved in the HDM-CS group, and further revealed an enrichment of Mmp12, Il33 and Il13, and gene expression consistent with greater expansion of alternatively activated macrophages. VOC analysis also identified four compounds increased by CS exposure that were paradoxically reduced in the HDM-CS group. CONCLUSIONS: Early-life allergic airways disease worsened emphysematous lung pathology in CS-exposed mice and markedly alters the lung transcriptome.


Sujet(s)
Asthme , Fumer des cigarettes , Emphysème , Hypersensibilité , Emphysème pulmonaire , Humains , Animaux , Souris , Souris de lignée C57BL , Pyroglyphidae , Fumer des cigarettes/effets indésirables , Emphysème pulmonaire/étiologie , Inflammation
3.
Am J Respir Cell Mol Biol ; 69(1): 99-112, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37014138

RÉSUMÉ

The epidemiological patterns of incident chronic obstructive pulmonary disease (COPD) and lung adenocarcinoma are changing, with an increasing fraction of disease occurring in patients who are never-smokers or were not exposed to traditional risk factors. However, causative mechanism(s) are obscure. Overactivity of Src family kinases (SFKs) and myeloid cell-dependent inflammatory lung epithelial and endothelial damage are independent candidate mechanisms, but their pathogenic convergence has not been demonstrated. Here we present a novel preclinical model in which an activating mutation in Lyn, a nonreceptor SFK that is expressed in immune cells, epithelium, and endothelium-all strongly implicated in the pathogenesis of COPD-causes spontaneous inflammation, early-onset progressive emphysema, and lung adenocarcinoma. Surprisingly, even though activated macrophages, elastolytic enzymes, and proinflammatory cytokines were prominent, bone marrow chimeras formally demonstrated that myeloid cells were not disease initiators. Rather, lung disease arose from aberrant epithelial cell proliferation and differentiation, microvascular lesions within an activated endothelial microcirculation, and amplified EGFR (epidermal growth factor receptor) expression. In human bioinformatics analyses, LYN expression was increased in patients with COPD and was correlated with increased EGFR expression, a known lung oncogenic pathway, and LYN was linked to COPD. Our study shows that a singular molecular defect causes a spontaneous COPD-like immunopathology and lung adenocarcinoma. Furthermore, we identify Lyn and, by implication, its associated signaling pathways as new therapeutic targets for COPD and cancer. Moreover, our work may inform the development of molecular risk screening and intervention methods for disease susceptibility, progression, and prevention of these increasingly prevalent conditions.


Sujet(s)
Adénocarcinome pulmonaire , Emphysème , Tumeurs du poumon , Broncho-pneumopathie chronique obstructive , Emphysème pulmonaire , Humains , Adénocarcinome pulmonaire/génétique , Récepteurs ErbB/métabolisme , Tumeurs du poumon/génétique , Broncho-pneumopathie chronique obstructive/métabolisme , Emphysème pulmonaire/génétique , src-Family kinases/métabolisme
4.
Front Pharmacol ; 13: 835848, 2022.
Article de Anglais | MEDLINE | ID: mdl-35273509

RÉSUMÉ

Asthmatics are highly susceptible to developing lower respiratory tract infections caused by Streptococcus pneumoniae (SPN, the pneumococcus). It has recently emerged that underlying allergic airway disease creates a lung microenvironment that is defective in controlling pneumococcal lung infections. In the present study, we examined how house dust mite (HDM) aeroallergen exposure altered immunity to acute pneumococcal lung infection. Alveolar macrophage (AM) isolated from HDM-exposed mice expressed alternatively activated macrophage (AAM) markers including YM1, FIZZ1, IL-10, and ARG-1. In vivo, prior HDM exposure resulted in accumulation of AAMs in the lungs and 2-log higher bacterial titres in the bronchoalveolar (BAL) fluid of SPN-infected mice (Day 2). Acute pneumococcal infection further increased the expression of IL-10 and ARG1 in the lungs of HDM-exposed mice. Moreover, prior HDM exposure attenuated neutrophil extracellular traps (NETs) formation in the lungs and dsDNA levels in the BAL fluid of SPN-infected mice. In addition, HDM-SPN infected animals had significantly increased BAL fluid cellularity driven by an influx of macrophages/monocytes, neutrophils, and eosinophils. Increased lung inflammation and mucus production was also evident in HDM-sensitised mice following acute pneumococcal infection, which was associated with exacerbated airway hyperresponsiveness. Of note, PCV13 vaccination modestly reduced pneumococcal titres in the BAL fluid of HDM-exposed animals and did not prevent BAL inflammation. Our findings provide new insights on the relationship between pneumococcal lung infections and allergic airways disease, where defective AM phagocytosis and NETosis are implicated in increased susceptibility to pneumococcal infection.

