251
|
Schrumpf JA, Amatngalim GD, Veldkamp JB, Verhoosel RM, Ninaber DK, Ordonez SR, van der Does AM, Haagsman HP, Hiemstra PS. Proinflammatory Cytokines Impair Vitamin D-Induced Host Defense in Cultured Airway Epithelial Cells. Am J Respir Cell Mol Biol 2017; 56:749-761. [PMID: 28231019 DOI: 10.1165/rcmb.2016-0289oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Vitamin D is a regulator of host defense against infections and induces expression of the antimicrobial peptide hCAP18/LL-37. Vitamin D deficiency is associated with chronic inflammatory lung diseases and respiratory infections. However, it is incompletely understood if and how (chronic) airway inflammation affects vitamin D metabolism and action. We hypothesized that long-term exposure of primary bronchial epithelial cells to proinflammatory cytokines alters their vitamin D metabolism, antibacterial activity, and expression of hCAP18/LL-37. To investigate this, primary bronchial epithelial cells were differentiated at the air-liquid interface for 14 days in the presence of the proinflammatory cytokines, TNF-α and IL-1β (TNF-α/IL-1β), and subsequently exposed to vitamin D (inactive 25(OH)D3 and active 1,25(OH)2D3). Expression of hCAP18/LL-37, vitamin D receptor, and enzymes involved in vitamin D metabolism (CYP24A1 and CYP27B1) was determined using quantitative PCR, Western blot, and immunofluorescence staining. Furthermore, vitamin D-mediated antibacterial activity was assessed using nontypeable Haemophilus influenzae. We found that TNF-α/IL-1β treatment reduced vitamin D-induced expression of hCAP18/LL-37 and killing of nontypeable H. influenzae. In addition, CYP24A1 (a vitamin D-degrading enzyme) was increased by TNF-α/IL-1β, whereas CYP27B1 (that converts 25(OH)D3 to its active form) and vitamin D receptor expression remained unaffected. Furthermore, we have demonstrated that the TNF-α/IL-1β-mediated induction of CYP24A1 was, at least in part, mediated by the transcription factor specific protein 1, and the epidermal growth factor receptor-mitogen-activated protein kinase pathway. These findings indicate that TNF-α/IL-1β decreases vitamin D-mediated antibacterial activity and hCAP18/LL-37 expression via induction of CYP24A1 and suggest that chronic inflammation impairs protective responses induced by vitamin D.
Collapse
Affiliation(s)
- Jasmijn A Schrumpf
- 1 Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands; and
| | - Gimano D Amatngalim
- 1 Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands; and
| | - Joris B Veldkamp
- 1 Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands; and
| | - Renate M Verhoosel
- 1 Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands; and
| | - Dennis K Ninaber
- 1 Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands; and
| | - Soledad R Ordonez
- 2 Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands
| | - Anne M van der Does
- 1 Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands; and
| | - Henk P Haagsman
- 2 Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands
| | - Pieter S Hiemstra
- 1 Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands; and
| |
Collapse
|
252
|
Gottdiener JS. Intersection of 2 Epidemics: Asthma and Cardiovascular Disease. JACC-HEART FAILURE 2017; 5:505-506. [PMID: 28662938 DOI: 10.1016/j.jchf.2017.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 05/10/2017] [Indexed: 10/19/2022]
Affiliation(s)
- John S Gottdiener
- Division of Cardiology, Department of Medicine, University of Maryland Medical Center, Baltimore, Maryland.
| |
Collapse
|
253
|
Eichinger KM, Resetar E, Orend J, Anderson K, Empey KM. Age predicts cytokine kinetics and innate immune cell activation following intranasal delivery of IFNγ and GM-CSF in a mouse model of RSV infection. Cytokine 2017; 97:25-37. [PMID: 28558308 DOI: 10.1016/j.cyto.2017.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/20/2017] [Accepted: 05/23/2017] [Indexed: 12/22/2022]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in young children and is further associated with increased healthcare utilization and cost of care in the first years of life. Severe RSV disease during infancy has also been linked to the later development of allergic asthma, yet there remains no licensed RSV vaccine or effective treatment. Pre-clinical and clinical studies have shown that disease severity and development of allergic asthma are associated with differences in cytokine production. As a result, stimulation of the innate host immune response with immune potentiators is gaining attention for their prospective application in populations with limited immune responses to antigenic stimuli or against pathogens for which vaccines do not exist. Specifically, macrophage-activating cytokines such as interferon gamma (IFNγ) and granulocyte colony-stimulating factor (GM-CSF) are commercially available immune potentiators used to prevent infections in patients with chronic granulomatous disease and febrile neutropenia, respectively. Moreover, an increasing number of reports describe the protective function of IFNγ and GM-CSF as vaccine adjuvants. Although a positive correlation between cytokine production and age has previously been reported, little is known about age-dependent cytokine metabolism or immune activating responses in infant compared to adult lungs. Here we use a non-compartmental pharmacokinetic model in naïve and RSV-infected infant and adult BALB/c mice to determine the effect of age on IFNγ and GM-CSF elimination and innate cell activation following intranasal delivery.
Collapse
Affiliation(s)
- Katherine M Eichinger
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Erin Resetar
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jacob Orend
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Kacey Anderson
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA
| | - Kerry M Empey
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA; Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
254
|
Mertens TCJ, Karmouty-Quintana H, Taube C, Hiemstra PS. Use of airway epithelial cell culture to unravel the pathogenesis and study treatment in obstructive airway diseases. Pulm Pharmacol Ther 2017; 45:101-113. [PMID: 28502841 DOI: 10.1016/j.pupt.2017.05.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 04/19/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are considered as two distinct obstructive diseases. Both chronic diseases share a component of airway epithelial dysfunction. The airway epithelium is localized to deal with inhaled substances, and functions as a barrier preventing penetration of such substances into the body. In addition, the epithelium is involved in the regulation of both innate and adaptive immune responses following inhalation of particles, allergens and pathogens. Through triggering and inducing immune responses, airway epithelial cells contribute to the pathogenesis of both asthma and COPD. Various in vitro research models have been described to study airway epithelial cell dysfunction in asthma and COPD. However, various considerations and cautions have to be taken into account when designing such in vitro experiments. Epithelial features of asthma and COPD can be modelled by using a variety of disease-related invoking substances either alone or in combination, and by the use of primary cells isolated from patients. Differentiation is a hallmark of airway epithelial cells, and therefore models should include the ability of cells to differentiate, as can be achieved in air-liquid interface models. More recently developed in vitro models, including precision cut lung slices, lung-on-a-chip, organoids and human induced pluripotent stem cells derived cultures, provide novel state-of-the-art alternatives to the conventional in vitro models. Furthermore, advanced models in which cells are exposed to respiratory pathogens, aerosolized medications and inhaled toxic substances such as cigarette smoke and air pollution are increasingly used to model e.g. acute exacerbations. These exposure models are relevant to study how epithelial features of asthma and COPD are affected and provide a useful tool to study the effect of drugs used in treatment of asthma and COPD. These new developments are expected to contribute to a better understanding of the complex gene-environment interactions that contribute to development and progression of asthma and COPD.
