1
|
Wang Z, Vernaz J, Tagaras N, Boda B, Buerki-Thurnherr T, Reina G, Kissling VM, Constant S, Gupta G, Wick P. Silicon Carbide Nanowires Impair Mucociliary Clearance-Mediated Innate Immunity in Primary Human Bronchial Epithelial Cells. ACS NANO 2025; 19:21426-21445. [PMID: 40474814 PMCID: PMC12177949 DOI: 10.1021/acsnano.5c01981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 05/23/2025] [Accepted: 05/23/2025] [Indexed: 06/18/2025]
Abstract
The respiratory tract possesses mucociliary-driven innate immune defense mechanisms that protect the lungs from harmful environmental exposures, but when damaged, increase susceptibility to respiratory infections and diseases. Inhalation exposure to certain nanomaterials has been shown to trigger fibrosis and other respiratory conditions. However, there is a limited understanding of whether nanomaterials can impair mucociliary defense in lungs and its underlying mechanism. Here, we first investigated the fate of zero-dimensional, one-dimensional, and two-dimensional silicon- and carbon-based nanomaterials (silicon carbide nanowires (SiC NWs), silicon dioxide (SiO2), multiwalled carbon nanotubes (MWCNTs), and graphene nanosheets) in airway mucus. The results demonstrated that only SiC NWs escaped through the mucus gel without interactions, suggesting their potential to diffuse across the protective mucus layer. The hydrophobicity of the SiC NWs, associated with the low abundance of polar surface groups, such as silanols, was mainly responsible for the observed shielding of particle interactions with mucus components. Furthermore, repeated exposure to SiC NWs in primary bronchial epithelial cell cultures revealed abnormal ciliary structure and significantly (p < 0.05) compromised mucociliary clearance functions, however, no such effects were evident for other particles. mRNA expression analysis showed a significant (p < 0.05) increase in FOX-J1 transcripts, suggesting transcriptional dysregulation of ciliogenesis after exposure to SiC NWs. Finally, SiC NWs reduced epithelial barrier integrity and promoted pro-inflammatory and pro-fibrotic responses. These findings unravel the hazardous potential of SiC NWs upon inhalation exposure and identify the breaching and impairment of the mucociliary innate defense as a key event in their respiratory toxicity.
Collapse
Affiliation(s)
- Ziting Wang
- Nanomaterials
in Health Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and
Technology (Empa), St. Gallen9014, Switzerland
| | - Jimmy Vernaz
- Epithelix
Sàrl, 18 chemin
des Aulx, 1228Plan-les-Ouates, Switzerland
| | - Nikolaos Tagaras
- Nanomaterials
in Health Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and
Technology (Empa), St. Gallen9014, Switzerland
| | - Bernadett Boda
- Epithelix
Sàrl, 18 chemin
des Aulx, 1228Plan-les-Ouates, Switzerland
| | - Tina Buerki-Thurnherr
- Nanomaterials
in Health Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and
Technology (Empa), St. Gallen9014, Switzerland
| | - Giacomo Reina
- Nanomaterials
in Health Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and
Technology (Empa), St. Gallen9014, Switzerland
| | - Vera M. Kissling
- Nanomaterials
in Health Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and
Technology (Empa), St. Gallen9014, Switzerland
| | - Samuel Constant
- Epithelix
Sàrl, 18 chemin
des Aulx, 1228Plan-les-Ouates, Switzerland
| | - Govind Gupta
- Nanomaterials
in Health Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and
Technology (Empa), St. Gallen9014, Switzerland
| | - Peter Wick
- Nanomaterials
in Health Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and
Technology (Empa), St. Gallen9014, Switzerland
| |
Collapse
|
2
|
Leinardi R, Longo Sanchez-Calero C, Ibouraadaten S, Uwambayinema F, Yakoub Y, Pavan C, Claus R, Lemaire F, Ronsmans S, Ghosh M, Farczádi L, Moldovan H, Vanoirbeek JAJ, Turci F, Hoet PHM, Huaux F. Dynamic biodistribution of inhaled silica particles to extrapulmonary sites: Early and late translocation mechanisms with implication for particle biomonitoring. ENVIRONMENT INTERNATIONAL 2025; 199:109473. [PMID: 40250236 DOI: 10.1016/j.envint.2025.109473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
An innovative method based on inductively coupled plasma mass spectrometry (ICP-MS) was developed to quantify the time-dependent systemic redistribution pattern of pulmonary-deposited crystalline silica particles by measuring silicon (Si) levels in the lungs, distal organs, and biological fluids. The method was applied in a murine model and validated in blood and urine samples from two occupationally exposed cohorts (miners and porcelain industry workers). In mice, 30 % of silica particles deposited in the lungs via oropharyngeal administration accumulated in extrapulmonary sites in less than 4 months. An early translocation (within 3 days) resulted in silica distribution to liver and kidneys (13 %), followed by a delayed migration (up to 60 days) in mediastinal lymph nodes (12 %), spleen (1.7 %), and abdominal skin (1.7 %). The long-term increase of Si in urine suggested silica renal clearance. Our data also indicated that the toxic potential of particles is a key determinant of extrapulmonary redistribution. The interest of Si as biomarker of exposure has been confirmed in workers exposed to crystalline silica dust. In these individuals, elevated Si levels in blood and urine paralleled silica exposure. Our findings quantify the dynamics of silica biodistribution in extrapulmonary organs, offering new insights on the biomonitoring of silica exposure in different scenarios.
Collapse
Affiliation(s)
- Riccardo Leinardi
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 57, 1200 Woluwe-Saint-Lambert, Brussels, Belgium.
| | - Chiara Longo Sanchez-Calero
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 57, 1200 Woluwe-Saint-Lambert, Brussels, Belgium
| | - Saloua Ibouraadaten
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 57, 1200 Woluwe-Saint-Lambert, Brussels, Belgium
| | - Francine Uwambayinema
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 57, 1200 Woluwe-Saint-Lambert, Brussels, Belgium
| | - Yousof Yakoub
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 57, 1200 Woluwe-Saint-Lambert, Brussels, Belgium
| | - Cristina Pavan
- Department of Chemistry, University of Turin, Via P. Giuria 7, 10125 Turin, Italy; "G. Scansetti" Interdepartmental Centre for Studies on Asbestos and Other Toxic Particulates, University of Turin, Via P. Giuria 7, 10125 Turin, Italy
| | - Rani Claus
- Environment and Health Unit, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Frauke Lemaire
- Environment and Health Unit, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Steven Ronsmans
- Environment and Health Unit, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Manosij Ghosh
- Environment and Health Unit, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Lénárd Farczádi
- Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology (UMFST), Gh. Marinescu 38, 540142 Târgu Mureş, Romania
| | - Horatiu Moldovan
- Department of Occupational Medicine, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology (UMFST), Gh. Marinescu 38, 540142 Târgu Mureş, Romania
| | - Jeroen A J Vanoirbeek
- Environment and Health Unit, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Francesco Turci
- Department of Chemistry, University of Turin, Via P. Giuria 7, 10125 Turin, Italy; "G. Scansetti" Interdepartmental Centre for Studies on Asbestos and Other Toxic Particulates, University of Turin, Via P. Giuria 7, 10125 Turin, Italy
| | - Peter H M Hoet
- Environment and Health Unit, KU Leuven, Herestraat 49, 3000 Leuven, Belgium; Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 57, 1200 Woluwe-Saint-Lambert, Brussels, Belgium
| |
Collapse
|
3
|
Galli G, Leleu D, Depaire A, Blanco P, Contin-Bordes C, Truchetet ME. Crystalline silica on the lung-environment interface: Impact on immunity, epithelial cells, and therapeutic perspectives for autoimmunity. Autoimmun Rev 2025; 24:103730. [PMID: 39701338 DOI: 10.1016/j.autrev.2024.103730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Crystalline silica (the most abundant form of silicon dioxide) is a natural element that is ubiquitous in the Earth's crust. Chronic personal or professional exposure has been implicated in various pathologies, including silicosis and autoimmune diseases since the early 20th century. More recently, a specific pathogenic role for crystalline silica has been identified through its impact on lung epithelial cells as well as immune cells present at this organism barrier. This review summarizes the current in vitro and in vivo knowledge regarding the physiopathology of crystalline silica at the lung-environment interface, discusses its effects on innate and adaptive immune cells and epithelial cells, and reviews current therapeutic perspectives explored in mouse models to alleviate its impact, especially on autoimmune phenotypes.
Collapse
Affiliation(s)
- Gaël Galli
- Univ. Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, F-33000 Bordeaux, France; CHU de Bordeaux, FHU ACRONIM, Centre national de référence des maladies auto-immunes et systémiques rares Est/Sud-Ouest (RESO), F-33000 Bordeaux, France; CHU de Bordeaux, Service de Médecine Interne, Immunologie Clinique et Maladies Infectieuses, UMR 5164, F-33000 Bordeaux, France.
| | - Damien Leleu
- Univ. Bourgogne Franche-Comté, INSERM, LNC UMR1231, LabEx LipSTIC, F-21000 Dijon, France; CHRU Dijon Bourgogne, Laboratory of Clinical Chemistry, F-21000 Dijon, France
| | - Agathe Depaire
- Univ. Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, F-33000 Bordeaux, France; MED'INN'Pharma, F-25000 Besançon, France
| | - Patrick Blanco
- Univ. Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, F-33000 Bordeaux, France; CHU de Bordeaux, FHU ACRONIM, Centre national de référence des maladies auto-immunes et systémiques rares Est/Sud-Ouest (RESO), F-33000 Bordeaux, France; CHU de Bordeaux, Service d'Immunologie, UMR 5164, F-33000 Bordeaux, France
| | - Cécile Contin-Bordes
- Univ. Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, F-33000 Bordeaux, France; CHU de Bordeaux, FHU ACRONIM, Centre national de référence des maladies auto-immunes et systémiques rares Est/Sud-Ouest (RESO), F-33000 Bordeaux, France; CHU de Bordeaux, Service d'Immunologie, UMR 5164, F-33000 Bordeaux, France
| | - Marie-Elise Truchetet
- Univ. Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, F-33000 Bordeaux, France; CHU de Bordeaux, FHU ACRONIM, Centre national de référence des maladies auto-immunes et systémiques rares Est/Sud-Ouest (RESO), F-33000 Bordeaux, France; CHU de Bordeaux, Service de Rhumatologie, UMR 5164, F-33000 Bordeaux, France
| |
Collapse
|
4
|
Pavan C, Leinardi R, Benhida A, Ibouraadaten S, Yakoub Y, Brule SVD, Lison D, Turci F, Huaux F. Short- and long-term pathologic responses to quartz are induced by nearly free silanols formed during crystal fracturing. Part Fibre Toxicol 2024; 21:52. [PMID: 39633374 PMCID: PMC11619699 DOI: 10.1186/s12989-024-00611-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Inhalation of respirable crystalline silica particles, including quartz, is associated with an increased risk of developing pathologies, including persistent lung inflammation, fibrosis, cancer, and systemic autoimmunity. We demonstrated that the nearly free silanols (NFS) generated upon quartz fracturing trigger the early molecular events determining quartz toxicity. Here, we address the involvement of NFS in driving short- and long-term pathogenic responses, including lung inflammation, fibrosis, cancer, and autoimmunity in multiple mouse models. RESULTS In vivo pulmonary responses to as-grown NFS-poor quartz (gQ) and fractured NFS-rich quartz (gQ-f) of synthetic origin were compared to two NFS-rich reference quartz dusts (Min-U-Sil 5, mQ-f). Acute and persistent inflammation, as well as fibrosis, were assessed 3 and 60 days, respectively, after administering one dose of particles (2 mg) via oropharyngeal aspiration (o.p.a.) to C57BL/6 mice. The carcinogenic potential was assessed in a co-carcinogenicity study using A/J mice, which were pre-treated with 3-methylcholanthrene (3-MC) and administered four doses of quartz particles (4 × 1 mg, o.p.a.), then sacrificed after 10 months. Autoimmunity was evaluated in autoimmune-prone 129/Sv mice 4 months after particle administration (2 × 1.25 mg, o.p.a). Mice exposed to NFS-rich quartz exhibited a strong acute lung inflammatory response, characterized by pro-inflammatory cytokine release and leukocyte accumulation, which persisted for up to 60 days. No inflammatory effect was observed in mice treated with NFS-poor gQ. Fibrosis onset (i.e., increased levels of pro-fibrotic factors, hydroxyproline, and collagen) was prominent in mice exposed to NFS-rich but not to NFS-poor quartz. Additionally, lung cancer development (tumour numbers) and autoimmune responses (elevated IgG and anti-dsDNA autoantibody levels) were only observed after exposure to NFS-rich quartz. CONCLUSIONS Collectively, the results indicate that NFS, which occur upon fracturing of quartz particles, play a crucial role in the short- and long-term local and systemic responses to quartz. The assessment of NFS on amorphous or crystalline silica particles may help create a predictive model of silica pathogenicity.
Collapse
Affiliation(s)
- Cristina Pavan
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium.
- Department of Chemistry, University of Turin, Turin, Italy.