5.
Respirology ; 27(6): 427-436, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35176813

RÉSUMÉ

BACKGROUND AND OBJECTIVE: Inhalation of high concentrations of respirable crystalline silica (RCS) can lead to silicosis. RCS contains varying levels of iron, which can cause oxidative stress and stimulate ferritin production. This study evaluated iron-related and inflammatory markers in control and silicosis patients. METHODS: A cohort of stone benchtop industry workers (n = 18) were radiologically classified by disease severity into simple or complicated silicosis. Peripheral blood and bronchoalveolar lavage (BAL) were collected to measure iron, ferritin, C-reactive protein, serum amyloid A and serum silicon levels. Ferritin subunit expression in BAL and transbronchial biopsies was analysed by reverse transcription quantitative PCR. Lipid accumulation in BAL macrophages was assessed by Oil Red O staining. RESULTS: Serum iron levels were significantly elevated in patients with silicosis, with a strong positive association with serum ferritin levels. In contrast, markers of systemic inflammation were not increased in silicosis patients. Serum silicon levels were significantly elevated in complicated disease. BAL macrophages from silicosis patients were morphologically consistent with lipid-laden foamy macrophages. Ferritin light chain (FTL) mRNA expression in BAL macrophages was also significantly elevated in simple silicosis patients and correlated with systemic ferritin. CONCLUSION: Our findings suggest that elevated iron levels during the early phases of silicosis increase FTL expression in BAL macrophages, which drives elevated BAL and serum ferritin levels. Excess iron and ferritin were also associated with the emergence of a foamy BAL macrophage phenotype. Ferritin may represent an early disease marker for silicosis, where increased levels are independent of inflammation and may contribute to fibrotic lung remodelling.


Sujet(s)
Ferritines , Silicose , Marqueurs biologiques/métabolisme , Liquide de lavage bronchoalvéolaire/composition chimique , Ferritines/analyse , Ferritines/métabolisme , Humains , Inflammation/métabolisme , Fer/analyse , Fer/métabolisme , Lipides , Poumon/anatomopathologie , Macrophages/métabolisme , Silice
6.
JCI Insight ; 6(11)2021 06 08.
Article de Anglais | MEDLINE | ID: mdl-33945505

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant repair that diminishes lung function via mechanisms that remain poorly understood. CC chemokine receptor (CCR10) and its ligand CCL28 were both elevated in IPF compared with normal donors. CCR10 was highly expressed by various cells from IPF lungs, most notably stage-specific embryonic antigen-4-positive mesenchymal progenitor cells (MPCs). In vitro, CCL28 promoted the proliferation of CCR10+ MPCs while CRISPR/Cas9-mediated targeting of CCR10 resulted in the death of MPCs. Following the intravenous injection of various cells from IPF lungs into immunodeficient (NOD/SCID-γ, NSG) mice, human CCR10+ cells initiated and maintained fibrosis in NSG mice. Eph receptor A3 (EphA3) was among the highest expressed receptor tyrosine kinases detected on IPF CCR10+ cells. Ifabotuzumab-targeted killing of EphA3+ cells significantly reduced the numbers of CCR10+ cells and ameliorated pulmonary fibrosis in humanized NSG mice. Thus, human CCR10+ cells promote pulmonary fibrosis, and EphA3 mAb-directed elimination of these cells inhibits lung fibrosis.


Sujet(s)
Fibrose pulmonaire idiopathique/métabolisme , Cellules souches mésenchymateuses/métabolisme , Récepteur EphA3/métabolisme , Récepteurs CCR10/métabolisme , Pneumocytes/métabolisme , Animaux , Anticorps monoclonaux/pharmacologie , Systèmes CRISPR-Cas , Chimiokines CC/métabolisme , Fibroblastes/métabolisme , Techniques de knock-out de gènes , Humains , Fibrose pulmonaire idiopathique/anatomopathologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Souris , Souris de lignée NOD , Souris SCID
7.
Br J Pharmacol ; 178(8): 1869-1885, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33609280