Collapse
Affiliation(s)
- Tinne C J Mertens
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christian Taube
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
255
|
Zemski Berry KA, Murphy RC, Kosmider B, Mason RJ. Lipidomic characterization and localization of phospholipids in the human lung. J Lipid Res 2017; 58:926-933. [PMID: 28280112 PMCID: PMC5408611 DOI: 10.1194/jlr.m074955] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/17/2017] [Indexed: 01/22/2023] Open
Abstract
Lipids play a central role in lung physiology and pathology; however, a comprehensive lipidomic characterization of human pulmonary cells relevant to disease has not been performed. The cells involved in lung host defense, including alveolar macrophages (AMs), bronchial epithelial cells (BECs), and alveolar type II cells (ATIIs), were isolated from human subjects and lipidomic analysis by LC-MS and LC-MS/MS was performed. Additionally, pieces of lung tissue from the same donors were analyzed by MALDI imaging MS in order to determine lipid localization in the tissue. The unique distribution of phospholipids in ATIIs, BECs, and AMs from human subjects was accomplished by subjecting the large number of identified phospholipid molecular species to univariant statistical analysis. Specific MALDI images were generated based on the univariant statistical analysis data to reveal the location of specific cell types within the human lung slice. While the complex composition and function of the lipidome in various disease states is currently poorly understood, this method could be useful for the characterization of lipid alterations in pulmonary disease and may aid in a better understanding of disease pathogenesis.
Collapse
Affiliation(s)
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - Beata Kosmider
- Department of Medicine, National Jewish Health, Denver, CO 80206
| | - Robert J Mason
- Department of Medicine, National Jewish Health, Denver, CO 80206
| |
Collapse
|
256
|
Polosukhin VV, Richmond BW, Du RH, Cates JM, Wu P, Nian H, Massion PP, Ware LB, Lee JW, Kononov AV, Lawson WE, Blackwell TS. Secretory IgA Deficiency in Individual Small Airways Is Associated with Persistent Inflammation and Remodeling. Am J Respir Crit Care Med 2017; 195:1010-1021. [PMID: 27911098 PMCID: PMC5422646 DOI: 10.1164/rccm.201604-0759oc] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 12/01/2016] [Indexed: 01/01/2023] Open
Abstract
RATIONALE Maintenance of a surface immune barrier is important for homeostasis in organs with mucosal surfaces that interface with the external environment; however, the role of the mucosal immune system in chronic lung diseases is incompletely understood. OBJECTIVES We examined the relationship between secretory IgA (SIgA) on the mucosal surface of small airways and parameters of inflammation and airway wall remodeling in chronic obstructive pulmonary disease (COPD). METHODS We studied 1,104 small airways (<2 mm in diameter) from 50 former smokers with COPD and 39 control subjects. Small airways were identified on serial tissue sections and examined for epithelial morphology, SIgA, bacterial DNA, nuclear factor-κB activation, neutrophil and macrophage infiltration, and airway wall thickness. MEASUREMENTS AND MAIN RESULTS Morphometric evaluation of small airways revealed increased mean airway wall thickness and inflammatory cell counts in lungs from patients with COPD compared with control subjects, whereas SIgA level on the mucosal surface was decreased. However, when small airways were classified as SIgA intact or SIgA deficient, we found that pathologic changes were localized almost exclusively to SIgA-deficient airways, regardless of study group. SIgA-deficient airways were characterized by (1) abnormal epithelial morphology, (2) invasion of bacteria across the apical epithelial barrier, (3) nuclear factor-κB activation, (4) accumulation of macrophages and neutrophils, and (5) fibrotic remodeling of the airway wall. CONCLUSIONS Our findings support the concept that localized, acquired SIgA deficiency in individual small airways of patients with COPD allows colonizing bacteria to cross the epithelial barrier and drive persistent inflammation and airway wall remodeling, even after smoking cessation.
Collapse
Affiliation(s)
- Vasiliy V. Polosukhin
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Bradley W. Richmond
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Cell and Developmental Biology
| | - Rui-Hong Du
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Justin M. Cates
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Pingsheng Wu
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Hui Nian
- Department of Biostatistics, and
| | - Pierre P. Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Jae Woo Lee
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, California; and
| | - Alexey V. Kononov
- Department of Pathology, Omsk State Medical Academy, Omsk, Russian Federation
| | - William E. Lawson
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Timothy S. Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, and
- Department of Cell and Developmental Biology
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| |
Collapse
|
257
|
Zarcone MC, van Schadewijk A, Duistermaat E, Hiemstra PS, Kooter IM. Diesel exhaust alters the response of cultured primary bronchial epithelial cells from patients with chronic obstructive pulmonary disease (COPD) to non-typeable Haemophilus influenzae. Respir Res 2017; 18:27. [PMID: 28129777 PMCID: PMC5273858 DOI: 10.1186/s12931-017-0510-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 01/16/2017] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Exacerbations constitute a major cause of morbidity and mortality in patients suffering from chronic obstructive pulmonary disease (COPD). Both bacterial infections, such as those with non-typeable Haemophilus influenzae (NTHi), and exposures to diesel engine emissions are known to contribute to exacerbations in COPD patients. However, the effect of diesel exhaust (DE) exposure on the epithelial response to microbial stimulation is incompletely understood, and possible differences in the response to DE of epithelial cells from COPD patients and controls have not been studied. METHODS Primary bronchial epithelial cells (PBEC) were obtained from age-matched COPD patients (n = 7) and controls (n = 5). PBEC were cultured at the air-liquid interface (ALI) to achieve mucociliary differentiation. ALI-PBECs were apically exposed for 1 h to a stream of freshly generated whole DE or air. Exposure was followed by 3 h incubation in presence or absence of UV-inactivated NTHi before analysis of epithelial gene expression. RESULTS DE alone induced an increase in markers of oxidative stress (HMOX1, 50-100-fold) and of the integrated stress response (CHOP, 1.5-2-fold and GADD34, 1.5-fold) in cells from both COPD patients and controls. Exposure of COPD cultures to DE followed by NTHi caused an additive increase in GADD34 expression (up to 3-fold). Importantly, DE caused an inhibition of the NTHi-induced expression of the antimicrobial peptide S100A7, and of the chaperone protein HSP5A/BiP. CONCLUSIONS Our findings show that DE exposure of differentiated primary airway epithelial cells causes activation of the gene expression of HMOX1 and markers of integrated stress response to a similar extent in cells from COPD donors and controls. Furthermore, DE further increased the NTHi-induced expression of GADD34, indicating a possible enhancement of the integrated stress response. DE reduced the NTHi-induced expression of S100A7. These data suggest that DE exposure may cause adverse health effects in part by decreasing host defense against infection and by modulating stress responses.