- "G. Scansetti" Interdepartmental Centre for Studies on Asbestos and Other Toxic Particulates, University of Turin, Turin, Italy.
| | - Riccardo Leinardi
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Anissa Benhida
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Saloua Ibouraadaten
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Yousof Yakoub
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Sybille van den Brule
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Dominique Lison
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Francesco Turci
- Department of Chemistry, University of Turin, Turin, Italy
- "G. Scansetti" Interdepartmental Centre for Studies on Asbestos and Other Toxic Particulates, University of Turin, Turin, Italy
| | - François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium.
| |
Collapse
|
5
|
Abstract
PURPOSE OF REVIEW There has been a rapid increase in silicosis cases, particularly related to artificial stone. The key to management is avoidance of silica exposure. Despite this, many develop progressive disease and there are no routinely recommended treatments. This review provides a summary of the literature pertaining to pharmacological therapies for silicosis and examines the plausibility of success of such treatments given the disease pathogenesis. RECENT FINDINGS In-vitro and in-vivo models demonstrate potential efficacy for drugs, which target inflammasomes, cytokines, effector cells, fibrosis, autophagy, and oxidation. SUMMARY There is some evidence for potential therapeutic targets in silicosis but limited translation into human studies. Treatment of silicosis likely requires a multimodal approach, and there is considerable cross-talk between pathways; agents that modulate both inflammation, fibrosis, autophagy, and ROS production are likely to be most efficacious.
Collapse
Affiliation(s)
- Hayley Barnes
- Monash Centre for Occupational and Environmental Health, Monash University
- Department of Respiratory Medicine, Alfred Health
- Central Clinical School, Monash University, Melbourne
| | - Maggie Lam
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Michelle D Tate
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Ryan Hoy
- Monash Centre for Occupational and Environmental Health, Monash University
- Department of Respiratory Medicine, Alfred Health
| |
Collapse
|
6
|
Lescoat A, Leinardi R, Pouxvielh K, Yakoub Y, Lelong M, Pochet A, Dumontet E, Bellamri N, Le Tallec E, Pavan C, Turci F, Paris C, Huaux F, Lecureur V. Effects of different amosite preparations on macrophages, lung damages, and autoimmunity. J Mol Med (Berl) 2024; 102:197-211. [PMID: 38015242 DOI: 10.1007/s00109-023-02401-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
The underlying mechanisms of asbestos-related autoimmunity are poorly understood. As the size, surface reactivity, and free radical activity of asbestos particles are considered crucial regarding the health effects, this study aims to compare the effects of exposure to pristine amosite (pAmo) or milled amosite (mAmo) particles on lung damage, autoimmunity, and macrophage phenotype. Four months after lung exposure to 0.1 mg of amosite, BAL levels of lactate dehydrogenase, protein, free DNA, CCL2, TGF-β1, TIMP-1, and immunoglobulin A of pAmo-exposed C57Bl/6 mice were increased when compared to fluids from control- and mAmo-exposed mice. Effects in pAmo-exposed mice were associated with lung fibrosis and autoimmunity including anti-double-strand DNA autoantibody production. mAmo or pAmo at 20 µg/cm2 induced a pro-inflammatory phenotype characterized by a significant increase in TNFα and IL-6 secretion on human monocyte-derived macrophages (MDMs). mAmo and pAmo exposure induced a decrease in the efferocytosis capacities of MDMs, whereas macrophage abilities to phagocyte fluorescent beads were unchanged when compared to control MDMs. mAmo induced IL-6 secretion and reduced the percentage of MDMs expressing MHCII and CD86 markers involved in antigen and T-lymphocyte stimulation. By contrast, pAmo but not mAmo activated the NLRP3 inflammasome, as evaluated through quantification of caspase-1 activity and IL-1β secretion. Our results demonstrated that long-term exposure to pAmo may induce significant lung damage and autoimmune effects, probably through an alteration of macrophage phenotype, supporting in vivo the higher toxicity of entire amosite (pAmo) with respect to grinded amosite. However, considering their impact on efferocytosis and co-stimulation markers, mAmo effects should not be neglected. KEY MESSAGES: Lung fibrosis and autoimmunity induced by amosite particles depend on their physicochemical characteristics (size and surface) Inhalation exposure of mice to pristine amosite fibers is associated with lung fibrosis and autoimmunity Anti-dsDNA antibody is a marker of autoimmunity in mice exposed to pristine amosite fibers Activation of lung mucosa-associated lymphoid tissue, characterized by IgA production, after exposure to pristine amosite fibers Pristine and milled amosite particle exposure reduced the efferocytosis capacity of human-derived macrophages.
Collapse
Affiliation(s)
- Alain Lescoat
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35000, Rennes, France
- Department of Internal Medicine & Clinical Immunology, Rennes University Hospital, 35000, Rennes, France
| | - Riccardo Leinardi
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Expérimentale Et Clinique (IREC), Université Catholique de Louvain (UCL), Avenue Hippocrate 57, Bte B-1.57.06, 1200, Brussels, Belgium
| | - Kévin Pouxvielh
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35000, Rennes, France
| | - Yousof Yakoub
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Expérimentale Et Clinique (IREC), Université Catholique de Louvain (UCL), Avenue Hippocrate 57, Bte B-1.57.06, 1200, Brussels, Belgium
| | - Marie Lelong
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35000, Rennes, France
| | - Amandine Pochet
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Expérimentale Et Clinique (IREC), Université Catholique de Louvain (UCL), Avenue Hippocrate 57, Bte B-1.57.06, 1200, Brussels, Belgium
| | | | - Nessrine Bellamri
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35000, Rennes, France
| | - Erwan Le Tallec
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35000, Rennes, France
- Department of Internal Medicine & Clinical Immunology, Rennes University Hospital, 35000, Rennes, France
| | - Cristina Pavan
- "G. Scansetti" Interdepartmental Center for Studies On Asbestos and Other Toxic Particulates, University of Turin, Via Pietro Giuria 7, 10125, Turin, Italy
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, 10125, Turin, Italy
| | - Francesco Turci
- "G. Scansetti" Interdepartmental Center for Studies On Asbestos and Other Toxic Particulates, University of Turin, Via Pietro Giuria 7, 10125, Turin, Italy
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, 10125, Turin, Italy
| | - Christophe Paris
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35000, Rennes, France
- Service de Santé Au Travail Et Pathologie Professionnelle, CHU Rennes, 35000, Rennes, France
| | - François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Expérimentale Et Clinique (IREC), Université Catholique de Louvain (UCL), Avenue Hippocrate 57, Bte B-1.57.06, 1200, Brussels, Belgium
| | - Valérie Lecureur
- Univ Rennes, CHU Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35000, Rennes, France.
- UMR-INSERM 1085, Campus Santé, 2 Avenue du Pr Léon Bernard, 35043, Rennes Cedex, France.
| |
Collapse
|
7
|
Liu X, Dai K, Zhang X, Huang G, Lynn H, Rabata A, Liang J, Noble PW, Jiang D. Multiple Fibroblast Subtypes Contribute to Matrix Deposition in Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2023; 69:45-56. [PMID: 36927333 PMCID: PMC10324043 DOI: 10.1165/rcmb.2022-0292oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 03/16/2023] [Indexed: 03/18/2023] Open
Abstract
Progressive pulmonary fibrosis results from a dysfunctional tissue repair response and is characterized by fibroblast proliferation, activation, and invasion and extracellular matrix accumulation. Lung fibroblast heterogeneity is well recognized. With single-cell RNA sequencing, fibroblast subtypes have been reported by recent studies. However, the roles of fibroblast subtypes in effector functions in lung fibrosis are not well understood. In this study, we incorporated the recently published single-cell RNA-sequencing datasets on murine lung samples of fibrosis models and human lung samples of fibrotic diseases and analyzed fibroblast gene signatures. We identified and confirmed the novel fibroblast subtypes we reported recently across all samples of both mouse models and human lung fibrotic diseases, including idiopathic pulmonary fibrosis, systemic sclerosis-associated interstitial lung disease, and coronavirus disease (COVID-19). Furthermore, we identified specific cell surface proteins for each fibroblast subtype through differential gene expression analysis, which enabled us to isolate primary cells representing distinct fibroblast subtypes by flow cytometry sorting. We compared matrix production, including fibronectin, collagen, and hyaluronan, after profibrotic factor stimulation and assessed the invasive capacity of each fibroblast subtype. Our results suggest that in addition to myofibroblasts, lipofibroblasts and Ebf1+ (Ebf transcription factor 1+) fibroblasts are two important fibroblast subtypes that contribute to matrix deposition and also have enhanced invasive, proliferative, and contraction phenotypes. The histological locations of fibroblast subtypes are identified in healthy and fibrotic lungs by these cell surface proteins. This study provides new insights to inform approaches to targeting lung fibroblast subtypes to promote the development of therapeutics for lung fibrosis.
Collapse
Affiliation(s)
- Xue Liu
- Department of Medicine and Women’s Guild Lung Institute and
| | - Kristy Dai
- Department of Medicine and Women’s Guild Lung Institute and
| | - Xuexi Zhang
- Department of Medicine and Women’s Guild Lung Institute and
| | - Guanling Huang
- Department of Medicine and Women’s Guild Lung Institute and
| | - Heather Lynn
- Department of Medicine and Women’s Guild Lung Institute and
| | - Anas Rabata
- Department of Medicine and Women’s Guild Lung Institute and
| | - Jiurong Liang
- Department of Medicine and Women’s Guild Lung Institute and
| | - Paul W. Noble
- Department of Medicine and Women’s Guild Lung Institute and
| | - Dianhua Jiang
- Department of Medicine and Women’s Guild Lung Institute and
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
8
|
Heiss J, Grün K, Tempel L, Matasci M, Schrepper A, Schwarzer M, Bauer R, Förster M, Berndt A, Jung C, Schulze PC, Neri D, Franz M. Targeted Interleukin-9 delivery in pulmonary hypertension: Comparison of immunocytokine formats and effector cell study. Eur J Clin Invest 2023; 53:e13907. [PMID: 36377348 DOI: 10.1111/eci.13907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/19/2022] [Accepted: 10/27/2022] [Indexed: 11/16/2022]
Abstract
AIMS Pulmonary hypertension (PH) is accompanied by pulmonary vascular remodelling. By targeted delivery of Interleukin-9 (IL9) via the immunocytokine F8IL9, beneficial effects could be demonstrated in a mouse model of PH. This study aimed to compare two immunocytokine formats (single-chain Fv and full IgG) and to identify potential target cells of IL9. METHODS The Monocrotaline mouse model of PH (PH, n = 12) was chosen to evaluate the treatment effects of F8IL9F8 (n = 12) and F8IgGIL9 (n = 6) compared with sham-induced animals (control, n = 10), the dual endothelin receptor antagonist Macitentan (MAC, n = 12) or IL9-based immunocytokines with irrelevant antigen specificity (KSFIL9KSF, n = 12; KSFIgGIL9 n = 6). Besides comparative validation of treatment effects, the study was focused on the detection and quantification of mast cells (MCs) and regulatory T cells (Tregs). RESULTS There was a significantly elevated systolic right ventricular pressure (104 ± 36 vs. 45 ± 17 mmHg) and an impairment of right ventricular echocardiographic parameters (RVbasal: 2.52 ± 0.25 vs. 1.94 ± 0.13 mm) in untreated PH compared with controls (p < 0.05). Only the groups treated with F8IL9, irrespective of the format, showed consistent beneficial effects (p < 0.05). Moreover, F8IL9F8 but not F8IgGIL9 treatment significantly reduced lung tissue damage compared with untreated PH mice (p < 0.05). There was a significant increase in Tregs in F8IL9-treated compared with control animals, the untreated PH and the MAC group (p < 0.05). CONCLUSIONS Beneficial treatment effects of targeted IL9 delivery in a preclinical model of PH could be convincingly validated. IL9-mediated recruitment of Tregs into lung tissue might play a crucial role in the induction of anti-inflammatory and anti-proliferative mechanisms potentially contributing to a novel disease-modifying concept.
Collapse
Affiliation(s)
- Judith Heiss
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany.,Else Kröner Graduate School for Medical Students "JSAM", Jena University Hospital, Jena, Germany
| | - Katja Grün
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | - Laura Tempel
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | | | - Andrea Schrepper
- Department of Cardiothoracic Surgery, University Hospital Jena, Jena, Germany
| | - Michael Schwarzer
- Department of Cardiothoracic Surgery, University Hospital Jena, Jena, Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital Jena, Jena, Germany
| | - Martin Förster
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | - Alexander Berndt
- Section Pathology, Institute of Legal Medicine, University Hospital Jena, Jena, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | | | | | - Marcus Franz
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| |
Collapse
|
9
|
Effects of early dexamethasone treatment on several markers of inflammation and fibrosis in an animal model of lung silicosis in rats – A pilot study. ACTA MEDICA MARTINIANA 2022. [DOI: 10.2478/acm-2022-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Lung silicosis is primarily caused by inhalation of particles of silicon oxide (silica). Despite a huge progress in understanding the interactions among the pathomechanisms of lung silicosis in the last years, there is a lack of effective therapy. With respect to a wide therapeutic action of corticosteroids, the purpose of this pilot study was to evaluate early effects of dexamethasone on several markers of inflammation and lung fibrosis in a rat model of silicosis. The silicosis model was induced by a single transoral intratracheal instillation of silica (50 mg/ml/animal), while the controls received an equivalent volume of sterile saline. The treatment with intraperitoneal dexamethasone initiated the next day after the silica instillation and was given 2-times a week at a dose of 1 mg/kg, while the controls received an equivalent volume of saline. The animals were euthanized 14 or 28 days after the treatment onset. Total and differential counts of leukocytes in the blood and bronchoalveolar lavage (BAL) fluid were determined. The presence of collagen in the bronchioles and lung vessels was detected by Sirius red staining and a smooth muscle mass was detected by smooth muscle actin. In comparison to saline, the instillation of silica increased the total count of circulating leukocytes after 14 and 28 days of the experiment (both p<0.05), which was associated with higher counts of lymphocytes (p<0.05 after 14 days, p>0.05 after 28 days) and slight but non-significant increases in neutrophils and eosinophils (both p>0.05). Although the total cell count in the BAL fluid did not change significantly, the percentages and absolute counts of neutrophils, eosinophils, and lymphocytes (p<0.05, p<0.01 or p<0.001) elevated after 14 and 28 days of the experiment. Silica induced an accumulation of collagen in the bronchioles (p<0.001 after both 14 and 28 days) and pulmonary vessels (p<0.01 after both 14 and 28 days) and elevated a formation of smooth muscle mass (p<0.05 after 14 days, p<0.01 or p<0.001 after 28 days). Treatment with dexamethasone decreased circulating leukocytes (p<0.01) and lymphocytes (p<0.001) and increased neutrophils (p<0.05), which was associated with a slightly decreased total cell count in BAL (p>0.05), decline in lymphocytes (p<0.01), and slight decreases in neutrophils and eosinophils after 28 days of the treatment. Moreover, dexamethasone reduced the accumulation of collagen (p<0.01 after 14 days and p<0.001 after 28 days) and the formation of smooth muscle mass (p<0.01 for bronchioles and p>0.05 for vessels after 24 days, p<0.001 for both bronchioles and vessels after 28 days). In conclusion, early dexamethasone treatment mitigated silica-induced granulocytic-lymphocytic inflammation and decreased a formation of collagen and smooth muscle mass in the bronchiolar and vascular walls, demonstrating a therapeutic potential of dexamethasone in the lung silicosis.