RÉSUMÉ

BACKGROUND AND PURPOSE: Asthma is a chronic disease that displays heterogeneous clinical and molecular features. A phenotypic subset of late-onset severe asthmatics has debilitating fixed airflow obstruction, increased neutrophilic inflammation and a history of pneumonia. Influenza A virus (IAV) is an important viral cause of pneumonia and asthmatics are frequently hospitalised during IAV epidemics. This study aims to determine whether antagonising granulocyte colony stimulating factor receptor (G-CSFR) prevents pneumonia-associated severe asthma. EXPERIMENTAL APPROACH: Mice were sensitised to house dust mite (HDM) to establish allergic airway inflammation and subsequently infected with IAV (HKx31/H3N2 subtype). A neutralising monoclonal antibody against G-CSFR was therapeutically administered. KEY RESULTS: In IAV-infected mice with prior HDM sensitisation, a significant increase in airway fibrotic remodelling and airways hyper-reactivity was observed. A mixed granulocytic inflammatory profile consisting of neutrophils, macrophages and eosinophils was prominent and at a molecular level, G-CSF expression was significantly increased in HDMIAV-treated mice. Blockage of G-CSFR reduced neutrophilic inflammation in the bronchoalveolar and lungs by over 80% in HDMIAV-treated mice without altering viral clearance. Markers of NETosis (dsDNA and myeloperoxidase in bronchoalveolar), tissue injury (LDH activity in bronchoalveolar) and oedema (total bronchoalveolar-fluid protein) were also significantly reduced with anti-G-CSFR treatment. In addition, anti-G-CSFR antagonism significantly reduced bronchoalveolar gelatinase activity, active TFGß lung levels, collagen lung expression, airways fibrosis and airways hyper-reactivity in HDMIAV-treated mice. CONCLUSIONS AND IMPLICATIONS: We have shown that antagonising G-CSFR-dependent neutrophilic inflammation reduced pathological disruption of the mucosal barrier and airways fibrosis in an IAV-induced severe asthma model.


Sujet(s)
Asthme , Récepteur de facteur de croissance granulocytaire , Animaux , Asthme/traitement médicamenteux , Asthme/anatomopathologie , Asthme/virologie , Liquide de lavage bronchoalvéolaire , Modèles animaux de maladie humaine , Fibrose , Sous-type H3N2 du virus de la grippe A , Poumon/anatomopathologie , Souris , Pyroglyphidae , Récepteur de facteur de croissance granulocytaire/antagonistes et inhibiteurs
8.
Am J Respir Crit Care Med ; 202(11): 1540-1550, 2020 12 01.
Article de Anglais | MEDLINE | ID: mdl-32692579

RÉSUMÉ

Rationale: Declining lung function in patients with interstitial lung disease is accompanied by epithelial remodeling and progressive scarring of the gas-exchange region. There is a need to better understand the contribution of basal cell hyperplasia and associated mucosecretory dysfunction to the development of idiopathic pulmonary fibrosis (IPF).Objectives: We sought to decipher the transcriptome of freshly isolated epithelial cells from normal and IPF lungs to discern disease-dependent changes within basal stem cells.Methods: Single-cell RNA sequencing was used to map epithelial cell types of the normal and IPF human airways. Organoid and air-liquid interface cultures were used to investigate functional properties of basal cell subtypes.Measurements and Main Results: We found that basal cells included multipotent and secretory primed subsets in control adult lung tissue. Secretory primed basal cells include an overlapping molecular signature with basal cells obtained from the distal lung tissue of IPF lungs. We confirmed that NOTCH2 maintains undifferentiated basal cells and restricts basal-to-ciliated differentiation, and we present evidence that NOTCH3 functions to restrain secretory differentiation.Conclusions: Basal cells are dynamically regulated in disease and are specifically biased toward the expansion of the secretory primed basal cell subset in IPF. Modulation of basal cell plasticity may represent a relevant target for therapeutic intervention in IPF.