Collapse
Affiliation(s)
- Maria C Zarcone
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Annemarie van Schadewijk
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | | | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Ingeborg M Kooter
- Netherlands Organization for Applied Scientific Research, Utrecht, The Netherlands
| |
Collapse
|
258
|
Airway mucus, inflammation and remodeling: emerging links in the pathogenesis of chronic lung diseases. Cell Tissue Res 2017; 367:537-550. [PMID: 28108847 DOI: 10.1007/s00441-016-2562-z] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022]
Abstract
Airway mucus obstruction is a hallmark of many chronic lung diseases including rare genetic disorders such as cystic fibrosis (CF) and primary ciliary dyskinesia, as well as common lung diseases such as asthma and chronic obstructive pulmonary disease (COPD), which have emerged as a leading cause of morbidity and mortality worldwide. However, the role of excess airway mucus in the in vivo pathogenesis of these diseases remains poorly understood. The generation of mice with airway-specific overexpression of epithelial Na+ channels (ENaC), exhibiting airway surface dehydration (mucus hyperconcentration), impaired mucociliary clearance (MCC) and mucus plugging, led to a model of muco-obstructive lung disease that shares key features of CF and COPD. In this review, we summarize recent progress in the understanding of causes of impaired MCC and in vivo consequences of airway mucus obstruction that can be inferred from studies in βENaC-overexpressing mice. These studies confirm that mucus hyperconcentration on airway surfaces plays a critical role in the pathophysiology of impaired MCC, mucus adhesion and airway plugging that cause airflow obstruction and provide a nidus for bacterial infection. In addition, these studies support the emerging concept that excess airway mucus per se, probably via several mechanisms including hypoxic epithelial necrosis, retention of inhaled irritants or allergens, and potential immunomodulatory effects, is a potent trigger of chronic airway inflammation and associated lung damage, even in the absence of bacterial infection. Finally, these studies suggest that improvement of mucus clearance may be a promising therapeutic strategy for a spectrum of muco-obstructive lung diseases.
Collapse
|
259
|
Bergman P, Seyedoleslami Esfahani S, Engström Y. Drosophila as a Model for Human Diseases—Focus on Innate Immunity in Barrier Epithelia. Curr Top Dev Biol 2017; 121:29-81. [DOI: 10.1016/bs.ctdb.2016.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
260
|
Adenovirus-mediated Foxp3 expression in lung epithelial cells ameliorates acute radiation-induced pneumonitis in mice. Gene Ther 2016; 24:104-112. [PMID: 27996966 DOI: 10.1038/gt.2016.86] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 11/11/2016] [Accepted: 12/05/2016] [Indexed: 12/11/2022]
Abstract
Forkhead transcription factor 3 (Foxp3) has a critical role in regulatory T cells (Treg). There are an increasing number of researches concerning the functions of Foxp3 in other cells, including lung epithelial cells besides Treg. However, the roles of Foxp3 in lung epithelial cells remain poorly understood. To examine the potential therapeutic benefits of Foxp3 for lung inflammation, this study investigates the effect of adenovirus-mediated Foxp3 overexpression in a radiation-induced lung damage model. Foxp3-EGFP expressing adenovirus was administered by intratracheal injection three times over 14 days after focal X-ray irradiation. To evaluate effects of Foxp3 overexpression in radiation-induced lung inflammation, immune cell profiles of bronchoalveolar lavage (BAL) fluid were analyzed. Foxp3 gene-delivered mice showed significant inhibition of immune cell infiltration, such as eosinophils, lymphocytes, macrophages and neutrophils in BAL fluid. Histopathological analysis also showed that Foxp3 overexpression inhibits inflammatory cell recruitment and collagen deposition in lung tissues. In addition, expression of inflammatory and fibrosis-related genes was decreased in the Foxp3 expression adenovirus-infected group. These results suggest that Foxp3 expression in lungs holds considerable therapeutic potential for attenuating inflammation and fibrosis in radiation-induced lung injury.
Collapse
|
261
|
Cohen TS. Role of MicroRNA in the Lung's Innate Immune Response. J Innate Immun 2016; 9:243-249. [PMID: 27915347 DOI: 10.1159/000452669] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 10/18/2016] [Indexed: 12/22/2022] Open
Abstract
The immune response to respiratory pathogens must be robust enough to defend the host yet properly constrained such that inflammation-induced tissue damage is avoided. MicroRNA (miRNA) are small noncoding RNA which posttranscriptionally influence gene expression. In this review, we discuss recent experimental evidence of the contribution of miRNA to the lung's response to bacterial and viral pathogens.
Collapse
Affiliation(s)
- Taylor S Cohen
- Department of Infectious Disease, Medimmune, Gaithersburg, MD, USA
| |
Collapse
|
262
|
Fritzsching B, Hagner M, Dai L, Christochowitz S, Agrawal R, van Bodegom C, Schmidt S, Schatterny J, Hirtz S, Brown R, Goritzka M, Duerr J, Zhou-Suckow Z, Mall MA. Impaired mucus clearance exacerbates allergen-induced type 2 airway inflammation in juvenile mice. J Allergy Clin Immunol 2016; 140:190-203.e5. [PMID: 27865862 DOI: 10.1016/j.jaci.2016.09.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 08/22/2016] [Accepted: 09/05/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Type 2 airway inflammation plays a central role in the pathogenesis of allergen-induced asthma, but the underlying mechanisms remain poorly understood. Recently, we demonstrated that reduced mucociliary clearance, a characteristic feature of asthma, produces spontaneous type 2 airway inflammation in juvenile β-epithelial Na+ channel (Scnn1b)-transgenic (Tg) mice. OBJECTIVE We sought to determine the role of impaired mucus clearance in the pathogenesis of allergen-induced type 2 airway inflammation and identify cellular sources of the signature cytokine IL-13. METHODS We challenged juvenile Scnn1b-Tg and wild-type mice with Aspergillus fumigatus and house dust mite allergen and compared the effects on airway eosinophilia, type 2 cytokine levels, goblet cell metaplasia, and airway hyperresponsiveness. Furthermore, we determined cellular sources of IL-13 and effects of genetic deletion of the key type 2 signal-transducing molecule signal transducer and activator of transcription 6 (STAT6) and evaluated the effects of therapeutic improvement of mucus clearance. RESULTS Reduced mucociliary allergen clearance exacerbated Stat6-dependent secretion of type 2 cytokines, airway eosinophilia, and airway hyperresponsiveness in juvenile Scnn1b-Tg mice. IL-13 levels were increased in airway epithelial cells, macrophages, type 2 innate lymphoid cells, and TH2 cells along with increased Il33 expression in the airway epithelium of Scnn1b-Tg mice. Treatment with the epithelial Na+ channel blocker amiloride, improving airway surface hydration and mucus clearance, reduced allergen-induced inflammation in Scnn1b-Tg mice. CONCLUSION Our data support that impaired clearance of inhaled allergens triggering IL-13 production by multiple cell types in the airways plays an important role in the pathogenesis of type 2 airway inflammation and suggests therapeutic improvement of mucociliary clearance as a novel treatment strategy for children with allergen-induced asthma.