Collapse
|
10
|
Pulmonary Toxicity of Silica Linked to Its Micro- or Nanometric Particle Size and Crystal Structure: A Review. NANOMATERIALS 2022; 12:nano12142392. [PMID: 35889616 PMCID: PMC9318389 DOI: 10.3390/nano12142392] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023]
Abstract
Silicon dioxide (SiO2) is a mineral compound present in the Earth’s crust in two mineral forms: crystalline and amorphous. Based on epidemiological and/or biological evidence, the pulmonary effects of crystalline silica are considered well understood, with the development of silicosis, emphysema, chronic bronchitis, or chronic obstructive pulmonary disease. The structure and capacity to trigger oxidative stress are recognized as relevant determinants in crystalline silica’s toxicity. In contrast, natural amorphous silica was long considered nontoxic, and was often used as a negative control in experimental studies. However, as manufactured amorphous silica nanoparticles (or nanosilica or SiNP) are becoming widely used in industrial applications, these paradigms must now be reconsidered at the nanoscale (<100 nm). Indeed, recent experimental studies appear to point towards significant toxicity of manufactured amorphous silica nanoparticles similar to that of micrometric crystalline silica. In this article, we present an extensive review of the nontumoral pulmonary effects of silica based on in vitro and in vivo experimental studies. The findings of this review are presented both for micro- and nanoscale particles, but also based on the crystalline structure of the silica particles.
Collapse
|
11
|
Taucher E, Mykoliuk I, Lindenmann J, Smolle-Juettner FM. Implications of the Immune Landscape in COPD and Lung Cancer: Smoking Versus Other Causes. Front Immunol 2022; 13:846605. [PMID: 35386685 PMCID: PMC8978964 DOI: 10.3389/fimmu.2022.846605] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 12/30/2022] Open
Abstract
Cigarette smoking is reported in about one third of adults worldwide. A strong relationship between cigarette smoke exposure and chronic obstructive pulmonary disease (COPD) as well as lung cancer has been proven. However, about 15% of lung cancer cases, and between one fourth and one third of COPD cases, occur in never-smokers. The effects of cigarette smoke on the innate as well as the adaptive immune system have been widely investigated. It is assumed that certain immunologic features contribute to lung cancer and COPD development in the absence of smoking as the major risk factor. In this article, we review different immunological aspects of lung cancer and COPD with a special focus on non-smoking related risk factors.
Collapse
Affiliation(s)
- Elisabeth Taucher
- Division of Pulmonology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Iurii Mykoliuk
- Division of Thoracic Surgery, Department of Surgery, Medical University Graz, Graz, Austria
| | - Joerg Lindenmann
- Division of Thoracic Surgery, Department of Surgery, Medical University Graz, Graz, Austria
| | | |
Collapse
|
12
|
Moog MT, Hinze C, Bormann T, Aschenbrenner F, Knudsen L, DeLuca DS, Jonigk D, Neubert L, Welte T, Gauldie J, Kolb M, Maus UA. B Cells Are Not Involved in the Regulation of Adenoviral TGF-β1- or Bleomycin-Induced Lung Fibrosis in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1259-1271. [PMID: 35149532 DOI: 10.4049/jimmunol.2100767] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is an irreversible, age-related diffuse parenchymal lung disease of poorly defined etiology. Many patients with IPF demonstrate distinctive lymphocytic interstitial infiltrations within remodeled lung tissue with uncertain pathogenetic relevance. Histopathological examination of explant lung tissue of patients with IPF revealed accentuated lymphoplasmacellular accumulations in close vicinity to, or even infiltrating, remodeled lung tissue. Similarly, we found significant accumulations of B cells interfused with T cells within remodeled lung tissue in two murine models of adenoviral TGF-β1 or bleomycin (BLM)-induced lung fibrosis. Such B cell accumulations coincided with significantly increased lung collagen deposition, lung histopathology, and worsened lung function in wild-type (WT) mice. Surprisingly, B cell-deficient µMT knockout mice exhibited similar lung tissue remodeling and worsened lung function upon either AdTGF-β1 or BLM as for WT mice. Comparative transcriptomic profiling of sorted B cells collected from lungs of AdTGF-β1- and BLM-exposed WT mice identified a large set of commonly regulated genes, but with significant enrichment observed for Gene Ontology terms apparently not related to lung fibrogenesis. Collectively, although we observed B cell accumulations in lungs of IPF patients as well as two experimental models of lung fibrosis, comparative profiling of characteristic features of lung fibrosis between WT and B cell-deficient mice did not support a major involvement of B cells in lung fibrogenesis in mice.
Collapse
Affiliation(s)
- Marie T Moog
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | - Christopher Hinze
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | - Tina Bormann
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany
| | | | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - David S DeLuca
- German Center for Lung Research, partner site Biomedical Research in Endstage and Obstructive Lung Disease Hanover, Hannover, Germany
| | - Danny Jonigk
- German Center for Lung Research, partner site Biomedical Research in Endstage and Obstructive Lung Disease Hanover, Hannover, Germany
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Lavinia Neubert
- German Center for Lung Research, partner site Biomedical Research in Endstage and Obstructive Lung Disease Hanover, Hannover, Germany
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Tobias Welte
- German Center for Lung Research, partner site Biomedical Research in Endstage and Obstructive Lung Disease Hanover, Hannover, Germany
- Clinic for Pneumology, Hannover Medical School, Hannover, Germany; and
| | - Jack Gauldie
- Department of Medicine, Pathology, and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Martin Kolb
- Department of Medicine, Pathology, and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ulrich A Maus
- Division of Experimental Pneumology, Hannover Medical School, Hannover, Germany;
- German Center for Lung Research, partner site Biomedical Research in Endstage and Obstructive Lung Disease Hanover, Hannover, Germany
| |
Collapse
|
13
|
Wang J, Hu K, Cai X, Yang B, He Q, Wang J, Weng Q. Targeting PI3K/AKT signaling for treatment of idiopathic pulmonary fibrosis. Acta Pharm Sin B 2022; 12:18-32. [PMID: 35127370 PMCID: PMC8799876 DOI: 10.1016/j.apsb.2021.07.023] [Citation(s) in RCA: 230] [Impact Index Per Article: 76.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/13/2021] [Accepted: 07/09/2021] [Indexed: 01/03/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive fibrotic interstitial pneumonia with unknown causes. The incidence rate increases year by year and the prognosis is poor without cure. Recently, phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB/AKT) signaling pathway can be considered as a master regulator for IPF. The contribution of the PI3K/AKT in fibrotic processes is increasingly prominent, with PI3K/AKT inhibitors currently under clinical evaluation in IPF. Therefore, PI3K/AKT represents a critical signaling node during fibrogenesis with potential implications for the development of novel anti-fibrotic strategies. This review epitomizes the progress that is being made in understanding the complex interpretation of the cause of IPF, and demonstrates that PI3K/AKT can directly participate to the greatest extent in the formation of IPF or cooperate with other pathways to promote the development of fibrosis. We further summarize promising PI3K/AKT inhibitors with IPF treatment benefits, including inhibitors in clinical trials and pre-clinical studies and natural products, and discuss how these inhibitors mitigate fibrotic progression to explore possible potential agents, which will help to develop effective treatment strategies for IPF in the near future.
Collapse
Affiliation(s)
- Jincheng Wang
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Kaili Hu
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xuanyan Cai
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Wang
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
14
|
Deleterious Role of Th9 Cells in Pulmonary Fibrosis. Cells 2021; 10:cells10113209. [PMID: 34831433 PMCID: PMC8621886 DOI: 10.3390/cells10113209] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 12/05/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease of unknown etiology. Immune disorders play an important role in IPF pathogenesis. Here, we show that Th9 cells differentiate and activate in the lung tissue of patients with IPF and bleomycin (BLM)-induced lung fibrosis mice. Moreover, we found that Th9 cells promote pulmonary fibrosis in two ways. On the one hand, Th9 cells promote fibroblast differentiation, activation, and collagen secretion by secreting IL-9. On the other hand, they promote differentiation of Th0 cells into Th2 cells by secreting IL-4. Th9 cells and Th2 cells can promote each other, accelerating the Th1/Th2 imbalance and eventually forming a positive feedback of pulmonary fibrosis. In addition, we found that neutralizing IL-9 in both preventive and therapeutic settings ameliorates bleomycin-induced pulmonary fibrosis. Furthermore, we identified several critical signaling pathways involved in the effect of neutralizing IL-9 on pulmonary fibrosis by proteomics study. From an immunological perspective, we elucidated the novel role and underlying mechanism of Th9 cells in pulmonary fibrosis. Our study suggested that Th9-based immunotherapy may be employed as a treatment strategy for IPF.
Collapse
|
15
|
Kim MG, Yun D, Kang CL, Hong M, Hwang J, Moon KC, Jeong CW, Kwak C, Kim DK, Oh KH, Joo KW, Kim YS, Lee DS, Han SS. Kidney VISTA prevents IFNγ-IL-9 axis-mediated tubulointerstitial fibrosis after acute glomerular injury. J Clin Invest 2021; 132:151189. [PMID: 34752423 PMCID: PMC8718152 DOI: 10.1172/jci151189] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 11/05/2021] [Indexed: 11/17/2022] Open
Abstract
Severe glomerular injury ultimately leads to tubulointerstitial fibrosis that determines patient outcome, but the immunological molecules connecting these processes remain undetermined. The present study addressed whether V-domain Ig suppressor of T cell activation (VISTA), constitutively expressed in kidney macrophages, plays a protective role in tubulointerstitial fibrotic transformation after acute antibody-mediated glomerulonephritis. After acute glomerular injury using nephrotoxic serum, tubules in the VISTA-deficient (Vsir–/–) kidney suffered more damage than those in WT kidneys. When interstitial immune cells were examined, the contact frequency of macrophages with infiltrated T cells increased and the immunometabolic features of T cells changed to showing high oxidative phosphorylation and fatty acid metabolism and overproduction of IFN-γ. The Vsir–/– parenchymal tissue cells responded to this altered milieu of interstitial immune cells as more IL-9 was produced, which augmented tubulointerstitial fibrosis. Blocking antibodies against IFN-γ and IL-9 protected the above pathological process in VISTA-depleted conditions. In human samples with acute glomerular injury (e.g., antineutrophil cytoplasmic autoantibody vasculitis), high VISTA expression in tubulointerstitial immune cells was associated with low tubulointerstitial fibrosis and good prognosis. Therefore, VISTA is a sentinel protein expressed in kidney macrophages that prevents tubulointerstitial fibrosis via the IFN-γ/IL-9 axis after acute antibody-mediated glomerular injury.
Collapse
Affiliation(s)
- Min-Gang Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Donghwan Yun
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Chae Lin Kang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Minki Hong
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Juhyeon Hwang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Kyung Chul Moon
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Chang Wook Jeong
- Department of Urology, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Cheol Kwak
- Department of Urology, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea, Republic of
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea, Republic of
| |
Collapse
|
16
|
Arai T, Matsuoka H, Hirose M, Kida H, Yamamoto S, Ogata Y, Mori M, Hatsuda K, Sugimoto C, Tachibana K, Akira M, Inoue Y. Prognostic significance of serum cytokines during acute exacerbation of idiopathic interstitial pneumonias treated with thrombomodulin. BMJ Open Respir Res 2021; 8:e000889. [PMID: 34326155 PMCID: PMC8323382 DOI: 10.1136/bmjresp-2021-000889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/11/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Acute exacerbation (AE) has been reported to herald a poor prognosis in idiopathic pulmonary fibrosis and is now thought to do so in idiopathic interstitial pneumonias (IIPs). However, the pathophysiology of AE-IIPs is not sufficiently understood. In our previously reported SETUP trial, we found better survival in patients with AE-IIPs treated with corticosteroids and thrombomodulin than in those treated with corticosteroids alone. In that study, we collected serum samples to evaluate changes in cytokine levels and retrospectively examined the prognostic significance and pathophysiological role of serum cytokines in patients with AE-IIPs. METHODS This study included 28 patients from the SETUP trial for whom serial serum samples had been prospectively obtained. AE-IIPs were diagnosed using the Japanese Respiratory Society criteria. All patients were treated with intravenous thrombomodulin and corticosteroids from 2014 to 2016. Serum levels of 27 cytokines were measured using Bio-Plex. The high-resolution CT pattern at the time of diagnosis of AE was classified as diffuse or non-diffuse. RESULTS Univariate analysis revealed that higher serum levels of interleukin (IL)-2, IL-7, IL-9, IL-12, IL13, basic fibroblast growth factor, granulocyte-macrophage colony-stimulating factor, interferon-γ inducible protein-10, platelet-derived growth factor and regulated on activation, normal T cell expressed and secreted (RANTES) at AE were significant predictors of 90-day survival. The HRCT pattern was also a significant clinical predictor of 90-day survival. Multivariate analysis with stepwise selection identified a higher serum RANTES level at AE to be a significant predictor of 90-day survival, including after adjustment for HRCT pattern. Multivariate analysis with stepwise selection suggested that a marked increase in the serum IL-10 level on day 8 could predict 90-day mortality. CONCLUSIONS A higher serum RANTES level at AE the time of diagnosis predicted a good survival outcome, and an elevated serum IL-10 level on day 8 predicted a poor survival outcome. TRIAL REGISTRATION NUMBER UMIN000014969.