Sujet(s)
Plasticité cellulaire , Prolifération cellulaire/génétique , Auto-renouvellement cellulaire/génétique , Cellules épithéliales/cytologie , Fibrose pulmonaire idiopathique/génétique , Muqueuse respiratoire/cytologie , Sujet âgé , Pneumocytes/cytologie , Pneumocytes/métabolisme , Membrane basale , Études cas-témoins , Cellules épithéliales/métabolisme , Femelle , Analyse de profil d'expression de gènes , Humains , Fibrose pulmonaire idiopathique/métabolisme , Mâle , Adulte d'âge moyen , RNA-Seq , Muqueuse respiratoire/métabolisme , Analyse sur cellule unique , Transcriptome , Jeune adulte
9.
Antioxid Redox Signal ; 32(13): 929-942, 2020 05 01.
Article de Anglais | MEDLINE | ID: mdl-31190565

RÉSUMÉ

Aims: Reactive oxygen species (ROS) are highly reactive molecules generated in different subcellular sites or compartments, including endosomes via the NOX2-containing nicotinamide adenine dinucleotide phosphate oxidase during an immune response and in mitochondria during cellular respiration. However, while endosomal NOX2 oxidase promotes innate inflammation to influenza A virus (IAV) infection, the role of mitochondrial ROS (mtROS) has not been comprehensively investigated in the context of viral infections in vivo. Results: In this study, we show that pharmacological inhibition of mtROS, with intranasal delivery of MitoTEMPO, resulted in a reduction in airway/lung inflammation, neutrophil infiltration, viral titers, as well as overall morbidity and mortality in mice infected with IAV (Hkx31, H3N2). MitoTEMPO treatment also attenuated apoptotic and necrotic neutrophils and macrophages in airway and lung tissue. At an early phase of influenza infection, that is, day 3 there were significantly lower amounts of IL-1ß protein in the airways, but substantially higher amounts of type I IFN-ß following MitoTEMPO treatment. Importantly, blocking mtROS did not appear to alter the initiation of an adaptive immune response by lung dendritic cells, nor did it affect lung B and T cell populations that participate in humoral and cellular immunity. Innovation/Conclusion: Influenza virus infection promotes mtROS production, which drives innate immune inflammation and this exacerbates viral pathogenesis. This pathogenic cascade highlights the therapeutic potential of local mtROS antioxidant delivery to alleviate influenza virus pathology.


Sujet(s)
Inflammation/immunologie , Mitochondries/immunologie , Infections à Orthomyxoviridae/immunologie , Espèces réactives de l'oxygène/immunologie , Animaux , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Virus de la grippe A/effets des médicaments et des substances chimiques , Virus de la grippe A/immunologie , Mâle , Souris , Souris de lignée C57BL , Mitochondries/effets des médicaments et des substances chimiques , Composés organiques du phosphore/administration et posologie , Composés organiques du phosphore/pharmacologie , Infections à Orthomyxoviridae/traitement médicamenteux , Infections à Orthomyxoviridae/anatomopathologie , Pipéridines/administration et posologie , Pipéridines/pharmacologie , Espèces réactives de l'oxygène/antagonistes et inhibiteurs
10.
Expert Opin Biol Ther ; 19(6): 539-546, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-30900913

RÉSUMÉ

INTRODUCTION: Lifelong maintenance of a healthy lung requires resident stem cells to proliferate according to tissue requirements. Once thought to be a quiescent tissue, evolving views of the complex differentiation landscape of lung stem and progenitor cells have broad implications for our understanding of how the lung is maintained, as well as the development of new therapies for promoting endogenous regeneration in lung disease. AREAS COVERED: This review collates a large body of research relating to the hierarchical organization of epithelial stem cells in the adult lung and their role in tissue homeostasis and regeneration after injury. To identify relevant studies, PubMed was queried using one or a combination of the terms 'lung', 'airway', 'alveoli', 'stem cells', 'progenitor', 'repair' and 'regeneration'. EXPERT OPINION: This review discusses how new technologies and injury models have challenged the demarcations between stem and progenitor cell populations.


Sujet(s)
Maladies pulmonaires/thérapie , Poumon/physiologie , Régénération , Animaux , Différenciation cellulaire , Humains , Alvéoles pulmonaires/cytologie , Transplantation de cellules souches , Cellules souches/cytologie , Cellules souches/métabolisme
11.
Methods Mol Biol ; 1842: 167-181, 2018.
Article de Anglais | MEDLINE | ID: mdl-30196408

RÉSUMÉ

Identification, isolation, and clonal culture of stem cells is essential for understanding their proliferative and differentiation potential, and the cellular and molecular mechanisms that regulate their fate. Akin to development in vivo, the in vitro growth of adult lung epithelial stem cells requires support of mesenchymal-derived growth factors. In the adult mouse lung, epithelial stem/progenitor cells are defined by the phenotype CD45neg CD31neg EpCAMpos CD104pos CD24low, and mesenchymal cells are defined by the phenotype CD45neg CD31neg EpCAMneg Sca-1hi. Here we describe a method for primary cell isolation from the adult mouse lung, a flow cytometry strategy for fractionation of epithelial stem/progenitor cells and mesenchymal cells, and a three-dimensional epithelial colony-forming assay.