Collapse
Affiliation(s)
- Benedikt Fritzsching
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Matthias Hagner
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Lu Dai
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Sandra Christochowitz
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Raman Agrawal
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Charlotte van Bodegom
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Simone Schmidt
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Jolanthe Schatterny
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Stephanie Hirtz
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Ryan Brown
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Michelle Goritzka
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Julia Duerr
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Zhe Zhou-Suckow
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany
| | - Marcus A Mall
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
263
|
Amatngalim GD, Broekman W, Daniel NM, van der Vlugt LEPM, van Schadewijk A, Taube C, Hiemstra PS. Cigarette Smoke Modulates Repair and Innate Immunity following Injury to Airway Epithelial Cells. PLoS One 2016; 11:e0166255. [PMID: 27829065 PMCID: PMC5102360 DOI: 10.1371/journal.pone.0166255] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 10/25/2016] [Indexed: 11/18/2022] Open
Abstract
Cigarette smoking is the main risk factor associated with chronic obstructive pulmonary disease (COPD), and contributes to COPD development and progression by causing epithelial injury and inflammation. Whereas it is known that cigarette smoke (CS) may affect the innate immune function of airway epithelial cells and epithelial repair, this has so far not been explored in an integrated design using mucociliary differentiated airway epithelial cells. In this study, we examined the effect of whole CS exposure on wound repair and the innate immune activity of mucociliary differentiated primary bronchial epithelial cells, upon injury induced by disruption of epithelial barrier integrity or by mechanical wounding. Upon mechanical injury CS caused a delayed recovery in the epithelial barrier integrity and wound closure. Furthermore CS enhanced innate immune responses, as demonstrated by increased expression of the antimicrobial protein RNase 7. These differential effects on epithelial repair and innate immunity were both mediated by CS-induced oxidative stress. Overall, our findings demonstrate modulation of wound repair and innate immune responses of injured airway epithelial cells that may contribute to COPD development and progression.
Collapse
Affiliation(s)
- Gimano D Amatngalim
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Winifred Broekman
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nadia M Daniel
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Christian Taube
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
264
|
Anas AA, Yang J, Daan de Boer J, Roelofs JJTH, Hou B, de Vos AF, van der Poll T. General, but not myeloid or type II lung epithelial cell, myeloid differentiation factor 88 deficiency abrogates house dust mite induced allergic lung inflammation. Clin Exp Immunol 2016; 187:204-212. [PMID: 27625307 DOI: 10.1111/cei.12867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2016] [Indexed: 12/27/2022] Open
Abstract
Asthma is a highly prevalent chronic allergic inflammatory disease of the airways affecting people worldwide. House dust mite (HDM) is the most common allergen implicated in human allergic asthma. HDM-induced allergic responses are thought to depend upon activation of pathways involving Toll-like receptors and their adaptor protein myeloid differentiation factor 88 (MyD88). We sought here to determine the role of MyD88 in myeloid and type II lung epithelial cells in the development of asthma-like allergic disease using a mouse model. Repeated exposure to HDM caused allergic responses in control mice characterized by influx of eosinophils into the bronchoalveolar space and lung tissue, lung pathology and mucus production and protein leak into bronchoalveolar lavage fluid. All these responses were abrogated in mice with a general deficiency of MyD88 but unaltered in mice with MyD88 deficiency, specifically in myeloid or type II lung epithelial cells. We conclude that cells other than myeloid or type II lung epithelial cells are responsible for MyD88-dependent HDM-induced allergic airway inflammation.
Collapse
Affiliation(s)
- A A Anas
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - J Yang
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - J Daan de Boer
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - J J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - B Hou
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chaoyang District, Beijing, China
| | - A F de Vos
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - T van der Poll
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.,Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
265
|
Hiemstra PS, Bals R. Basic science of electronic cigarettes: assessment in cell culture and in vivo models. Respir Res 2016; 17:127. [PMID: 27717371 PMCID: PMC5055681 DOI: 10.1186/s12931-016-0447-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/05/2016] [Indexed: 12/20/2022] Open
Abstract
Electronic cigarettes (e-cigarettes, ECIGs) were introduced into the market a decade ago as an alternative to tobacco smoking. Whether ECIGs are safe and whether they qualify as smoking cessation tool is currently unknown. Their use has markedly expanded in that period, despite the fact that potential toxic effects of the vapour created by the e-cigarette and the nicotine-containing cartridge fluid have been incompletely studied. Marketing targets diverse groups including older smokers but also young people. Whereas the adverse health effects of nicotine inhaled by users of ECIGs has been well documented, less is known about the other components. An increasing number of in vitro and in vivo studies demonstrate a range of adverse effects of both the vapour created by ECIGs as well as the nicotine-containing fluid. Importantly, these studies demonstrate that toxicity from ECIGs, although this may be less than that caused by tobacco products, not only arises from its nicotine content. Furthermore, there are no data on the long-term consequences of ECIG use. The wide range of ECIG products available to consumers and the lack of standardisation of toxicological approaches towards ECIG evaluation complicates the assessment of adverse health effects of their use. Here we review the current data on preclinical studies on ECIGs describing their effects in cell culture and animal models.
Collapse
Affiliation(s)
- Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert Bals
- Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421, Homburg, Germany.
| |
Collapse
|
266
|
Short Palate, Lung, and Nasal Epithelial Clone 1 Has Antimicrobial and Antibiofilm Activities against the Burkholderia cepacia Complex. Antimicrob Agents Chemother 2016; 60:6003-12. [PMID: 27458217 DOI: 10.1128/aac.00975-16] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/17/2016] [Indexed: 02/07/2023] Open
Abstract
The opportunistic bacteria of the Burkholderia cepacia complex (Bcc) are extremely pathogenic to cystic fibrosis (CF) patients, and acquisition of Bcc bacteria is associated with a significant increase in mortality. Treatment of Bcc infections is difficult because the bacteria are multidrug resistant and able to survive in biofilms. Short palate, lung, and nasal epithelial clone 1 (SPLUNC1) is an innate defense protein that is secreted by the upper airways and pharynx. While SPLUNC1 is known to have antimicrobial functions, its effects on Bcc strains are unclear. We therefore tested the hypothesis that SPLUNC1 is able to impair Bcc growth and biofilm formation. We found that SPLUNC1 exerted bacteriostatic effects against several Bcc clinical isolates, including B. cenocepacia strain J2315 (50% inhibitory concentration [IC50] = 0.28 μM), and reduced biofilm formation and attachment (IC50 = 0.11 μM). We then determined which domains of SPLUNC1 are responsible for its antimicrobial activity. Deletions of SPLUNC1's N terminus and α6 helix did not affect its function. However, deletion of the α4 helix attenuated antimicrobial activity, while the corresponding α4 peptide displayed antimicrobial activity. Chronic neutrophilia is a hallmark of CF lung disease, and neutrophil elastase (NE) cleaves SPLUNC1. However, we found that the ability of SPLUNC1 to disrupt biofilm formation was significantly potentiated by NE pretreatment. While the impact of CF on SPLUNC1-Bcc interactions is not currently known, our data suggest that understanding this interaction may have important implications for CF lung disease.