Collapse
Affiliation(s)
- Toru Arai
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai City, Japan
| | - Hiroto Matsuoka
- Department of Respiratory Medicine, Osaka Prefectural Hospital Organization Osaka Habikino Medical Center, Habikino City, Japan
| | - Masaki Hirose
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai City, Japan
| | - Hiroshi Kida
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka City, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita City, Japan
| | - Suguru Yamamoto
- Department of Respiratory Medicine, National Hospital Organization Osaka Minami Medical Center, Kawachinagano City, Japan
| | - Yoshitaka Ogata
- Department of Critical Care Medicine, Yao Tokushukai Hospital, Yao City, Japan
| | - Masahide Mori
- Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka City, Japan
| | - Kazuyoshi Hatsuda
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai City, Japan
| | - Chikatoshi Sugimoto
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai City, Japan
| | - Kazunobu Tachibana
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai City, Japan
| | - Masanori Akira
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai City, Japan
| | - Yoshikazu Inoue
- Clinical Research Center, National Hospital Organization Kinki-Chuo Chest Medical Center, Sakai City, Japan
| |
Collapse
|
17
|
Gouyou B, Grün K, Kerschenmeyer A, Villa A, Matasci M, Schrepper A, Pfeil A, Bäz L, Jung C, Schulze PC, Neri D, Franz M. Therapeutic Evaluation of Antibody-Based Targeted Delivery of Interleukin 9 in Experimental Pulmonary Hypertension. Int J Mol Sci 2021; 22:ijms22073460. [PMID: 33801620 PMCID: PMC8037792 DOI: 10.3390/ijms22073460] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/22/2022] Open
Abstract
Background and Aims: Pulmonary hypertension (PH) is a heterogeneous disorder associated with poor prognosis. For the majority of patients, only limited therapeutic options are available. Thus, there is great interest to develop novel treatment strategies focusing on pulmonary vascular and right ventricular remodeling. Interleukin 9 (IL9) is a pleiotropic cytokine with pro- and anti-inflammatory functions. The aim of this study was to evaluate the therapeutic activity of F8IL9F8 consisting of IL9 fused to the F8 antibody, specific to the alternatively-spliced EDA domain of fibronectin, which is abundantly expressed in pulmonary vasculature and right ventricular myocardium in PH. Methods: The efficacy of F8IL9F8 in attenuating PH progression in the monocrotaline mouse model was evaluated in comparison to an endothelin receptor antagonist (ERA) or an IL9 based immunocytokine with irrelevant antibody specificity (KSFIL9KSF). Treatment effects were assessed by right heart catheterization, echocardiography as well as histological and immunohistochemical tissue analyses. Results: Compared to controls, systolic right ventricular pressure (RVPsys) was significantly elevated and a variety of right ventricular echocardiographic parameters were significantly impaired in all MCT-induced PH groups except for the F8IL9F8 group. Both, F8IL9F8 and ERA treatments lead to a significant reduction in RVPsys and an improvement of echocardiographic parameters when compared to the MCT group not observable for the KSFIL9KSF group. Only F8IL9F8 significantly reduced lung tissue damage and displayed a significant decrease of leukocyte and macrophage accumulation in the lungs and right ventricles. Conclusions: Our study provides first pre-clinical evidence for the use of F8IL9F8 as a new therapeutic agent for PH in terms of a disease-modifying concept addressing cardiovascular remodeling.
Collapse
Affiliation(s)
- Baptiste Gouyou
- Philochem AG, CH-8112 Otelfingen, Switzerland; (B.G.); (A.K.); (A.V.); (M.M.); (D.N.)
| | - Katja Grün
- Department of Internal Medicine I, Univerisity Hospital Jena, 07747 Jena, Germany; (K.G.); (L.B.); (P.C.S.)
| | - Anne Kerschenmeyer
- Philochem AG, CH-8112 Otelfingen, Switzerland; (B.G.); (A.K.); (A.V.); (M.M.); (D.N.)
| | - Alessandra Villa
- Philochem AG, CH-8112 Otelfingen, Switzerland; (B.G.); (A.K.); (A.V.); (M.M.); (D.N.)
| | - Mattia Matasci
- Philochem AG, CH-8112 Otelfingen, Switzerland; (B.G.); (A.K.); (A.V.); (M.M.); (D.N.)
| | - Andrea Schrepper
- Department of Cardiothoracic Surgery, Univerisity Hospital Jena, 07747 Jena, Germany;
| | - Alexander Pfeil
- Department of Internal Medicine III, Univerisity Hospital Jena, 07747 Jena, Germany;
| | - Laura Bäz
- Department of Internal Medicine I, Univerisity Hospital Jena, 07747 Jena, Germany; (K.G.); (L.B.); (P.C.S.)
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - P. Christian Schulze
- Department of Internal Medicine I, Univerisity Hospital Jena, 07747 Jena, Germany; (K.G.); (L.B.); (P.C.S.)
| | - Dario Neri
- Philochem AG, CH-8112 Otelfingen, Switzerland; (B.G.); (A.K.); (A.V.); (M.M.); (D.N.)
| | - Marcus Franz
- Department of Internal Medicine I, Univerisity Hospital Jena, 07747 Jena, Germany; (K.G.); (L.B.); (P.C.S.)
- Correspondence: ; Tel.: +49-3641-9324127
| |
Collapse
|
18
|
Ma Q. Polarization of Immune Cells in the Pathologic Response to Inhaled Particulates. Front Immunol 2020; 11:1060. [PMID: 32625201 PMCID: PMC7311785 DOI: 10.3389/fimmu.2020.01060] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/01/2020] [Indexed: 12/30/2022] Open
Abstract
Polarization of immune cells is commonly observed in host responses associated with microbial immunity, inflammation, tumorigenesis, and tissue repair and fibrosis. In this process, immune cells adopt distinct programs and perform specialized functions in response to specific signals. Accumulating evidence indicates that inhalation of micro- and nano-sized particulates activates barrier immune programs in the lung in a time- and context-dependent manner, including type 1 and type 2 inflammation, and T helper (Th) 17 cell, regulatory T cell (Treg), innate lymphoid cell (ILC), and myeloid-derived suppressor cell (MDSC) responses, which highlight the polarization of several major immune cell types. These responses facilitate the pulmonary clearance and repair under physiological conditions. When exposure persists and overwhelms the clearance capacity, they foster the chronic progression of inflammation and development of progressive disease conditions, such as fibrosis and cancer. The pulmonary response to insoluble particulates thus represents a distinctive disease process wherein non-infectious, persistent exposures stimulate the polarization of immune cells to orchestrate dynamic inflammatory and immune reactions, leading to pulmonary and pleural chronic inflammation, fibrosis, and malignancy. Despite large variations in particles and their associated disease outcomes, the early response to inhaled particles often follows a common path. The initial reactions entail a barrier immune response dominated by type 1 inflammation that features active phagocytosis by M1 macrophages and recruitment of neutrophils, both of which are fueled by Th1 and proinflammatory cytokines. Acute inflammation is immediately followed by resolution and tissue repair mediated through specialized pro-resolving mediators (SPMs) and type 2 cytokines and cells including M2 macrophages and Th2 lymphocytes. As many particles and fibers cannot be digested by phagocytes, resolution is often extended and incomplete, and type 2 inflammation becomes heightened, which promotes interstitial fibrosis, granuloma formation, and tumorigenesis. Recent studies also reveal the involvement of Th17-, Treg-, ILC-, and MDSC-mediated responses in the pathogenesis caused by inhaled particulates. This review synopsizes the progress in understanding the interplay between inhaled particles and the pulmonary immune functions in disease pathogenesis, with focus on particle-induced polarization of immune cells and its role in the development of chronic inflammation, fibrosis, and cancer in the lung.
Collapse
Affiliation(s)
- Qiang Ma
- Receptor Biology Laboratory, Toxicology and Molecular Biology Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| |
Collapse
|
19
|
In Vitro and In Vivo Toxicity Studies on Cymbopogon giganteus Chiov. Leaves Essential Oil from Benin. J Toxicol 2020; 2020:8261058. [PMID: 32399030 PMCID: PMC7204255 DOI: 10.1155/2020/8261058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/14/2019] [Accepted: 10/24/2019] [Indexed: 12/17/2022] Open
Abstract
Cymbopogon giganteus Chiov. (Poaceae) is a medicinal plant used to treat various diseases in traditional medicine in several African countries. The present study aims to evaluate the oral and inhalation toxicity as well as the mutagenic effects of the essential oil of Cymbopogon giganteus leaves (EOCG) from a sample collected in Benin. Mutagenic potential was assessed by the Ames test using Salmonella typhimurium strains TA98 and TA100. Oral acute toxicity was carried out by administration of a single dose of 2000 mg/kg b.w. to Wistar rats while oral subacute toxicity was assessed by daily administration of 50 and 500 mg/kg of EOCG for 28 days. Finally, inhalation toxicity was assessed by administration of a single dose of 0.125%, 0.5%, 2% or 5% v/v of EOCG emulsions in 0.05% v/v lecithin solution in sterile water for the first experiment, and in a second one by administration of single dose of 0.125% or 0.5% v/v. A broncho-alveolar lavage was performed after 3 h or 24 h, respectively. The results show that EOCG is not mutagenic on Salmonella typhimurium strains at the highest concentration tested (200 μg/plate). In the acute oral toxicity study, EOCG induce neither mortality nor toxicity, showing that the LD50 is greater than 2000 mg/kg. The subacute oral toxicity study at both doses did not show any significant difference in body weight, relative organ weight, hematological and/or biochemical parameters or histopathology as compared to the control group. EOCG induced mortality and inflammation in lungs 3 h after administration of a single dose of 5% or 2% v/v. Single doses of 0.125% or 0.5% v/v did not induce inflammation, cell recruitment nor cytotoxicity in lungs 3 h or 24 h after administration, suggesting safety at these concentrations. This first report on the in vivo toxicity will be useful to guide safe uses of EOCG.
Collapse
|
20
|
Sugimoto N, Suzukawa M, Nagase H, Koizumi Y, Ro S, Kobayashi K, Yoshihara H, Kojima Y, Kamiyama-Hara A, Hebisawa A, Ohta K. IL-9 Blockade Suppresses Silica-induced Lung Inflammation and Fibrosis in Mice. Am J Respir Cell Mol Biol 2019; 60:232-243. [PMID: 30240278 DOI: 10.1165/rcmb.2017-0287oc] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Recapitulative animal models of idiopathic pulmonary fibrosis (IPF) and related diseases are lacking, which inhibits our ability to fully clarify the pathogenesis of these diseases. Although lung fibrosis in mouse models is often induced by bleomycin, silica-induced lung fibrosis is more sustainable and more progressive. Therefore, in this study, we sought to elucidate the mediator(s) responsible for the pathogenesis of lung fibrosis, through the use of a mouse model of silica-induced lung fibrosis. With a single nasal administration of 16 mg of silica, lung inflammation (assessed by elevated cellular components in the BAL fluids [BALFs]) and lung fibrosis (assessed by lung histology and lung hydroxyproline levels) were induced and sustained for as long as 24 weeks. Of the mediators measured in the BALFs, IL-9 was characteristically elevated gradually, and peaked at 24 weeks after silica administration. Treatment of silica-challenged mice with anti-IL-9-neutralizing antibody inhibited lung fibrosis, as assessed by lung hydroxyproline level, and suppressed the levels of major mediators, including IL-1β, IL-6, IL-12, CCL2, CXCL1, and TNF-α in BALFs. Moreover, human lung specimens from patients with IPF have shown high expression of IL-9 in alveolar macrophages, CD4-positive cells, and receptors for IL-9 in airway epithelial cells. Collectively, these data suggest that IL-9 plays an important role in the pathogenesis of lung fibrosis in diseases such as IPF.