Sujet(s)
Cellules souches adultes/cytologie , Cellules souches adultes/métabolisme , Poumon/cytologie , Animaux , Marqueurs biologiques , Techniques de culture cellulaire , Test clonogénique , Cellules épithéliales/cytologie , Cellules épithéliales/métabolisme , Humains , Immunophénotypage , Souris , Phénotype , Muqueuse respiratoire/cytologie
12.
Development ; 144(14): 2618-2628, 2017 07 15.
Article de Anglais | MEDLINE | ID: mdl-28619823

RÉSUMÉ

Mechanisms that regulate tissue-specific progenitors for maintenance and differentiation during development are poorly understood. Here, we demonstrate that the co-repressor protein Sin3a is crucial for lung endoderm development. Loss of Sin3a in mouse early foregut endoderm led to a specific and profound defect in lung development with lung buds failing to undergo branching morphogenesis and progressive atrophy of the proximal lung endoderm with complete epithelial loss at later stages of development. Consequently, neonatal pups died at birth due to respiratory insufficiency. Further analysis revealed that loss of Sin3a resulted in embryonic lung epithelial progenitor cells adopting a senescence-like state with permanent cell cycle arrest in G1 phase. This was mediated at least partially through upregulation of the cell cycle inhibitors Cdkn1a and Cdkn2c. At the same time, loss of endodermal Sin3a also disrupted cell differentiation of the mesoderm, suggesting aberrant epithelial-mesenchymal signaling. Together, these findings reveal that Sin3a is an essential regulator for early lung endoderm specification and differentiation.


Sujet(s)
Cellules souches embryonnaires/cytologie , Cellules souches embryonnaires/métabolisme , Poumon/embryologie , Poumon/métabolisme , Protéines de répression/métabolisme , Animaux , Animaux nouveau-nés , Points de contrôle du cycle cellulaire , Différenciation cellulaire , Lignage cellulaire/génétique , Lignage cellulaire/physiologie , Inhibiteur p18 de kinase cycline-dépendante/génétique , Inhibiteur p18 de kinase cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/génétique , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Endoderme/cytologie , Endoderme/embryologie , Endoderme/métabolisme , Cellules épithéliales/cytologie , Cellules épithéliales/métabolisme , Femelle , Régulation de l'expression des gènes au cours du développement , Poumon/cytologie , Souris , Souris knockout , Organogenèse/génétique , Organogenèse/physiologie , Grossesse , Protéines de répression/déficit , Protéines de répression/génétique , Transduction du signal , Complexe Sin3-histone désacétylases-corépresseurs
13.
Stem Cell Reports ; 7(5): 817-825, 2016 11 08.
Article de Anglais | MEDLINE | ID: mdl-27773701

RÉSUMÉ

Recent studies have implicated keratin 5 (KRT5)+ cells in repopulation of damaged lung tissue following severe H1N1 influenza virus infection. However, the origins of the cells repopulating the injured alveolar region remain controversial. We sought to determine the cellular dynamics of lung repair following influenza infection and define whether nascent KRT5+ cells repopulating alveolar epithelium were derived from pre-existing alveolar or airway progenitor cells. We found that the wound-healing response begins with proliferation of SOX2+ SCGB1A1- KRT5- progenitor cells in airways. These cells generate nascent KRT5+ cells as an early response to airway injury and yield progeny that colonize damaged alveolar parenchyma. Moreover, we show that local alveolar progenitors do not contribute to nascent KRT5+ cells after injury. Repopulation of injured airway and alveolar regions leads to proximalization of distal airways by pseudostratified epithelium and of alveoli by airway-derived epithelial cells that lack the normal characteristics of mature airway or alveolar epithelium.