Collapse
|
267
|
Sanak M. Eicosanoid Mediators in the Airway Inflammation of Asthmatic Patients: What is New? ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2016; 8:481-90. [PMID: 27582398 PMCID: PMC5011047 DOI: 10.4168/aair.2016.8.6.481] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 06/09/2016] [Accepted: 06/09/2016] [Indexed: 12/31/2022]
Abstract
Lipid mediators contribute to inflammation providing both pro-inflammatory signals and terminating the inflammatory process by activation of macrophages. Among the most significant biologically lipid mediators, these are produced by free-radical or enzymatic oxygenation of arachidonic acid named "eicosanoids". There were some novel eicosanoids identified within the last decade, and many of them are measurable in clinical samples by affordable chromatography-mass spectrometry equipment or sensitive immunoassays. In this review, we present some recent advances in understanding of the signaling by eicosanoid mediators during asthmatic airway inflammation. Eicosanoid profiling in the exhaled breath condensate, induced sputum, or their metabolites measurements in urine is complementary to the cellular phenotyping of asthmatic inflammation. Special attention is paid to aspirin-exacerbated respiratory disease, a phenotype of asthma manifested by the most profound changes in the profile of eicosanoids produced. A hallmark of this type of asthma with hypersensitivity to non-steroid anti-inflammatory drugs (NSAIDs) is to increase biosynthesis of cysteinyl leukotrienes on the systemic level. It depends on transcellular biosynthesis of leukotriene C4 by platelets that adhere to granulocytes releasing leukotriene A4. However, other abnormalities are also reported in this type of asthma as a resistance to anti-inflammatory activity of prostaglandin E2 or a robust eosinophil interferon-γ response resulting in cysteinyl leukotrienes production. A novel mechanism is also discussed in which an isoprostane structurally related to prostaglandin E2 is released into exhaled breath condensate during a provoked asthmatic attack. However, it is concluded that any single eicosanoid or even their complex profile can hardly provide a thorough explanation for the mechanism of asthmatic inflammation.
Collapse
Affiliation(s)
- Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
268
|
Affiliation(s)
- Renat Shaykhiev
- Department of Genetic Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
269
|
Blume C, David J, Bell RE, Laver JR, Read RC, Clark GC, Davies DE, Swindle EJ. Modulation of Human Airway Barrier Functions during Burkholderia thailandensis and Francisella tularensis Infection Running Title: Airway Barrier Functions during Bacterial Infections. Pathogens 2016; 5:pathogens5030053. [PMID: 27527221 PMCID: PMC5039433 DOI: 10.3390/pathogens5030053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 07/25/2016] [Accepted: 07/26/2016] [Indexed: 11/16/2022] Open
Abstract
The bronchial epithelium provides protection against pathogens from the inhaled environment through the formation of a highly-regulated barrier. In order to understand the pulmonary diseases melioidosis and tularemia caused by Burkholderia thailandensis and Fransicella tularensis, respectively, the barrier function of the human bronchial epithelium were analysed. Polarised 16HBE14o- or differentiated primary human bronchial epithelial cells (BECs) were exposed to increasing multiplicities of infection (MOI) of B. thailandensis or F. tularensis Live Vaccine Strain and barrier responses monitored over 24-72 h. Challenge of polarized BECs with either bacterial species caused an MOI- and time-dependent increase in ionic permeability, disruption of tight junctions, and bacterial passage from the apical to the basolateral compartment. B. thailandensis was found to be more invasive than F. tularensis. Both bacterial species induced an MOI-dependent increase in TNF-α release. An increase in ionic permeability and TNF-α release was induced by B. thailandensis in differentiated BECs. Pretreatment of polarised BECs with the corticosteroid fluticasone propionate reduced bacterial-dependent increases in ionic permeability, bacterial passage, and TNF-α release. TNF blocking antibody Enbrel(®) reduced bacterial passage only. BEC barrier properties are disrupted during respiratory bacterial infections and targeting with corticosteroids or anti-TNF compounds may represent a therapeutic option.
Collapse
Affiliation(s)
- Cornelia Blume
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK.
| | - Jonathan David
- Defence Science and Technology Laboratory (Dstl), Porton Down, Salisbury SP4 0JQ, UK.
| | - Rachel E Bell
- Defence Science and Technology Laboratory (Dstl), Porton Down, Salisbury SP4 0JQ, UK.
- Centre for Molecular and Cellular Biology of Inflammation, Guy's Campus, King's College London, London SE1 1UL, UK.
| | - Jay R Laver
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK.
| | - Robert C Read
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK.
- Southampton NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton SO16 6YD, UK.
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Graeme C Clark
- Defence Science and Technology Laboratory (Dstl), Porton Down, Salisbury SP4 0JQ, UK.
| | - Donna E Davies
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK.
- Southampton NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton SO16 6YD, UK.
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Emily J Swindle
- Academic Unit of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, University Hospital Southampton, Tremona Road, Southampton SO16 6YD, UK.
- Southampton NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton SO16 6YD, UK.
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
270
|
Stolarczyk M, Amatngalim GD, Yu X, Veltman M, Hiemstra PS, Scholte BJ. ADAM17 and EGFR regulate IL-6 receptor and amphiregulin mRNA expression and release in cigarette smoke-exposed primary bronchial epithelial cells from patients with chronic obstructive pulmonary disease (COPD). Physiol Rep 2016; 4:e12878. [PMID: 27561911 PMCID: PMC5002905 DOI: 10.14814/phy2.12878] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/08/2016] [Accepted: 07/09/2016] [Indexed: 01/31/2023] Open
Abstract
Aberrant activity of a disintegrin and metalloprotease 17 (ADAM17), also known as TACE, and epidermal growth factor receptor (EGFR) has been suggested to contribute to chronic obstructive pulmonary disease (COPD) development and progression. The aim of this study was to investigate the role of these proteins in activation of primary bronchial epithelial cells differentiated at the air-liquid interface (ALI-PBEC) by whole cigarette smoke (CS), comparing cells from COPD patients with non-COPD CS exposure of ALI-PBEC enhanced ADAM17-mediated shedding of the IL-6 receptor (IL6R) and the EGFR agonist amphiregulin (AREG) toward the basolateral compartment, which was more pronounced in cells from COPD patients than in non-COPD controls. CS transiently increased IL6R and AREG mRNA in ALI-PBEC to a similar extent in cultures from both groups, suggesting that posttranslational events determine differential shedding between COPD and non-COPD cultures. We show for the first time by in situ proximity ligation (PLA) that CS strongly enhances interactions of phosphorylated ADAM17 with AREG and IL-6R in an intracellular compartment, suggesting that CS-induced intracellular trafficking events precede shedding to the extracellular compartment. Both EGFR and ADAM17 activity contribute to CS-induced IL-6R and AREG protein shedding and to mRNA expression, as demonstrated using selective inhibitors (AG1478 and TMI-2). Our data are consistent with an autocrine-positive feedback mechanism in which CS triggers shedding of EGFR agonists evoking EGFR activation, in ADAM17-dependent manner, and subsequently transduce paracrine signaling toward myeloid cells and connective tissue. Reducing ADAM17 and EGFR activity could therefore be a therapeutic approach for the tissue remodeling and inflammation observed in COPD.