Collapse
Affiliation(s)
- Naoya Sugimoto
- 1 Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan; and
| | - Maho Suzukawa
- 2 National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - Hiroyuki Nagase
- 1 Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan; and
| | - Yuta Koizumi
- 1 Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan; and
| | - Shoki Ro
- 1 Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan; and
| | - Konomi Kobayashi
- 1 Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan; and
| | - Hisanao Yoshihara
- 1 Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan; and
| | - Yasuhiro Kojima
- 1 Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan; and
| | - Asae Kamiyama-Hara
- 1 Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan; and
| | - Akira Hebisawa
- 2 National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| | - Ken Ohta
- 2 National Hospital Organization Tokyo National Hospital, Tokyo, Japan
| |
Collapse
|
21
|
Sironval V, Palmai-Pallag M, Vanbever R, Huaux F, Mejia J, Lucas S, Lison D, van den Brule S. HIF-1α is a key mediator of the lung inflammatory potential of lithium-ion battery particles. Part Fibre Toxicol 2019; 16:35. [PMID: 31533843 PMCID: PMC6751682 DOI: 10.1186/s12989-019-0319-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/29/2019] [Indexed: 01/30/2023] Open
Abstract
Background Li-ion batteries (LIB) are increasingly used worldwide. They are made of low solubility micrometric particles, implying a potential for inhalation toxicity in occupational settings and possibly for consumers. LiCoO2 (LCO), one of the most used cathode material, induces inflammatory and fibrotic lung responses in mice. LCO also stabilizes hypoxia-inducible factor (HIF) -1α, a factor implicated in inflammation, fibrosis and carcinogenicity. Here, we investigated the role of cobalt, nickel and HIF-1α as determinants of toxicity, and evaluated their predictive value for the lung toxicity of LIB particles in in vitro assays. Results By testing a set of 5 selected LIB particles (LCO, LiNiMnCoO2, LiNiCoAlO2) with different cobalt and nickel contents, we found a positive correlation between their in vivo lung inflammatory activity, and (i) Co and Ni particle content and their bioaccessibility and (ii) the stabilization of HIF-1α in the lung. Inhibition of HIF-1α with chetomin or PX-478 blunted the lung inflammatory response to LCO in mice. In IL-1β deficient mice, HIF-1α was the upstream signal of the inflammatory lung response to LCO. In vitro, the level of HIF-1α stabilization induced by LIB particles in BEAS-2B cells correlated with the intensity of lung inflammation induced by the same particles in vivo. Conclusions We conclude that HIF-1α, stabilized in lung cells by released Co and Ni ions, is a mechanism-based biomarker of lung inflammatory responses induced by LIB particles containing Co/Ni. Documenting the Co/Ni content of LIB particles, their bioaccessibility and their capacity to stabilize HIF-1α in vitro can be used to predict the lung inflammatory potential of LIB particles. Electronic supplementary material The online version of this article (10.1186/s12989-019-0319-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Violaine Sironval
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 57 - bte B1.57.06, 1200, Brussels, Belgium.
| | - Mihaly Palmai-Pallag
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 57 - bte B1.57.06, 1200, Brussels, Belgium
| | - Rita Vanbever
- Louvain Drug Research Institute, Université catholique de Louvain, Avenue Mounier 73 - bte B1.73.12, 1200, Brussels, Belgium
| | - François Huaux
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 57 - bte B1.57.06, 1200, Brussels, Belgium
| | - Jorge Mejia
- Research Centre for the Physics of Matter and Radiation (PMR-LARN), NARILIS, Université de Namur, rue de Bruxelles 61, 5000, Namur, Belgium
| | - Stéphane Lucas
- Research Centre for the Physics of Matter and Radiation (PMR-LARN), NARILIS, Université de Namur, rue de Bruxelles 61, 5000, Namur, Belgium
| | - Dominique Lison
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 57 - bte B1.57.06, 1200, Brussels, Belgium
| | - Sybille van den Brule
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 57 - bte B1.57.06, 1200, Brussels, Belgium
| |
Collapse
|
22
|
Xylourgidis N, Min K, Ahangari F, Yu G, Herazo-Maya JD, Karampitsakos T, Aidinis V, Binzenhöfer L, Bouros D, Bennett AM, Kaminski N, Tzouvelekis A. Role of dual-specificity protein phosphatase DUSP10/MKP-5 in pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2019; 317:L678-L689. [PMID: 31483681 DOI: 10.1152/ajplung.00264.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Mitogen-activated protein kinase (MAPK) phosphatase 5 (MKP-5) is a member of the dual-specificity family of protein tyrosine phosphatases that negatively regulates p38 MAPK and the JNK. MKP-5-deficient mice exhibit improved muscle repair and reduced fibrosis in an animal model of muscular dystrophy. Here, we asked whether the effects of MKP-5 on muscle fibrosis extend to other tissues. Using a bleomycin-induced model of pulmonary fibrosis, we found that MKP-5-deficient mice were protected from the development of lung fibrosis, expressed reduced levels of hydroxyproline and fibrogenic genes, and displayed marked polarization towards an M1-macrophage phenotype. We showed that the profibrogenic effects of the transforming growth factor-β1 (TGF-β1) were inhibited in MKP-5-deficient lung fibroblasts. MKP-5-deficient fibroblasts exhibited enhanced p38 MAPK activity, impaired Smad3 phosphorylation, increased Smad7 levels, and decreased expression of fibrogenic genes. Myofibroblast differentiation was attenuated in MKP-5-deficient fibroblasts. Finally, we found that MKP-5 expression was increased in idiopathic pulmonary fibrosis (IPF)-derived lung fibroblasts but not in whole IPF lungs. These data suggest that MKP-5 plays an essential role in promoting lung fibrosis. Our results couple MKP-5 with the TGF-β1 signaling machinery and imply that MKP-5 inhibition may serve as a therapeutic target for human lung fibrosis.
Collapse
Affiliation(s)
- Nikos Xylourgidis
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Kisuk Min
- Department of Kinesiology, University of Texas, El Paso, Texas
| | - Farida Ahangari
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Guoying Yu
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Jose D Herazo-Maya
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut
| | | | - Vassilis Aidinis
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming," Athens, Greece
| | - Leonhard Binzenhöfer
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Demosthenes Bouros
- First Academic Department of Pneumonology, University of Athens, Athens, Greece
| | - Anton M Bennett
- Department of Pharmacology, Yale School of Medicine, New Haven, Connecticut
| | - Naftali Kaminski
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Argyrios Tzouvelekis
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut.,Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming," Athens, Greece
| |
Collapse
|
23
|
He J, Wang L, Zhang C, Shen W, Zhang Y, Liu T, Hu H, Xie X, Luo F. Interleukin-9 promotes tumorigenesis through augmenting angiogenesis in non-small cell lung cancer. Int Immunopharmacol 2019; 75:105766. [PMID: 31352324 DOI: 10.1016/j.intimp.2019.105766] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 06/23/2019] [Accepted: 07/15/2019] [Indexed: 02/05/2023]
Abstract
IL-9 is a proinflammatory cytokine that plays a critical role in autoimmunity and inflammatory diseases. However, its role in tumorigenesis has not been well studied. In this study, we found that IL-9 expression was significantly increased and associated with poor progression in human non-small cell lung cancer (NSCLC). Ectopic expression of IL-9 in NSCLC cells did not affect cell proliferation and apoptosis in vitro, but markedly promoted tumor growth in vivo. Immune-profile analysis showed no significant changes in the frequencies of infiltrated immune cells in the tumor site, neither in nude mice nor in immune-competent mice. However, we found that VEGF and microvessel density (MVD) were significantly increased in xenografts. IL-9 could promote cell growth and tube formation of HUVEC cells in vitro. In addition, correlation analysis implied a significant positive relationship between the density of IL-9 and VEGF, as well as MVD in human NSCLC tissues. Finally, we found that IL-9 stimulated tumor angiogenesis via STAT3 signaling. Together, our findings demonstrate a promoting role of IL-9 in lung cancer development, probably through promoting tumor angiogenesis. IL-9 thus may represent a new prognostic marker and therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Jun He
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China; Department of Oncology, The Third Hospital of Mianyang (Sichuan Mental Health Center), Mianyang, Sichuan 621000, China
| | - Li Wang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Chengda Zhang
- Department of Gastroenterology, The Third Hospital of MianYang (Sichuan Mental Health Center), Mianyang, Sichuan 621000, China
| | - Wenbin Shen
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Yong Zhang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Tao Liu
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610000, China
| | - Haoyue Hu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Xiaoxiao Xie
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China
| | - Feng Luo
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610000, China.
| |
Collapse
|
24
|
Elevation of IL-6 and IL-33 Levels in Serum Associated with Lung Fibrosis and Skeletal Muscle Wasting in a Bleomycin-Induced Lung Injury Mouse Model. Mediators Inflamm 2019; 2019:7947596. [PMID: 31049028 PMCID: PMC6458868 DOI: 10.1155/2019/7947596] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/15/2019] [Accepted: 01/31/2019] [Indexed: 02/07/2023] Open
Abstract
Weight loss due to skeletal muscle atrophy in patients with chronic pulmonary disease is negatively correlated with clinical outcome. Pulmonary fibrosis is a chronic and progressive interstitial lung disease characterized by the dysregulated deposition of the extracellular matrix (ECM) with the destruction of normal tissue, resulting in end-stage organ failure. BLM-induced fibrosis is one of several different experimental models of pulmonary fibrosis, characterized by inflammation and excessive ECM deposition. We directly induced mouse lung injury by the intratracheal administration of bleomycin and monitored the physiological and biochemical changes in lung and skeletal muscle tissues by using lung function testing, ELISA, Western blotting, and immunohistochemistry. Here, we found that BLM-induced lung fibrosis with thickened interstitial lung tissue, including fibronectin and collagen, was correlated with the increased serum concentrations of IL-6 and IL-33 and accompanied by reduced lung function, including FRC (functional residual capacity), C chord (lung compliance), IC (inspiratory capacity), VC (vital capacity), TLC (total lung capacity), and FVC (forced vital capacity) (p < 0.05). The activity of AKT in lung tissue was suppressed, but conversely, the activity of STAT3 was enhanced during lung fibrosis in mice. In addition, we found that the amount of sST2, the soluble form of the IL-33 receptor, was dramatically decreased in lung fibrosis tissues. The skeletal muscle tissue isolated from lung injury mice increased the activation of STAT3 and AMPK, accompanied by an increased amount of Atrogin-1 protein in BLM-induced lung fibrosis mice. The mouse myoblast cell-based model showed that IL-6 and IL-33 specifically activated STAT3 and AMPK signaling, respectively, to induce the expression of the muscle-specific proteolysis markers MuRF1 and Atrogin-1. These data suggested that increased levels of IL-6 and IL-33 in the serum of mice with BLM-induced lung injury may cause lung fibrosis with thickened interstitial lung tissue accompanied by reduced lung function and muscle mass through the activation of STAT3 and AMPK signals.
Collapse
|
25
|
Heukels P, Moor C, von der Thüsen J, Wijsenbeek M, Kool M. Inflammation and immunity in IPF pathogenesis and treatment. Respir Med 2019; 147:79-91. [DOI: 10.1016/j.rmed.2018.12.015] [Citation(s) in RCA: 320] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 11/21/2018] [Accepted: 12/29/2018] [Indexed: 12/11/2022]
|
26
|
Rojas-Quintero J, Wang X, Owen CA. Dusting Off IL-9 as a New Therapeutic Target for Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2019; 60:141-143. [PMID: 30281328 PMCID: PMC6376410 DOI: 10.1165/rcmb.2018-0319ed] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Joselyn Rojas-Quintero
- 1 Division of Pulmonary and Critical Care Medicine Brigham and Women's Hospital and Harvard Medical School Boston, Massachusetts
| | - Xiaoyun Wang
- 1 Division of Pulmonary and Critical Care Medicine Brigham and Women's Hospital and Harvard Medical School Boston, Massachusetts
| | - Caroline A Owen
- 1 Division of Pulmonary and Critical Care Medicine Brigham and Women's Hospital and Harvard Medical School Boston, Massachusetts
| |
Collapse
|
27
|
Sironval V, Reylandt L, Chaurand P, Ibouraadaten S, Palmai-Pallag M, Yakoub Y, Ucakar B, Rose J, Poleunis C, Vanbever R, Marbaix E, Lison D, van den Brule S. Respiratory hazard of Li-ion battery components: elective toxicity of lithium cobalt oxide (LiCoO 2) particles in a mouse bioassay. Arch Toxicol 2018; 92:1673-1684. [PMID: 29550861 DOI: 10.1007/s00204-018-2188-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
Rechargeable Li-ion batteries (LIB) are increasingly produced and used worldwide. LIB electrodes are made of micrometric and low solubility particles, consisting of toxicologically relevant elements. The health hazard of these materials is not known. Here, we investigated the respiratory hazard of three leading LIB components (LiFePO4 or LFP, Li4Ti5O12 or LTO, and LiCoO2 or LCO) and their mechanisms of action. Particles were characterized physico-chemically and elemental bioaccessibility was documented. Lung inflammation and fibrotic responses, as well as particle persistence and ion bioavailability, were assessed in mice after aspiration of LIB particles (0.5 or 2 mg); crystalline silica (2 mg) was used as reference. Acute inflammatory lung responses were recorded with the 3 LIB particles and silica, LCO being the most potent. Inflammation persisted 2 m after LFP, LCO and silica, in association with fibrosis in LCO and silica lungs. LIB particles persisted in the lungs after 2 m. Endogenous iron co-localized with cobalt in LCO lungs, indicating the formation of ferruginous bodies. Fe and Co ions were detected in the broncho-alveolar lavage fluids of LFP and LCO lungs, respectively. Hypoxia-inducible factor (HIF) -1α, a marker of fibrosis and of the biological activity of Co ions, was upregulated in LCO and silica lungs. This study identified, for the first time, the respiratory hazard of LIB particles. LCO was at least as potent as crystalline silica to induce lung inflammation and fibrosis. Iron and cobalt, but not lithium, ions appear to contribute to LFP and LCO toxicity, respectively.