Sujet(s)
Pneumocytes/métabolisme , Différenciation cellulaire , Kératine-5/métabolisme , Muqueuse respiratoire/cytologie , Muqueuse respiratoire/métabolisme , Facteurs de transcription SOX-B1/métabolisme , Cellules souches/cytologie , Cellules souches/métabolisme , Pneumocytes/cytologie , Pneumocytes/virologie , Animaux , Marqueurs biologiques , Lignage cellulaire , Auto-renouvellement cellulaire/génétique , Sous-type H1N1 du virus de la grippe A , Souris , Souris transgéniques , Modèles biologiques , Infections à Orthomyxoviridae/génétique , Infections à Orthomyxoviridae/métabolisme , Infections à Orthomyxoviridae/anatomopathologie , Infections à Orthomyxoviridae/virologie , Muqueuse respiratoire/virologie , Facteurs de transcription SOX-B1/génétique
14.
Sci Rep ; 6: 22751, 2016 Mar 04.
Article de Anglais | MEDLINE | ID: mdl-26940954

RÉSUMÉ

There is emerging epidemiological data to suggest that upper respiratory tract bacterial colonisation in infancy may increase the risk of developing respiratory dysfunction later in life, and respiratory viruses are known to precipitate persistent colonisation. This study utilized a neonatal mouse model of Streptococcus pneumonia (SP) and influenza A virus (IAV) co-infection, where bronchoalveolar leukocyte infiltration had resolved by adulthood. Only co-infection resulted in persistent nasopharyngeal colonisation over 40 days and a significant increase in airway resistance in response to in vivo methacholine challenge. A significant increase in hysteresivity was also observed in IAV and co-infected mice, consistent with ventilatory heterogeneity and structural changes in the adult lung. Airway hyper-responsiveness was not associated with a detectable increase in goblet cell transdifferentiation, peribronchial smooth muscle bulk or collagen deposition in regions surrounding the airways. Increased reactivity was not observed in precision cut lung slices challenged with methacholine in vitro. Histologically, the airway epithelium appeared normal and expression of epithelial integrity markers (ZO-1, occludin-1 and E-cadherin) were not altered. In summary, neonatal co-infection led to persistent nasopharyngeal colonisation and increased airway responsiveness that was not associated with detectable smooth muscle or mucosal epithelial abnormalities, however increased hysteresivity may reflect ventilation heterogeneity.


Sujet(s)
Asthme/anatomopathologie , Virus de la grippe A/croissance et développement , Poumon/microbiologie , Poumon/physiologie , Infections à Orthomyxoviridae/complications , Infections à pneumocoques/complications , Streptococcus pneumoniae/croissance et développement , Résistance des voies aériennes , Animaux , Animaux nouveau-nés , Bronchoconstricteurs/administration et posologie , Co-infection/complications , Femelle , Histocytochimie , Poumon/anatomopathologie , Chlorure de méthacholine/administration et posologie , Souris de lignée C57BL , Tests de la fonction respiratoire
15.
Br J Pharmacol ; 173(4): 635-48, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26013585

RÉSUMÉ

Cigarette smoking has reached epidemic proportions within many regions of the world and remains the highest risk factor for chronic obstructive pulmonary disease (COPD) and lung cancer. Squamous cell lung cancer is commonly detected in heavy smokers, where the risk of developing lung cancer is not solely defined by tobacco consumption. Although therapies that target common driver mutations in adenocarcinomas are showing some promise, they are proving ineffective in smoking-related squamous cell lung cancer. Since COPD is characterized by an excessive inflammatory and oxidative stress response, this review details how aberrant innate, adaptive and systemic inflammatory processes can contribute to lung cancer susceptibility in COPD. Activated leukocytes release increasing levels of proteases and free radicals as COPD progresses and tertiary lymphoid aggregates accumulate with increasing severity. Reactive oxygen species promote formation of reactive carbonyls that are not only tumourigenic through initiating DNA damage, but can directly alter the function of regulatory proteins involved in host immunity and tumour suppressor functions. Systemic inflammation is also markedly increased during infective exacerbations in COPD and the interplay between tumour-promoting serum amyloid A (SAA) and IL-17A is discussed. SAA is also an endogenous allosteric modifier of FPR2 expressed on immune and epithelial cells, and the therapeutic potential of targeting this receptor is proposed as a novel strategy for COPD-lung cancer overlap.