Collapse
Affiliation(s)
| | - Gimano D Amatngalim
- Pulmonology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Xiao Yu
- Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Mieke Veltman
- Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Pieter S Hiemstra
- Pulmonology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Bob J Scholte
- Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
271
|
Lee H, Zhang D, Minhas J, Jin Y. Extracellular Vesicles Facilitate the Intercellular Communications in the Pathogenesis of Lung Injury. ACTA ACUST UNITED AC 2016; 5. [PMID: 27722038 DOI: 10.4172/2168-9296.1000175] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are a group of heterogeneous, nano-sized structures surrounded by lipid bilayer membranes that are released by cells. Depending on their size and mechanisms of formation, EVs are often referred to as exosomes, microvesicles (MVs) and apoptotic bodies (AB). EVs are evolutionally conserved vesicles that mediate intercellular communications and cross-talk, via transferring proteins, lipids and nucleic acids. Accumulating evidence suggests that EVs exert essential physiological and pathological functions on both their mother and recipient cells. Therefore, growing interests focus on the potentials of EVs to serve as novel targets for the development of therapeutic and diagnostic strategies. Currently, extensive reports are yielded from cancer research. However, besides malignancy, EVs may also serve as crucial regulators in other devastating conditions, such as the acute respiratory distress syndrome (ARDS) and acute lung injury (ALI). The generation, regulation and function of EVs in ARDS/ALI are largely unexplored. In this mini review, we will briefly review the current understanding of EVs and their known physiological/pathological functions in the pathogenesis of ARDS/ALI. Previously, only scattered reports have been published in this field. We believe that further investigations focusing on EVs and their compositions will shed light on novel insights in the research of ARDS/ALI.
Collapse
Affiliation(s)
- Heedoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, USA
| | - Duo Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, USA
| | - Jasleen Minhas
- Internal Medicine, North Shore Medical Center, Salem Hospital, 81 Highland Ave, Salem, USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, USA
| |
Collapse
|
272
|
Ralhan A, Laval J, Lelis F, Ballbach M, Grund C, Hector A, Hartl D. Current Concepts and Controversies in Innate Immunity of Cystic Fibrosis Lung Disease. J Innate Immun 2016; 8:531-540. [PMID: 27362371 PMCID: PMC6738757 DOI: 10.1159/000446840] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/17/2016] [Accepted: 05/17/2016] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) lung disease is characterized by chronic infection and inflammation. The inflammatory response in CF is dominated by the activation of the innate immune system. Bacteria and fungi represent the key pathogens chronically colonizing the CF airways. In response, innate immune pattern recognition receptors, expressed by airway epithelial and myeloid cells, sense the microbial threat and release chemoattractants to recruit large numbers of neutrophils into CF airways. However, neutrophils fail to efficiently clear the invading pathogens, but instead release harmful proteases and oxidants and finally cause tissue injury. Here, we summarize and discuss current concepts and controversies in the field of innate immunity in CF lung disease, facing the ongoing questions of whether inflammation is good or bad in CF and how innate immune mechanisms could be harnessed therapeutically.
Collapse
Affiliation(s)
- Anjali Ralhan
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Julie Laval
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Felipe Lelis
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Marlene Ballbach
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Charlotte Grund
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Andreas Hector
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
| | - Dominik Hartl
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany
- Immunology, Inflammation and Infectious Diseases (I3) Discovery and Translational Area, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
273
|
Menzel M, Akbarshahi H, Bjermer L, Uller L. Azithromycin induces anti-viral effects in cultured bronchial epithelial cells from COPD patients. Sci Rep 2016; 6:28698. [PMID: 27350308 PMCID: PMC4923851 DOI: 10.1038/srep28698] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 06/08/2016] [Indexed: 12/02/2022] Open
Abstract
Rhinovirus infection is a major cause of chronic obstructive pulmonary disease (COPD) exacerbations and may contribute to the development into severe stages of COPD. The macrolide antibiotic azithromycin may exert anti-viral actions and has been reported to reduce exacerbations in COPD. However, little is known about its anti-viral actions on bronchial epithelial cells at clinically relevant concentrations. Primary bronchial epithelial cells from COPD donors and healthy individuals were treated continuously with azithromycin starting 24 h before infection with rhinovirus RV16. Expression of interferons, RIG-I like helicases, pro-inflammatory cytokines and viral load were analysed. Azithromycin transiently increased expression of IFNβ and IFNλ1 and RIG-I like helicases in un-infected COPD cells. Further, azithromycin augmented RV16-induced expression of interferons and RIG-I like helicases in COPD cells but not in healthy epithelial cells. Azithromycin also decreased viral load. However, it only modestly altered RV16-induced pro-inflammatory cytokine expression. Adding budesonide did not reduce interferon-inducing effects of azithromycin. Possibly by inducing expression of RIG-I like helicases, azithromycin increased rhinovirus-induced expression of interferons in COPD but not in healthy bronchial epithelium. These effects would reduce bronchial viral load, supporting azithromycin’s emerging role in prevention of exacerbations of COPD.
Collapse
Affiliation(s)
- Mandy Menzel
- Respiratory Immunopharmacology, Department of Experimental Medical Science, Lund University, Sweden
| | - Hamid Akbarshahi
- Respiratory Immunopharmacology, Department of Experimental Medical Science, Lund University, Sweden
| | - Leif Bjermer
- Lung medicine and Allergology, Department of Clinical Sciences, Lund University, Sweden
| | - Lena Uller
- Respiratory Immunopharmacology, Department of Experimental Medical Science, Lund University, Sweden
| |
Collapse
|
274
|
Epithelial Anion Transport as Modulator of Chemokine Signaling. Mediators Inflamm 2016; 2016:7596531. [PMID: 27382190 PMCID: PMC4921137 DOI: 10.1155/2016/7596531] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 05/03/2016] [Accepted: 05/12/2016] [Indexed: 12/16/2022] Open
Abstract
The pivotal role of epithelial cells is to secrete and absorb ions and water in order to allow the formation of a luminal fluid compartment that is fundamental for the epithelial function as a barrier against environmental factors. Importantly, epithelial cells also take part in the innate immune system. As a first line of defense they detect pathogens and react by secreting and responding to chemokines and cytokines, thus aggravating immune responses or resolving inflammatory states. Loss of epithelial anion transport is well documented in a variety of diseases including cystic fibrosis, chronic obstructive pulmonary disease, asthma, pancreatitis, and cholestatic liver disease. Here we review the effect of aberrant anion secretion with focus on the release of inflammatory mediators by epithelial cells and discuss putative mechanisms linking these transport defects to the augmented epithelial release of chemokines and cytokines. These mechanisms may contribute to the excessive and persistent inflammation in many respiratory and gastrointestinal diseases.
Collapse
|
275
|
Smits HH, van der Vlugt LE, von Mutius E, Hiemstra PS. Childhood allergies and asthma: New insights on environmental exposures and local immunity at the lung barrier. Curr Opin Immunol 2016; 42:41-47. [PMID: 27254380 DOI: 10.1016/j.coi.2016.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 12/11/2022]
Abstract
While certain bacteria and respiratory viruses promote local inflammation and disease onset, a more diverse colonization of the different species in the (gut) microbiome may be linked to more regulatory responses and protection against asthma and allergies. These processes are also influenced in part by food intake, both targeting the composition of the gut microbiome and influencing the immune system via metabolites. Early life environmental microbial exposure also contributes to protection against asthma and allergy and is linked with an early activation of the innate immune system and the development of regulatory immune responses. Although greater mechanistic insight is needed, it is tempting to speculate that part of the environmental effect can be explained by modulation of the microbiome composition at mucosal surfaces, epithelial barrier function and/or local immunity. A review of the latest studies is provided.