Collapse
Affiliation(s)
- Violaine Sironval
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue E. Mounier 52, bte B1.52.12, 1200, Brussels, Belgium.
| | - Laurence Reylandt
- Institute of Mechanics, Materials and Civil Engineering, Université catholique de Louvain, Place Sainte Barbe, 2, bte L5.02.02, 1348, Louvain-la-Neuve, Belgium
| | - Perrine Chaurand
- CEREGE, Aix Marseille Université, CNRS, IRD, Collège de France, Avenue Louis Philibert, 13090, Aix en Provence, France
| | - Saloua Ibouraadaten
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue E. Mounier 52, bte B1.52.12, 1200, Brussels, Belgium
| | - Mihaly Palmai-Pallag
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue E. Mounier 52, bte B1.52.12, 1200, Brussels, Belgium
| | - Yousof Yakoub
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue E. Mounier 52, bte B1.52.12, 1200, Brussels, Belgium
| | - Bernard Ucakar
- Louvain Drug Research Institute, Université catholique de Louvain, Avenue Mounier 73, bte B1.73.12, 1200, Brussels, Belgium
| | - Jérôme Rose
- CEREGE, Aix Marseille Université, CNRS, IRD, Collège de France, Avenue Louis Philibert, 13090, Aix en Provence, France
| | - Claude Poleunis
- Institute of Condensed Matter and Nanosciences, Université catholique de Louvain, Place Louis Pasteur 1, bte L4.01.10, 1348, Louvain-la-Neuve, Belgium
| | - Rita Vanbever
- Louvain Drug Research Institute, Université catholique de Louvain, Avenue Mounier 73, bte B1.73.12, 1200, Brussels, Belgium
| | - Etienne Marbaix
- De Duve Institute, Université catholique de Louvain, Avenue Hippocrate 75, bte B1.75.02, 1200, Brussels, Belgium
| | - Dominique Lison
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue E. Mounier 52, bte B1.52.12, 1200, Brussels, Belgium
| | - Sybille van den Brule
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue E. Mounier 52, bte B1.52.12, 1200, Brussels, Belgium
| |
Collapse
|
28
|
Macrophage polarization and function: new prospects for fibrotic disease. Immunol Cell Biol 2017; 95:864-869. [PMID: 29044201 DOI: 10.1038/icb.2017.64] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/26/2017] [Accepted: 07/27/2017] [Indexed: 02/08/2023]
|
29
|
Gong F, Pan YH, Huang X, Zhu HY, Jiang DL. From bench to bedside: Therapeutic potential of interleukin-9 in the treatment of asthma. Exp Ther Med 2017; 13:389-394. [PMID: 28352305 PMCID: PMC5347659 DOI: 10.3892/etm.2017.4024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/28/2016] [Indexed: 11/05/2022] Open
Abstract
Initially identified as a T cell and mast cell growth factor, interleukin (IL)-9 has long been recognized as an important mediator of asthma. Recently, accumulating results from transgenic mice demonstrated that systemic or lung-specific overexpression of IL-9 caused asthma-associated symptoms. Moreover, anti-mIL-9 antibody (Ab) blocking treatment alleviated disease in animal models of asthma. In light of the large quantity of data from the murine models, MEDI-528, a humanized anti-IL-9 monoclonal Ab has been produced to assess the activity of IL-9 on human asthma. In order to ascertain whether it is a successful translation from bench to bedside, the biological features of IL-9 were evaluated and up-to-date information regarding the role of IL-9 in different experimental murine models and human asthma were summarized.
Collapse
Affiliation(s)
- Fang Gong
- Department of Respiratory Medicine, The Third Hospital Affiliated to Nantong University, Wuxi, Jiangsu 214041, P.R. China
| | - Yu-Hong Pan
- Department of Respiratory Medicine, The Third Hospital Affiliated to Nantong University, Wuxi, Jiangsu 214041, P.R. China
| | - Xuan Huang
- Department of Respiratory Medicine, The Third Hospital Affiliated to Nantong University, Wuxi, Jiangsu 214041, P.R. China
| | - Hua-Yan Zhu
- Department of Respiratory Medicine, The Third Hospital Affiliated to Nantong University, Wuxi, Jiangsu 214041, P.R. China
| | - Dong-Lin Jiang
- Department of Respiratory Medicine, The Third Hospital Affiliated to Nantong University, Wuxi, Jiangsu 214041, P.R. China
| |
Collapse
|
30
|
Abstract
Since the discovery of IL-9 almost three decades back as a growth factor, we have come a long way to understand its pleiotropic functions in the immune system. Despite its many functions, IL-9 still remains as an understudied cytokine. In the last decade, renewed emphasis has been provided to understand the biology of IL-9. Any growth factor or cytokine signals via its cognate receptor to mediate biological functions. In this chapter, we discuss the IL-9 signal transduction in different cell types, which would then exert its distinct functions.
Collapse
Affiliation(s)
- Dijendra Nath Roy
- Department of Bioengineering, National Institute of Technology, NIT-Agartala, Jirania, 799046, Tripura, India
| | - Ritobrata Goswami
- School of Bio Science, Sir JC Bose Laboratory Complex, Indian Institute of Technology, Kharagpur, 721302, West Bengal, India.
| |
Collapse
|
31
|
Wirsdörfer F, Jendrossek V. The Role of Lymphocytes in Radiotherapy-Induced Adverse Late Effects in the Lung. Front Immunol 2016; 7:591. [PMID: 28018357 PMCID: PMC5155013 DOI: 10.3389/fimmu.2016.00591] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/29/2016] [Indexed: 12/31/2022] Open
Abstract
Radiation-induced pneumonitis and fibrosis are dose-limiting side effects of thoracic irradiation. Thoracic irradiation triggers acute and chronic environmental lung changes that are shaped by the damage response of resident cells, by the resulting reaction of the immune system, and by repair processes. Although considerable progress has been made during the last decade in defining involved effector cells and soluble mediators, the network of pathophysiological events and the cellular cross talk linking acute tissue damage to chronic inflammation and fibrosis still require further definition. Infiltration of cells from the innate and adaptive immune systems is a common response of normal tissues to ionizing radiation. Herein, lymphocytes represent a versatile and wide-ranged group of cells of the immune system that can react under specific conditions in various ways and participate in modulating the lung environment by adopting pro-inflammatory, anti-inflammatory, or even pro- or anti-fibrotic phenotypes. The present review provides an overview on published data about the role of lymphocytes in radiation-induced lung disease and related damage-associated pulmonary diseases with a focus on T lymphocytes and B lymphocytes. We also discuss the suspected dual role of specific lymphocyte subsets during the pneumonitic phase and fibrotic phase that is shaped by the environmental conditions as well as the interaction and the intercellular cross talk between cells from the innate and adaptive immune systems and (damaged) resident epithelial cells and stromal cells (e.g., endothelial cells, mesenchymal stem cells, and fibroblasts). Finally, we highlight potential therapeutic targets suited to counteract pathological lymphocyte responses to prevent or treat radiation-induced lung disease.
Collapse
Affiliation(s)
- Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen , Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen , Germany
| |
Collapse
|
32
|
Duru N, Wolfson B, Zhou Q. Mechanisms of the alternative activation of macrophages and non-coding RNAs in the development of radiation-induced lung fibrosis. World J Biol Chem 2016; 7:231-239. [PMID: 27957248 PMCID: PMC5124699 DOI: 10.4331/wjbc.v7.i4.231] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/17/2016] [Accepted: 10/25/2016] [Indexed: 02/05/2023] Open
Abstract
Radiation-induced lung fibrosis (RILF) is a common side effect of thoracic irradiation therapy and leads to high mortality rates after cancer treatment. Radiation injury induces inflammatory M1 macrophage polarization leading to radiation pneumonitis, the first stage of RILF progression. Fibrosis occurs due to the transition of M1 macrophages to the anti-inflammatory pro-fibrotic M2 phenotype, and the resulting imbalance of macrophage regulated inflammatory signaling. Non-coding RNA signaling has been shown to play a large role in the regulation of the M2 mediated signaling pathways that are associated with the development and progression of fibrosis. While many studies show the link between M2 macrophages and fibrosis, there are only a few that explore their distinct role and the regulation of their signaling by non-coding RNA in RILF. In this review we summarize the current body of knowledge describing the roles of M2 macrophages in RILF, with an emphasis on the expression and functions of non-coding RNAs.
Collapse
|
33
|
Kolahian S, Fernandez IE, Eickelberg O, Hartl D. Immune Mechanisms in Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2016; 55:309-22. [DOI: 10.1165/rcmb.2016-0121tr] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
34
|
Blockade of Wnt/β-Catenin Pathway Aggravated Silica-Induced Lung Inflammation through Tregs Regulation on Th Immune Responses. Mediators Inflamm 2016; 2016:6235614. [PMID: 27069316 PMCID: PMC4812397 DOI: 10.1155/2016/6235614] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/13/2016] [Accepted: 02/15/2016] [Indexed: 12/31/2022] Open
Abstract
CD4+ T cells play an important role in regulating silica-induced inflammation and fibrosis. Recent studies showed that Wnt/β-catenin pathway could modulate the function and the differentiation of CD4+ T cells. Therefore, Wnt/β-catenin pathway may participate in the development and progress of silicosis. To investigate the role of Wnt/β-catenin pathway, we used lentivirus expressing β-catenin shRNA to block the Wnt/β-catenin pathway by intratracheal instillation to the mice model of silicosis. Treatment of lentivirus could significantly aggravate the silica-induced lung inflammation and attenuated the fibrosis at the late stage. By analyzing CD4+ T cells, we found that blockade of Wnt/β-catenin pathway suppressed regulatory T cells (Tregs). Reciprocally, enhanced Th17 response was responsible for the further accumulation of neutrophils and production of proinflammatory cytokines. In addition, blockade of Wnt/β-catenin pathway delayed the Th1/Th2 polarization by inhibiting Tregs and Th2 response. These results indicated that Wnt/β-catenin pathway could regulate Tregs to modulate Th immune response, which finally altered the pathological character of silicosis. Our study suggested that Wnt/β-catenin pathway might be a potential target to treat the silica-induced inflammation and fibrosis.
Collapse
|
35
|
Jiang S, Wang Z, Ouyang H, Liu Z, Li L, Shi Y. Aberrant expression of cytokine interleukin 9 along with interleukin 4 and interferon γ in connective tissue disease-associated interstitial lung disease: association with severity of pulmonary fibrosis. Arch Med Sci 2016; 12:101-6. [PMID: 26925124 PMCID: PMC4754357 DOI: 10.5114/aoms.2015.47877] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 02/07/2014] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Connective tissues diseases (CTDs) are a heterogeneous group of disorders that share certain clinical characteristics and disturbed immunoregulation. Interstitial lung diseases (ILDs), also known as diffuse parenchymal lung diseases, are among the most serious pulmonary complications associated with CTDs. Interleukin 9 (IL-9), IL-4 and interferon γ (IFN-γ) - cytokines with important roles in autoimmune disease - were studied in CTD patients and CTD-ILD patients. MATERIAL AND METHODS Sixty-one hospitalized untreated CTD patients were recruited, and 20 healthy volunteers were enrolled as controls. The 61 CTD patients were divided into a simple CTD group and a CTD-ILD group, and the plasma protein IL-9, IL-4 and IFN-γ levels were measured by enzyme-linked immunosorbent assay (ELISA). RESULTS The results indicate that the serum IL-9 levels were significantly higher in CTD-ILD and simple CTD patients than they were in healthy controls (each p < 0.05) and that the levels were elevated in CTD-ILD patients compared with simple CTD patients (p < 0.05). The IL-4 levels were higher in CTD-ILD patients than they were in the simple CTD patients (p < 0.05) and healthy controls (p < 0.01). In addition, the serum IL-9 levels were negatively correlated with the level of IFN-γ (r (2) = 0.34, p = 0.01), the estimated percentage of predicted forced vital capacity (FVC%) (r (2) = 0.36, p = 0.00) and the estimated percentage of predicted diffusing capacity (DLCO%) (r (2) = 0.27, p = 0.04) and were positively correlated with the IL-4 level (r (2) = 0.31, p = 0.01). CONCLUSIONS Interleukin-9 may play an important role in the pathogenesis of CTD and may contribute to the progression of interstitial lung injury in CTD patients.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Zhi Wang
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Han Ouyang
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Zhichun Liu
- Department of Rheumatology, The Second Affiliated Hospital of Soochow University, Soochow, China
| | - Lingyun Li
- Department of Pharmacology, Medical College of Soochow University, Soochow, China
| | - Yongbing Shi
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Soochow, China
| |
Collapse
|
36
|
A deleterious role for Th9/IL-9 in hepatic fibrogenesis. Sci Rep 2016; 6:18694. [PMID: 26728971 PMCID: PMC4700496 DOI: 10.1038/srep18694] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 11/23/2015] [Indexed: 12/12/2022] Open
Abstract
T helper 9 (Th9) cells, a recently recognized Th cell subset, are involved in autoimmune diseases. We aimed to investigate the role of Th9/interleukin-9 (IL-9) in the pathogenesis of hepatic fibrosis. Th9 and Th17 cells were quantified in chronic hepatitis B (CHB) patients with hepatic fibrosis, HBV-associated liver cirrhosis (LC) patients and healthy controls (HC). The percentages of Th9 and Th17 cells, concentrations of IL-9 and IL-17, as well as expression of IL-17, TNF-α, IL-6, IL-4, IL-21, TGF-β1 and IFN-γ were significantly increased in plasma of CHB and LC patients compared with those in HC. Splenic Th9 and Th17 cells, plasma concentrations and liver expression of IL-9 and IL-17A were significantly elevated in mice with hepatic fibrosis compared with controls. Neutralization of IL-9 in mice ameliorated hepatic fibrosis, attenuated the activation of hepatic stellate cells, reduced frequencies of Th9, Th17 and Th1 cells in spleen, and suppressed expression of IL-9, IL-17A, IFN-γ, TGF-β1, IL-6, IL-4 and TNF-α in plasma and liver respectively. Our data suggest a deleterious role of Th9/IL-9 in increasing hepatic fibrosis and exacerbating disease endpoints, indicating that Th9/IL9 based immunotherapy may be a promising approach for treating hepatic fibrosis.