Sujet(s)
Carcinome épidermoïde/immunologie , Carcinome épidermoïde/physiopathologie , Immunité/immunologie , Inflammation/immunologie , Tumeurs du poumon/immunologie , Tumeurs du poumon/physiopathologie , Broncho-pneumopathie chronique obstructive/immunologie , Broncho-pneumopathie chronique obstructive/physiopathologie , Humains , Inflammation/physiopathologie , Tumeurs du poumon/anatomopathologie
17.
Clin Sci (Lond) ; 129(9): 785-96, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26201093

RÉSUMÉ

Cigarette smoke (CS) is the major cause of chronic obstructive pulmonary disease (COPD). Interleukin-17A (IL-17A) is a pivotal cytokine that regulates lung immunity and inflammation. The aim of the present study was to investigate how IL-17A regulates CS-induced lung inflammation in vivo. IL-17A knockout (KO) mice and neutralization of IL-17A in wild-type (WT) mice reduced macrophage and neutrophil recruitment and chemokine (C-C motif) ligand 2 (CCL2), CCL3 and matrix metalloproteinase (MMP)-12 mRNA expression in response to acute CS exposure. IL-17A expression was increased in non-obese diabetic (NOD) severe combined immunodeficiency SCID) mice with non-functional B- and T-cells over a 4-week CS exposure period, where macrophages accumulated to the same extent as in WT mice. Gene expression analysis by QPCR (quantitative real-time PCR) of isolated immune cell subsets detected increased levels of IL-17A transcript in macrophages, neutrophils and NK/NKT cells in the lungs of CS-exposed mice. In order to further explore the relative contribution of innate immune cellular sources, intracellular IL-17A staining was performed. In the present study, we demonstrate that CS exposure primes natural killer (NK), natural killer T (NKT) and γδ T-cells to produce more IL-17A protein and CS alone increased the frequency of IL17+ γδ T-cells in the lung, whereas IL-17A protein was not detected in macrophages and neutrophils. Our data suggest that activation of innate cellular sources of IL-17A is an essential mediator of macrophage accumulation in CS-exposed lungs. Targeting non-conventional T-cell sources of IL-17A may offer an alternative strategy to reduce pathogenic macrophages in COPD.


Sujet(s)
Interleukine-17/immunologie , Macrophages/immunologie , Nicotiana/composition chimique , Pneumopathie infectieuse/immunologie , Fumée , Animaux , Liquide de lavage bronchoalvéolaire/cytologie , Liquide de lavage bronchoalvéolaire/immunologie , Cellules cultivées , Chimiokine CCL2/génétique , Chimiokine CCL2/immunologie , Chimiokine CCL2/métabolisme , Chimiokine CCL3/génétique , Chimiokine CCL3/immunologie , Chimiokine CCL3/métabolisme , Cytométrie en flux , Expression des gènes/immunologie , Immunité innée/génétique , Immunité innée/immunologie , Interleukine-17/génétique , Interleukine-17/métabolisme , Poumon/immunologie , Poumon/métabolisme , Poumon/anatomopathologie , Macrophages/métabolisme , Matrix metalloproteinase 12/génétique , Matrix metalloproteinase 12/immunologie , Matrix metalloproteinase 12/métabolisme , Souris de lignée C57BL , Souris de lignée NOD , Souris knockout , Souris SCID , Cellules T tueuses naturelles/immunologie , Cellules T tueuses naturelles/métabolisme , Infiltration par les neutrophiles/immunologie , Pneumopathie infectieuse/génétique , Pneumopathie infectieuse/métabolisme , Récepteur lymphocytaire T antigène, gamma-delta/immunologie , Récepteur lymphocytaire T antigène, gamma-delta/métabolisme , RT-PCR
18.
Methods Mol Biol ; 1235: 231-41, 2015.
Article de Anglais | MEDLINE | ID: mdl-25388397

RÉSUMÉ

Clonal culture of stem cells is crucial for their identification, and the characterization of the cellular and molecular mechanisms that regulate their proliferation and differentiation. In the adult mouse lung, epithelial stem/progenitor cells are defined by the phenotype CD45(neg) CD31(neg) EpCAM(pos) CD104(pos) CD24(low). Here we describe a tissue dissociation and flow cytometry strategy for the detection and isolation of adult mouse lung epithelial stem/progenitor cells, and a three-dimensional colony-forming assay for their clonal culture in vitro.