Collapse
Affiliation(s)
- Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Luciën Epm van der Vlugt
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands; Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erika von Mutius
- Dr von Hauner Children's Hospital, Ludwig Maximilians University of Munich, Munich, Germany; Comprehensive Pneumology Centre Munich (CPC-M), Member of the German Center for Lung Research, Germany
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
276
|
Zarcone MC, Duistermaat E, van Schadewijk A, Jedynska A, Hiemstra PS, Kooter IM. Cellular response of mucociliary differentiated primary bronchial epithelial cells to diesel exhaust. Am J Physiol Lung Cell Mol Physiol 2016; 311:L111-23. [PMID: 27190060 DOI: 10.1152/ajplung.00064.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/17/2016] [Indexed: 12/22/2022] Open
Abstract
Diesel emissions are the main source of air pollution in urban areas, and diesel exposure is linked with substantial adverse health effects. In vitro diesel exposure models are considered a suitable tool for understanding these effects. Here we aimed to use a controlled in vitro exposure system to whole diesel exhaust to study the effect of whole diesel exhaust concentration and exposure duration on mucociliary differentiated human primary bronchial epithelial cells (PBEC). PBEC cultured at the air-liquid interface were exposed for 60 to 375 min to three different dilutions of diesel exhaust (DE). The DE mixture was generated by an engine at 47% load, and characterized for particulate matter size and distribution and chemical and gas composition. Cytotoxicity and epithelial barrier function was assessed, as well as mRNA expression and protein release analysis. DE caused a significant dose-dependent increase in expression of oxidative stress markers (HMOX1 and NQO1; n = 4) at 6 h after 150 min exposure. Furthermore, DE significantly increased the expression of the markers of the integrated stress response CHOP and GADD34 and of the proinflammatory chemokine CXCL8, as well as release of CXCL8 protein. Cytotoxic effects or effects on epithelial barrier function were observed only after prolonged exposures to the highest DE dose. These results demonstrate the suitability of our model and that exposure dose and duration and time of analysis postexposure are main determinants for the effects of DE on differentiated primary human airway epithelial cells.
Collapse
Affiliation(s)
- Maria C Zarcone
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands;
| | - Evert Duistermaat
- Netherlands Organization for Applied Scientific Research, Zeist, The Netherlands; and
| | | | - Aleksandra Jedynska
- Netherlands Organization for Applied Scientific Research Utrecht, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ingeborg M Kooter
- Netherlands Organization for Applied Scientific Research Utrecht, The Netherlands
| |
Collapse
|
277
|
Kadiyala V, Sasse SK, Altonsy MO, Berman R, Chu HW, Phang TL, Gerber AN. Cistrome-based Cooperation between Airway Epithelial Glucocorticoid Receptor and NF-κB Orchestrates Anti-inflammatory Effects. J Biol Chem 2016; 291:12673-12687. [PMID: 27076634 DOI: 10.1074/jbc.m116.721217] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Indexed: 12/11/2022] Open
Abstract
Antagonism of pro-inflammatory transcription factors by monomeric glucocorticoid receptor (GR) has long been viewed as central to glucocorticoid (GC) efficacy. However, the mechanisms and targets through which GCs exert therapeutic effects in diseases such as asthma remain incompletely understood. We previously defined a surprising cooperative interaction between GR and NF-κB that enhanced expression of A20 (TNFAIP3), a potent inhibitor of NF-κB. Here we extend this observation to establish that A20 is required for maximal cytokine repression by GCs. To ascertain the global extent of GR and NF-κB cooperation, we determined genome-wide occupancy of GR, the p65 subunit of NF-κB, and RNA polymerase II in airway epithelial cells treated with dexamethasone, TNF, or both using chromatin immunoprecipitation followed by deep sequencing. We found that GR recruits p65 to dimeric GR binding sites across the genome and discovered additional regulatory elements in which GR-p65 cooperation augments gene expression. GR targets regulated by this mechanism include key anti-inflammatory and injury response genes such as SERPINA1, which encodes α1 antitrypsin, and FOXP4, an inhibitor of mucus production. Although dexamethasone treatment reduced RNA polymerase II occupancy of TNF targets such as IL8 and TNFAIP2, we were unable to correlate specific binding sequences for GR or occupancy patterns with repressive effects on transcription. Our results suggest that cooperative anti-inflammatory gene regulation by GR and p65 contributes to GC efficacy, whereas tethering interactions between GR and p65 are not universally required for GC-based gene repression.
Collapse
Affiliation(s)
- Vineela Kadiyala
- From the Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Sarah K Sasse
- From the Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Mohammed O Altonsy
- From the Department of Medicine, National Jewish Health, Denver, Colorado 80206,; Department of Zoology, Sohag University, Sohag 825224, Egypt, and
| | - Reena Berman
- From the Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Hong W Chu
- From the Department of Medicine, National Jewish Health, Denver, Colorado 80206
| | - Tzu L Phang
- Department of Medicine, University of Colorado, Denver, Colorado 80045
| | - Anthony N Gerber
- From the Department of Medicine, National Jewish Health, Denver, Colorado 80206,; Department of Medicine, University of Colorado, Denver, Colorado 80045.
| |
Collapse
|
278
|
Reuter S, Beckert H, Taube C. Take the Wnt out of the inflammatory sails: modulatory effects of Wnt in airway diseases. J Transl Med 2016; 96:177-85. [PMID: 26595171 DOI: 10.1038/labinvest.2015.143] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/08/2015] [Accepted: 09/28/2015] [Indexed: 12/11/2022] Open
Abstract
Bronchial asthma and chronic obstructive pulmonary disease (COPD) are chronic diseases that are associated with inflammation and structural changes in the airways and lungs. Recent findings have implicated Wnt pathways in critically regulating inflammatory responses, especially in asthma. Furthermore, canonical and noncanonical Wnt pathways are involved in structural changes such as airway remodeling, goblet cell metaplasia, and airway smooth muscle (ASM) proliferation. In COPD, Wnt pathways are not only associated with structural changes in the airways but also involved in the development of emphysema. The present review summarizes the role and function of the canonical and noncanonical Wnt pathway with regard to airway inflammation and structural changes in asthma and COPD. Further identification of the role and function of different Wnt molecules and pathways could help to develop novel therapeutic options for these diseases.