Collapse
|
37
|
Wermuth PJ, Jimenez SA. The significance of macrophage polarization subtypes for animal models of tissue fibrosis and human fibrotic diseases. Clin Transl Med 2015; 4:2. [PMID: 25852818 PMCID: PMC4384891 DOI: 10.1186/s40169-015-0047-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/20/2015] [Indexed: 12/15/2022] Open
Abstract
The systemic and organ-specific human fibrotic disorders collectively represent one of the most serious health problems world-wide causing a large proportion of the total world population mortality. The molecular pathways involved in their pathogenesis are complex and despite intensive investigations have not been fully elucidated. Whereas chronic inflammatory cell infiltration is universally present in fibrotic lesions, the central role of monocytes and macrophages as regulators of inflammation and fibrosis has only recently become apparent. However, the precise mechanisms involved in the contribution of monocytes/macrophages to the initiation, establishment, or progression of the fibrotic process remain largely unknown. Several monocyte and macrophage subpopulations have been identified, with certain phenotypes promoting inflammation whereas others display profibrotic effects. Given the unmet need for effective treatments for fibroproliferative diseases and the crucial regulatory role of monocyte/macrophage subpopulations in fibrogenesis, the development of therapeutic strategies that target specific monocyte/macrophage subpopulations has become increasingly attractive. We will provide here an overview of the current understanding of the role of monocyte/macrophage phenotype subpopulations in animal models of tissue fibrosis and in various systemic and organ-specific human fibrotic diseases. Furthermore, we will discuss recent approaches to the design of effective anti-fibrotic therapeutic interventions by targeting the phenotypic differences identified between the various monocyte and macrophage subpopulations.
Collapse
Affiliation(s)
- Peter J Wermuth
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Bluemle Life Science Building Suite 509, 233 South 10th Street, Philadelphia, PA 19107-5541 USA
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Bluemle Life Science Building Suite 509, 233 South 10th Street, Philadelphia, PA 19107-5541 USA
| |
Collapse
|
38
|
Huaux F, Lo Re S, Giordano G, Uwambayinema F, Devosse R, Yakoub Y, Panin N, Palmai-Pallag M, Rabolli V, Delos M, Marbaix E, Dauguet N, Couillin I, Ryffel B, Renauld JC, Lison D. IL-1α induces CD11b(low) alveolar macrophage proliferation and maturation during granuloma formation. J Pathol 2015; 235:698-709. [PMID: 25421226 DOI: 10.1002/path.4487] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 10/31/2014] [Accepted: 11/21/2014] [Indexed: 12/28/2022]
Abstract
Macrophages play a central role in immune and tissue responses of granulomatous lung diseases induced by pathogens and foreign bodies. Circulating monocytes are generally viewed as central precursors of these tissue effector macrophages. Here, we provide evidence that granulomas derive from alveolar macrophages serving as a local reservoir for the expansion of activated phagocytic macrophages. By exploring lung granulomatous responses to silica particles in IL-1-deficient mice, we found that the absence of IL-1α, but not IL-1β, was associated with reduced CD11b(high) phagocytic macrophage accumulation and fewer granulomas. This defect was associated with impaired alveolar clearance and resulted in the development of pulmonary alveolar proteinosis (PAP). Reconstitution of IL-1α(-/-) mice with recombinant IL-1α restored lung clearance functions and the pulmonary accumulation of CD11b(high) phagocytic macrophages. Mechanistically, IL-1α induced the proliferation of CD11b(low) alveolar macrophages and differentiated these cells into CD11b(high) macrophages which perform critical phagocytic functions and organize granuloma. We newly discovered here that IL-1α triggers lung responses requiring macrophage proliferation and maturation from tissue-resident macrophages.
Collapse
Affiliation(s)
- François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
van den Brule S, Beckers E, Chaurand P, Liu W, Ibouraadaten S, Palmai-Pallag M, Uwambayinema F, Yakoub Y, Avellan A, Levard C, Haufroid V, Marbaix E, Thill A, Lison D, Rose J. Nanometer-long Ge-imogolite nanotubes cause sustained lung inflammation and fibrosis in rats. Part Fibre Toxicol 2014; 11:67. [PMID: 25497478 PMCID: PMC4276264 DOI: 10.1186/s12989-014-0067-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 11/13/2014] [Indexed: 11/16/2022] Open
Abstract
Background Ge-imogolites are short aluminogermanate tubular nanomaterials with attractive prospected industrial applications. In view of their nano-scale dimensions and high aspect ratio, they should be examined for their potential to cause respiratory toxicity. Here, we evaluated the respiratory biopersistence and lung toxicity of 2 samples of nanometer-long Ge-imogolites. Methods Rats were intra-tracheally instilled with single wall (SW, 70 nm length) or double wall (DW, 62 nm length) Ge-imogolites (0.02-2 mg/rat), as well as with crocidolite and the hard metal particles WC-Co, as positive controls. The biopersistence of Ge-imogolites and their localization in the lung were assessed by ICP-MS, X-ray fluorescence, absorption spectroscopy and computed micro-tomography. Acute inflammation and genotoxicity (micronuclei in isolated type II pneumocytes) was assessed 3 d post-exposure; chronic inflammation and fibrosis after 2 m. Results Cytotoxic and inflammatory responses were shown in bronchoalveolar lavage 3 d after instillation with Ge-imogolites. Sixty days after exposure, a persistent dose-dependent inflammation was still observed. Total lung collagen, reflected by hydroxyproline lung content, was increased after SW and DW Ge-imogolites. Histology revealed lung fibre reorganization and accumulation in granulomas with epithelioid cells and foamy macrophages and thickening of the alveolar walls. Overall, the inflammatory and fibrotic responses induced by SW and DW Ge-imogolites were more severe (on a mass dose basis) than those induced by crocidolite. A persistent fraction of Ge-imogolites (15% of initial dose) was mostly detected as intact structures in rat lungs 2 m after instillation and was localized in fibrotic alveolar areas. In vivo induction of micronuclei was significantly increased 3 d after SW and DW Ge-imogolite instillation at non-inflammatory doses, indicating the contribution of primary genotoxicity. Conclusions We showed that nm-long Ge-imogolites persist in the lung and promote genotoxicity, sustained inflammation and fibrosis, indicating that short high aspect ratio nanomaterials should not be considered as innocuous materials. Our data also suggest that Ge-imogolite structure and external surface determine their toxic activity. Electronic supplementary material The online version of this article (doi:10.1186/s12989-014-0067-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sybille van den Brule
- Louvain centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Avenue E. Mounier 52 - bte B1.52.12, 1200, Brussels, Belgium.
| | - Emilie Beckers
- Louvain centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Avenue E. Mounier 52 - bte B1.52.12, 1200, Brussels, Belgium.
| | - Perrine Chaurand
- CEREGE, Aix Marseille Université, CNRS, IRD, UM34, UMR 7330, Europole de l'arbois - BP 80, 13545, Aix en Provence, France. .,iCEINT, International Consortium for the Environmental Implications of Nanotechnology, CNRS, Duke University, 13545, Aix en Provence, France.
| | - Wei Liu
- CEREGE, Aix Marseille Université, CNRS, IRD, UM34, UMR 7330, Europole de l'arbois - BP 80, 13545, Aix en Provence, France. .,iCEINT, International Consortium for the Environmental Implications of Nanotechnology, CNRS, Duke University, 13545, Aix en Provence, France.
| | - Saloua Ibouraadaten
- Louvain centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Avenue E. Mounier 52 - bte B1.52.12, 1200, Brussels, Belgium.
| | - Mihaly Palmai-Pallag
- Louvain centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Avenue E. Mounier 52 - bte B1.52.12, 1200, Brussels, Belgium.
| | - Francine Uwambayinema
- Louvain centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Avenue E. Mounier 52 - bte B1.52.12, 1200, Brussels, Belgium.
| | - Yousof Yakoub
- Louvain centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Avenue E. Mounier 52 - bte B1.52.12, 1200, Brussels, Belgium.
| | - Astrid Avellan
- CEREGE, Aix Marseille Université, CNRS, IRD, UM34, UMR 7330, Europole de l'arbois - BP 80, 13545, Aix en Provence, France. .,iCEINT, International Consortium for the Environmental Implications of Nanotechnology, CNRS, Duke University, 13545, Aix en Provence, France.
| | - Clément Levard
- CEREGE, Aix Marseille Université, CNRS, IRD, UM34, UMR 7330, Europole de l'arbois - BP 80, 13545, Aix en Provence, France. .,iCEINT, International Consortium for the Environmental Implications of Nanotechnology, CNRS, Duke University, 13545, Aix en Provence, France.
| | - Vincent Haufroid
- Louvain centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Avenue E. Mounier 52 - bte B1.52.12, 1200, Brussels, Belgium. .,Department of Clinical Chemistry, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Avenue Hippocrate 10, 1200, Brussels, Belgium.
| | - Etienne Marbaix
- De Duve Institute, Université catholique de Louvain, Avenue Hippocrate 75 - bte B1.75.02, 1200, Brussels, Belgium.
| | - Antoine Thill
- Laboratoire Interdisciplinaire sur l'Organisation Nanométrique et Supramoléculaire, CEA Saclay, 91191, Gif sur Yvette, France.
| | - Dominique Lison
- Louvain centre for Toxicology and Applied Pharmacology, Université catholique de Louvain, Avenue E. Mounier 52 - bte B1.52.12, 1200, Brussels, Belgium.
| | - Jérôme Rose
- CEREGE, Aix Marseille Université, CNRS, IRD, UM34, UMR 7330, Europole de l'arbois - BP 80, 13545, Aix en Provence, France. .,iCEINT, International Consortium for the Environmental Implications of Nanotechnology, CNRS, Duke University, 13545, Aix en Provence, France.
| |
Collapse
|
40
|
Shimizu Y, Dobashi K, Nagase H, Ohta K, Sano T, Matsuzaki S, Ishii Y, Satoh T, Koka M, Yokoyama A, Ohkubo T, Ishii Y, Kamiya T. Co-localization of iron binding on silica with p62/sequestosome1 (SQSTM1) in lung granulomas of mice with acute silicosis. J Clin Biochem Nutr 2014; 56:74-83. [PMID: 25834305 PMCID: PMC4306660 DOI: 10.3164/jcbn.14-44] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 09/02/2014] [Indexed: 01/05/2023] Open
Abstract
The cellular mechanisms involved in the development of silicosis have not been fully elucidated. This study aimed to examine influence of silica-induced lung injury on autophagy. Suspensions of crystalline silica particles were administered transnasally to C57BL/6j mice. Immunohistochemical examination for Fas and p62 protein expression was performed using lung tissue specimens. Two-dimensional and quantitative analysis of silica deposits in the lungs were performed in situ using lung tissue sections by an in-air microparticle induced X-ray emission (in-air micro-PIXE) analysis system, which was based on irrradiation of specimens with a proton ion microbeam. Quantitative analysis showed a significant increase of iron levels on silica particles (assessed as the ratio of Fe relative to Si) on day 56 compared with day 7 (p<0.05). Fas and p62 were expressed by histiocytes in granulomas on day 7, and the expressions persisted for day 56. Fas- and p62-expressing histiocytes were co-localized in granulomas with silica particles that showed an increase of iron levels on silica particles in mouse lungs. Iron complexed with silica induces apoptosis, and may lead to dysregulations of autophagy in histiocytes of granulomas, and these mechanisms may contribute to granuloma development and progression in silicosis.