Sujet(s)
Cellules souches adultes/cytologie , Séparation cellulaire/méthodes , Cellules épithéliales/cytologie , Cytométrie en flux/méthodes , Poumon/cytologie , Animaux , Antigènes néoplasiques/analyse , Antigènes CD24/analyse , Molécules d'adhérence cellulaire/analyse , Techniques de culture cellulaire/méthodes , Prolifération cellulaire , Cellules cultivées , Test clonogénique/méthodes , Molécule d'adhérence des cellules épithéliales , Intégrine bêta4/analyse , Antigènes CD45/analyse , Souris , Antigènes CD31/analyse
19.
Int J Biochem Cell Biol ; 56: 82-91, 2014 Nov.
Article de Anglais | MEDLINE | ID: mdl-25450456

RÉSUMÉ

In response to recurrent exposure to environmental insults such as allergens, pollution, irritants, smoke and viral/bacterial infection, the epithelium of the lung is continually damaged. Homeostasis of the lung requires a balance between immune regulation and promotion of tissue regeneration, which requires the co-ordinated proliferation and differentiation of stem and progenitor cells. In this review we reflect on the current understanding of lung epithelial stem and progenitor cells and advocate a model hierarchy in which self-renewing multipotent lung epithelial stem cells give rise to lineage restricted progenitor cells that repopulate airway and alveolar epithelial cell lineages during homeostasis and repair. We also discuss the role of mesenchymal progenitor cells in maintaining the structural integrity of the lung and propose a model in which mesenchymal cells act as the quintessential architects of lung regeneration by providing molecular signals, such as FGF-10, to regulate the fate and specificity of epithelial stem and progenitor cells. Moreover, we discuss the current status and future prospects for translating lung stem cell therapies to the clinic to replace, repair, or regenerate diseased lung tissue. This article is part of a directed issue entitled: Regenerative Medicine: the challenge of translation.


Sujet(s)
Poumon/cytologie , Poumon/physiologie , Régénération/physiologie , Médecine régénérative/méthodes , Cellules souches/cytologie , Différenciation cellulaire , Prolifération cellulaire , Cellules épithéliales/cytologie , Homéostasie/physiologie , Humains , Modèles biologiques , Médecine régénérative/tendances
20.
FASEB J ; 28(9): 3867-77, 2014 Sep.
Article de Anglais | MEDLINE | ID: mdl-24846388

RÉSUMÉ

Serum amyloid A (SAA) is expressed locally in chronic inflammatory conditions such as chronic obstructive pulmonary disease (COPD), where macrophages that do not accord with the classic M1/M2 paradigm also accumulate. In this study, the role of SAA in regulating macrophage differentiation was investigated in vitro using human blood monocytes from healthy subjects and patients with COPD and in vivo using an airway SAA challenge model in BALB/c mice. Differentiation of human monocytes with SAA stimulated the proinflammatory monokines IL-6 and IL-1ß concurrently with the M2 markers CD163 and IL-10. Furthermore, SAA-differentiated macrophages stimulated with lipopolysaccharide (LPS) expressed markedly higher levels of IL-6 and IL-1ß. The ALX/FPR2 antagonist WRW4 reduced IL-6 and IL-1ß expression but did not significantly inhibit phagocytic and efferocytic activity. In vivo, SAA administration induced the development of a CD11c(high)CD11b(high) macrophage population that generated higher levels of IL-6, IL-1ß, and G-CSF following ex vivo LPS challenge. Blocking CSF-1R signaling effectively reduced the number of CD11c(high)CD11b(high) macrophages by 71% and also markedly inhibited neutrophilic inflammation by 80%. In conclusion, our findings suggest that SAA can promote a distinct CD11c(high)CD11b(high) macrophage phenotype, and targeting this population may provide a novel approach to treating chronic inflammatory conditions associated with persistent SAA expression.


Sujet(s)
Différenciation cellulaire , Poumon/cytologie , Macrophages/cytologie , Broncho-pneumopathie chronique obstructive/anatomopathologie , Récepteur du facteur de stimulation des colonies de macrophages/métabolisme , Protéine amyloïde A sérique/métabolisme , Animaux , Technique de Western , Études cas-témoins , Prolifération cellulaire , Cellules cultivées , Cytométrie en flux , Hématopoïèse , Humains , Inflammation/immunologie , Inflammation/métabolisme , Inflammation/anatomopathologie , Médiateurs de l'inflammation/métabolisme , Interleukine-1 bêta/métabolisme , Lipopolysaccharides/pharmacologie , Poumon/immunologie , Poumon/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Mâle , Souris , Souris de lignée BALB C , Monocytes/cytologie , Monocytes/immunologie , Monocytes/métabolisme , Granulocytes neutrophiles/cytologie , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Phagocytose/physiologie , Broncho-pneumopathie chronique obstructive/immunologie , Broncho-pneumopathie chronique obstructive/métabolisme , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , Récepteur du facteur de stimulation des colonies de macrophages/génétique , RT-PCR , Protéine amyloïde A sérique/génétique , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...