Collapse
Affiliation(s)
- Sebastian Reuter
- Priority Area Asthma and Allergy, Research Center Borstel, Airway Research Center North, Member of the German Center for Lung Research, Borstel, Germany
| | - Hendrik Beckert
- III Medical Clinic, University Medical Center, Mainz, Germany
| | - Christian Taube
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
279
|
Hiemstra PS, Amatngalim GD, van der Does AM, Taube C. Antimicrobial Peptides and Innate Lung Defenses: Role in Infectious and Noninfectious Lung Diseases and Therapeutic Applications. Chest 2016; 149:545-551. [PMID: 26502035 DOI: 10.1378/chest.15-1353] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/31/2015] [Accepted: 09/21/2015] [Indexed: 11/01/2022] Open
Abstract
Respiratory infections are a major clinical problem, and treatment is increasingly complicated by the emergence of microbial antibiotic resistance. Development of new antibiotics is notoriously costly and slow; therefore, alternative strategies are needed. Antimicrobial peptides, central effector molecules of the immune system, are being considered as alternatives to conventional antibiotics. These peptides display a range of activities, including not only direct antimicrobial activity, but also immunomodulation and wound repair. In the lung, airway epithelial cells and neutrophils in particular contribute to their synthesis. The relevance of antimicrobial peptides for host defense against infection has been demonstrated in animal models and is supported by observations in patient studies, showing altered expression and/or unfavorable circumstances for their action in a variety of lung diseases. Importantly, antimicrobial peptides are active against microorganisms that are resistant against conventional antibiotics, including multidrug-resistant bacteria. Several strategies have been proposed to use these peptides in the treatment of infections, including direct administration of antimicrobial peptides, enhancement of their local production, and creation of more favorable circumstances for their action. In this review, recent developments in antimicrobial peptides research in the lung and clinical applications for novel therapies of lung diseases are discussed.
Collapse
Affiliation(s)
- Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Gimano D Amatngalim
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne M van der Does
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christian Taube
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
280
|
Gela A, Bhongir RKV, Mori M, Keenan P, Mörgelin M, Erjefält JS, Herwald H, Egesten A, Kasetty G. Osteopontin That Is Elevated in the Airways during COPD Impairs the Antibacterial Activity of Common Innate Antibiotics. PLoS One 2016; 11:e0146192. [PMID: 26731746 PMCID: PMC4712133 DOI: 10.1371/journal.pone.0146192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 12/14/2015] [Indexed: 11/18/2022] Open
Abstract
Bacterial infections of the respiratory tract contribute to exacerbations and disease progression in chronic obstructive pulmonary disease (COPD). There is also an increased risk of invasive pneumococcal disease in COPD. The underlying mechanisms are not fully understood but include impaired mucociliary clearance and structural remodeling of the airways. In addition, antimicrobial proteins that are constitutively expressed or induced during inflammatory conditions are an important part of the airway innate host defense. In the present study, we show that osteopontin (OPN), a multifunctional glycoprotein that is highly upregulated in the airways of COPD patients co-localizes with several antimicrobial proteins expressed in the airways. In vitro, OPN bound lactoferrin, secretory leukocyte peptidase inhibitor (SLPI), midkine, human beta defensin-3 (hBD-3), and thymic stromal lymphopoietin (TSLP) but showed low or no affinity for lysozyme and LL-37. Binding of OPN impaired the antibacterial activity against the important bacterial pathogens Streptococcus pneumoniae and Pseudomonas aeruginosa. Interestingly, OPN reduced lysozyme-induced killing of S. pneumoniae, a finding that could be explained by binding of OPN to the bacterial surface, thereby shielding the bacteria. A fragment of OPN generated by elastase of P. aeruginosa retained some inhibitory effect. Some antimicrobial proteins have additional functions. However, the muramidase-activity of lysozyme and the protease inhibitory function of SLPI were not affected by OPN. Taken together, OPN can contribute to the impairment of innate host defense by interfering with the function of antimicrobial proteins, thus increasing the vulnerability to acquire infections during COPD.
Collapse
Affiliation(s)
- Anele Gela
- Respiratory Medicine & Allergology, Department of Clinical Sciences Lund, Lund University, SE-221 84, Lund, Sweden
| | - Ravi K. V. Bhongir
- Respiratory Medicine & Allergology, Department of Clinical Sciences Lund, Lund University, SE-221 84, Lund, Sweden
| | - Michiko Mori
- Airway Inflammation Unit, Department of Experimental Medical Sciences, Lund University, SE-221 84, Lund, Sweden
| | - Paul Keenan
- Respiratory Medicine & Allergology, Department of Clinical Sciences Lund, Lund University, SE-221 84, Lund, Sweden
| | - Matthias Mörgelin
- Infection Medicine, Department of Clinical Sciences Lund, Lund University, SE-221 84, Lund, Sweden
| | - Jonas S. Erjefält
- Airway Inflammation Unit, Department of Experimental Medical Sciences, Lund University, SE-221 84, Lund, Sweden
| | - Heiko Herwald
- Infection Medicine, Department of Clinical Sciences Lund, Lund University, SE-221 84, Lund, Sweden
| | - Arne Egesten
- Respiratory Medicine & Allergology, Department of Clinical Sciences Lund, Lund University, SE-221 84, Lund, Sweden
| | - Gopinath Kasetty
- Respiratory Medicine & Allergology, Department of Clinical Sciences Lund, Lund University, SE-221 84, Lund, Sweden
- * E-mail:
| |
Collapse
|
281
|
Saglani S, Lloyd CM. Novel concepts in airway inflammation and remodelling in asthma. Eur Respir J 2015; 46:1796-804. [PMID: 26541520 DOI: 10.1183/13993003.01196-2014] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/25/2015] [Indexed: 02/03/2023]
Abstract
The hallmark pathological features of asthma include airway eosinophilic inflammation and structural changes (remodelling) which are associated with an irreversible loss in lung function that tracks from childhood to adulthood. In parallel with changes in function, pathological abnormalities occur early, during the pre-school years, are established by school age and subsequently remain (even though symptoms may remit for periods during adulthood). Given the equal importance of inflammation and remodelling in asthma pathogenesis, there is a significant disparity in studies undertaken to investigate the contribution of each. The majority focus on the role of inflammation, and although novel therapeutics such as those targeted against T-helper cell type 2 (Th2) mediators have arisen, it is apparent that targeting inflammation alone has not allowed disease modification. Therefore, unless airway remodelling is addressed for future therapeutic strategies, it is unlikely that we will progress towards a cure for asthma. Having acknowledged these limitations, the focus of this review is to highlight the gaps in our current knowledge about the mechanisms underlying airway remodelling, the relationships between remodelling, inflammation and function, remodelling and clinical phenotypes, and the importance of utilising innovative and realistic pre-clinical models to uncover effective, disease-modifying therapeutic strategies.
Collapse
Affiliation(s)
- Sejal Saglani
- Inflammation, Repair and Development Section, National Heart & Lung Institute, Imperial College London, London, UK Dept of Respiratory Paediatrics, Royal Brompton Hospital, London, UK
| | - Clare M Lloyd
- Inflammation, Repair and Development Section, National Heart & Lung Institute, Imperial College London, London, UK
| |
Collapse
|
282
|
Hiemstra PS. Parallel activities and interactions between antimicrobial peptides and complement in host defense at the airway epithelial surface. Mol Immunol 2015; 68:28-30. [DOI: 10.1016/j.molimm.2015.07.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 07/23/2015] [Accepted: 07/24/2015] [Indexed: 01/28/2023]
|