Collapse
Affiliation(s)
- Yasuo Shimizu
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu-machi, Tochigi 321-0293, Japan ; Department of Respiratory Medicine, Maebashi Red Cross Hospital, 3-21-36 Asahi-cho, Maebashi-shi, Tochigi 371-0014, Japan ; Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi-shi, Gunma 371-8511, Japan
| | - Kunio Dobashi
- Gunma University School of Health Sciences, 3-39-22 Showa-machi, Maebashi-shi, Gunma 371-8514, Japan
| | - Hiroyuki Nagase
- Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Ken Ohta
- Department of Respiratory Diseases, National Hospital Organization Tokyo National Hospital, Tokyo, 3-1-1 Takeoka, Kiyose-shi, Tokyo 204-8585, Japan
| | - Takaaki Sano
- Department of Diagnostic Pathology, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi-shi, Gunma 371-8511, Japan
| | - Shinichi Matsuzaki
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, 3-39-15 Showa-machi, Maebashi-shi, Gunma 371-8511, Japan
| | - Yoshiki Ishii
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu-machi, Tochigi 321-0293, Japan
| | - Takahiro Satoh
- Japan Atomic Energy Agency, Takasaki Advanced Radiation Research Institute, 1233 Watanuki-machi, Takasaki-shi, Gunma 370-1292, Japan
| | - Masashi Koka
- Japan Atomic Energy Agency, Takasaki Advanced Radiation Research Institute, 1233 Watanuki-machi, Takasaki-shi, Gunma 370-1292, Japan
| | - Akihito Yokoyama
- Japan Atomic Energy Agency, Takasaki Advanced Radiation Research Institute, 1233 Watanuki-machi, Takasaki-shi, Gunma 370-1292, Japan
| | - Takeru Ohkubo
- Japan Atomic Energy Agency, Takasaki Advanced Radiation Research Institute, 1233 Watanuki-machi, Takasaki-shi, Gunma 370-1292, Japan
| | - Yasuyuki Ishii
- Japan Atomic Energy Agency, Takasaki Advanced Radiation Research Institute, 1233 Watanuki-machi, Takasaki-shi, Gunma 370-1292, Japan
| | - Tomihiro Kamiya
- Japan Atomic Energy Agency, Takasaki Advanced Radiation Research Institute, 1233 Watanuki-machi, Takasaki-shi, Gunma 370-1292, Japan
| |
Collapse
|
41
|
|
42
|
van den Brule S, Huaux F, Uwambayinema F, Ibouraadaten S, Yakoub Y, Palmai-Pallag M, Trottein F, Renauld JC, Lison D. Lung inflammation and thymic atrophy after bleomycin are controlled by the prostaglandin D2 receptor DP1. Am J Respir Cell Mol Biol 2014; 50:212-22. [PMID: 24003988 DOI: 10.1165/rcmb.2012-0520oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Acute lung injury (ALI) can be accompanied by secondary systemic manifestations. In a model of ALI induced by bleomycin (bleo), we examined the response of D prostanoid receptor 1 (DP1)-deficient mice (DP1(-/-)) to better understand these processes. DP1 deficiency aggravated the toxicity of bleo as indicated by enhanced body weight loss, mortality, and lung inflammation including bronchoalveolar permeability and neutrophilia. Thymic atrophy was also observed after bleo and was strongly exacerbated in DP1(-/-) mice. This resulted from the enhanced depletion of immature T lymphocytes in the thymus of DP1(-/-) mice, a phenomenon usually related to increased glucocorticoid release in blood. Serum corticosterone was more elevated in DP1(-/-) mice after bleo than in wild-type (wt) mice. Thymocytes of DP1(-/-) mice were not more sensitive to dexamethasone in vitro, and systemic delivery of dexamethasone or peritoneal inflammation after LPS induced a similar thymic atrophy in wt and DP1(-/-) mice, indicating that pulmonary DP1 was critical to the control of thymic atrophy after bleo. DP1(-/-) mice showed increased lung and/or blood mediators involved in neutrophil recruitment and/or glucocorticoid production/thymic atrophy (osteopontin, leukemia inhibitory factor, and keratinocyte-derived chemokine) after bleo. Finally, local pulmonary DP1 activation or inhibition in wt mice abrogated or amplified thymic atrophy after bleo, respectively. Altogether, our data reveal that ALI can perturb the systemic T-cell pool by inducing thymic atrophy and that both pathological processes are controlled by the pulmonary DP1 receptor. This new pathway represents a potential therapeutic target in ALI.
Collapse
|
43
|
Fructo-oligosaccharide attenuates the production of pro-inflammatory cytokines and the activation of JNK/Jun pathway in the lungs of d-galactose-treated Balb/cJ mice. Eur J Nutr 2013; 53:449-56. [DOI: 10.1007/s00394-013-0545-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 06/04/2013] [Indexed: 10/26/2022]
|
44
|
Lo Re S, Lison D, Huaux F. CD4+ T lymphocytes in lung fibrosis: diverse subsets, diverse functions. J Leukoc Biol 2013; 93:499-510. [PMID: 23159927 DOI: 10.1189/jlb.0512261] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
The discovery of several subsets of CD4(+) Th lymphocytes has contributed to refine and to challenge our understanding of the roles of CD4(+) T cells in the pathogenesis of fibrotic lung diseases. Here, we review recent findings, indicating that CD4(+) T subpopulations possess contrasting pro- and antifibrotic activities in human and experimental lung fibrosis. Special attention is given to delineate the activity of the newly discovered CD4(+) T lymphocyte subsets (Tregs, Th22, and Th9) on fibroblast function and matrix deposition through the release of growth factors, cytokines, and eicosanoids. It appears that the function of a CD4(+) T lymphocyte subset or of a cytokine can differ with the disease stage (acute vs. chronic), pulmonary localization (bronchial vs. alveolar), cellular level (epithelial cell vs. fibroblast), or immune environment (inflammatory or immunosuppressive). Integrating our recent understanding of the contrasting functions of T lymphocyte subsets in fibrosis provides new insights and opportunities for improved treatment strategies.
Collapse
Affiliation(s)
- Sandra Lo Re
- Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain, Avenue Mounier 52, Brussels, Belgium
| | | | | |
Collapse
|
45
|
Todd NW, Luzina IG, Atamas SP. Molecular and cellular mechanisms of pulmonary fibrosis. FIBROGENESIS & TISSUE REPAIR 2012; 5:11. [PMID: 22824096 PMCID: PMC3443459 DOI: 10.1186/1755-1536-5-11] [Citation(s) in RCA: 312] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/28/2012] [Indexed: 12/22/2022]
Abstract
Pulmonary fibrosis is a chronic lung disease characterized by excessive accumulation of extracellular matrix (ECM) and remodeling of the lung architecture. Idiopathic pulmonary fibrosis is considered the most common and severe form of the disease, with a median survival of approximately three years and no proven effective therapy. Despite the fact that effective treatments are absent and the precise mechanisms that drive fibrosis in most patients remain incompletely understood, an extensive body of scientific literature regarding pulmonary fibrosis has accumulated over the past 35 years. In this review, we discuss three broad areas which have been explored that may be responsible for the combination of altered lung fibroblasts, loss of alveolar epithelial cells, and excessive accumulation of ECM: inflammation and immune mechanisms, oxidative stress and oxidative signaling, and procoagulant mechanisms. We discuss each of these processes separately to facilitate clarity, but certainly significant interplay will occur amongst these pathways in patients with this disease.
Collapse
Affiliation(s)
- Nevins W Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | | |
Collapse
|
46
|
Poynter ME. Airway epithelial regulation of allergic sensitization in asthma. Pulm Pharmacol Ther 2012; 25:438-46. [PMID: 22579987 DOI: 10.1016/j.pupt.2012.04.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 04/18/2012] [Accepted: 04/27/2012] [Indexed: 02/07/2023]
Abstract
While many of the contributing cell types and mediators of allergic asthma are known, less well understood are the factors that influence the development of allergic responses that lead to the development of allergic asthma. As the first airway cell type to respond to inhaled factors, the epithelium orchestrates downstream interactions between dendritic cells (DCs) and CD4⁺ T cells that quantitatively and qualitatively dictate the degree and type of the allergic asthma phenotype, making the epithelium of critical importance for the genesis of allergies that later manifest in allergic asthma. Amongst the molecular processes of critical importance in airway epithelium is the transcription factor, nuclear factor-kappaB (NF-κB). This review will focus primarily on the genesis of pulmonary allergies and the participation of airway epithelial NF-κB activation therein, using examples from our own work on nitrogen dioxide (NO₂) exposure and genetic modulation of airway epithelial NF-κB activation. In addition, the mechanisms through which Serum Amyloid A (SAA), an NF-κB-regulated, epithelial-derived mediator, influences allergic sensitization and asthma severity will be presented. Knowledge of the molecular and cellular processes regulating allergic sensitization in the airways has the potential to provide powerful insight into the pathogenesis of allergy, as well as targets for the prevention and treatment of asthma.
Collapse
Affiliation(s)
- Matthew E Poynter
- Department of Medicine and Vermont Lung Center, University of Vermont, Given E410A, 89 Beaumont Avenue, Burlington, VT 05405, USA.
| |
Collapse
|
47
|
Abstract
Interleukin-9 (IL-9), cloned more than 20 years ago, was initially thought to be a Th2-specific cytokine. This assumption was initially confirmed by functional analyses showing that both IL-9 and Th2 cells play an important role in the pathogenesis of asthma, IgE class switch recombination, and resolution of parasitic infections. However, recently it was shown that IL-9-producing CD4(+) T cells represent the discrete T helper subset Th9 cells. Herein, we will review the cytokines and transcription factors known to promote the development of Th9 cells and their potential functional properties in relation to the biological activities of IL-9. In addition, we will discuss how Th9 cells are related to Th2, Th17, and T(reg) cells, as both an alternative source of IL-9 and in view of the fact that plasticity of CD4(+) T cell differentiation is currently a strong matter of debate in immunologic research.
Collapse
Affiliation(s)
- Michael Stassen
- Institute for Immunology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | | | | |
Collapse
|
48
|
Deniz O, Gumus S, Ors F, Yaman H, Battal B, Karaman B, Acikel CH, Ucar E, Cakir E, Tozkoparan E, Bilgic H. Serum lactate dehydrogenase levels significantly correlate with radiological extent of disease and spirometric values in patients with silicosis due to denim sandblasting. Clin Chem Lab Med 2012; 50:483-8. [DOI: 10.1515/cclm.2011.795] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 10/26/2011] [Indexed: 11/15/2022]
|
49
|
Ckless K, Hodgkins SR, Ather JL, Martin R, Poynter ME. Epithelial, dendritic, and CD4(+) T cell regulation of and by reactive oxygen and nitrogen species in allergic sensitization. BIOCHIMICA ET BIOPHYSICA ACTA 2011; 1810:1025-34. [PMID: 21397661 PMCID: PMC3140554 DOI: 10.1016/j.bbagen.2011.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Revised: 02/28/2011] [Accepted: 03/03/2011] [Indexed: 12/15/2022]
Abstract
BACKGROUND While many of the contributing cell types and mediators of allergic asthma are known, less well understood are the factors that induce allergy in the first place. Amongst the mediators speculated to affect initial allergen sensitization and the development of pathogenic allergic responses to innocuous inhaled antigens and allergens are exogenously or endogenously generated reactive oxygen species (ROS) and reactive nitrogen species (RNS). SCOPE OF REVIEW The interactions between ROS/RNS, dendritic cells (DCs), and CD4(+) T cells, as well as their modulation by lung epithelium, are of critical importance for the genesis of allergies that later manifest in allergic asthma. Therefore, this review will primarily focus on the initiation of pulmonary allergies and the role that ROS/RNS may play in the steps therein, using examples from our own work on the roles of NO(2) exposure and airway epithelial NF-κB activation. MAJOR CONCLUSIONS Endogenously generated ROS/RNS and those encountered from environmental sources interact with epithelium, DCs, and CD4(+) T cells to orchestrate allergic sensitization through modulation of the activities of each of these cell types, which quantitiatively and qualitatively dictate the degree and type of the allergic asthma phenotype. GENERAL SIGNIFICANCE Knowledge of the effects of ROS/RNS at the molecular and cellular levels has the potential to provide powerful insight into the balance between inhalational tolerance (the typical immunologic response to an innocuous inhaled antigen) and allergy, as well as to potentially provide mechanistic targets for the prevention and treatment of asthma.
Collapse
Affiliation(s)
- Karina Ckless
- Department of Chemistry, SUNY Plattsburgh, University of Vermont
| | | | - Jennifer L. Ather
- Department of Medicine and Vermont Lung Center, University of Vermont
| | - Rebecca Martin
- Department of Medicine and Vermont Lung Center, University of Vermont
| | | |
Collapse
|
50
|
Zou J, du Prel Carroll X, Liang X, Wang D, Li C, Yuan B, Leeper-Woodford S. Alterations of serum biomarkers associated with lung ventilation function impairment in coal workers: a cross-sectional study. Environ Health 2011; 10:83. [PMID: 21943057 PMCID: PMC3192731 DOI: 10.1186/1476-069x-10-83] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 09/27/2011] [Indexed: 05/04/2023]
Abstract
BACKGROUND Previous studies have demonstrated that alterations in certain circulating biomarkers may be correlated with Coal workers' pneumoconiosis (CWP). This study investigated the relationship between changes of serum biomarkers and pulmonary function during the development of CWP. METHODS Lung function parameters and specific serum indices were measured in 69 non-smoking coal workers, including 34 miners with CWP, 24 asymptomatic miners and 11 miners with minimal symptoms. The associations between changes in pulmonary function and serum indices were tested with Pearson's correlation coefficients. Multivariable analysis was used to estimate the predictive power of potential determinant variables for lung function. RESULTS Compared to healthy miners, lung function (FVC, FEV1, FEF50, FEF75, FEF25-75 % of predicted values) was decreased in miners with CWP (p < 0.05). Increased serum matrix metalloproteinase-9 (MMP-9) was associated with decreased FVC% of predicted values in the asymptomatic miners (r = -0.503, p = 0.014). CONCLUSIONS In coal mine workers, alterations of lung function parameters are associated with the development of CWP and with changes in circulating MMP-9, TIMP-9, IL-13 and IL-18R. These serum biomarkers may likely reflect the pathogenesis and progression of CWP in coal workers, and may provide for the importance of serum indicators in the early diagnosis of lung function injury in coal miners.
Collapse
Affiliation(s)
- Jimin Zou
- Department of Clinical Laboratory, Attached Kai Luan Hospital of North China Coal Medical College, Tangshan, 063000, China
| | - Xianming du Prel Carroll
- Department of Community Medicine, Mercer University School of Medicine, 1550 College Street, Macon, GA 31207, USA
- Department of Social Medicine and Health Education, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Xianhong Liang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, No. 6 Tiantan Xili, Beijing, 100050, China
| | - Dongmei Wang
- Department of Clinical Laboratory, Attached Kai Luan Hospital of North China Coal Medical College, Tangshan, 063000, China
| | - Chao Li
- Department of Clinical Laboratory, Attached Kai Luan Hospital of North China Coal Medical College, Tangshan, 063000, China
| | - Baojun Yuan
- Department of Clinical Laboratory, Attached Kai Luan Hospital of North China Coal Medical College, Tangshan, 063000, China
| | - Sandra Leeper-Woodford
- Division of Basic Medical Sciences, Mercer University School of Medicine, Macon, 1550 College Street, Macon, GA 31207, USA
| |
Collapse
|