151
|
Chaput C, Sander LE, Suttorp N, Opitz B. NOD-Like Receptors in Lung Diseases. Front Immunol 2013; 4:393. [PMID: 24312100 PMCID: PMC3836004 DOI: 10.3389/fimmu.2013.00393] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 11/07/2013] [Indexed: 12/15/2022] Open
Abstract
The lung is a particularly vulnerable organ at the interface of the body and the exterior environment. It is constantly exposed to microbes and particles by inhalation. The innate immune system needs to react promptly and adequately to potential dangers posed by these microbes and particles, while at the same time avoiding extensive tissue damage. Nucleotide-binding oligomerization domain-like receptors (NLRs) represent a group of key sensors for microbes and damage in the lung. As such they are important players in various infectious as well as acute and chronic sterile inflammatory diseases, such as pneumonia, chronic obstructive pulmonary disease (COPD), acute lung injury/acute respiratory distress syndrome, pneumoconiosis, and asthma. Activation of most known NLRs leads to the production and release of pro-inflammatory cytokines, and/or to the induction of cell death. We will review NLR functions in the lung during infection and sterile inflammation.
Collapse
Affiliation(s)
- Catherine Chaput
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin , Berlin , Germany
| | | | | | | |
Collapse
|
152
|
Herold S, Gabrielli NM, Vadász I. Novel concepts of acute lung injury and alveolar-capillary barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 2013; 305:L665-81. [PMID: 24039257 DOI: 10.1152/ajplung.00232.2013] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this review we summarize recent major advances in our understanding on the molecular mechanisms, mediators, and biomarkers of acute lung injury (ALI) and alveolar-capillary barrier dysfunction, highlighting the role of immune cells, inflammatory and noninflammatory signaling events, mechanical noxae, and the affected cellular and molecular entities and functions. Furthermore, we address novel aspects of resolution and repair of ALI, as well as putative candidates for treatment of ALI, including pharmacological and cellular therapeutic means.
Collapse
Affiliation(s)
- Susanne Herold
- Dept. of Internal Medicine, Justus Liebig Univ., Universities of Giessen and Marburg Lung Center, Klinikstrasse 33, 35392 Giessen, Germany.
| | | | | |
Collapse
|
153
|
Fröhlich S, Murphy N, Ryan D, Boylan JF. Acute respiratory distress syndrome: current concepts and future directions. Anaesth Intensive Care 2013; 41:463-72. [PMID: 23808504 DOI: 10.1177/0310057x1304100405] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Acute respiratory distress syndrome is one of the leading causes of death in critically ill patients. Recent advances in supportive care have led to a moderate improvement in mortality. In particular, a much lower mortality rate than expected was evident in the severest category of patients (requiring extracorporeal membrane oxygenation) in Australia during the recent H1N1 pandemic. Though improvements in supportive care may have provided some benefit, there remains an absence of effective biological agents that are necessary to achieve further incremental reduction in mortality. This article will review the evidence available for current treatment strategies and discuss future research directions that may eventually improve outcomes in this important global disease.
Collapse
Affiliation(s)
- S Fröhlich
- Department of Anaesthesia and Intensive Care Medicine, St Vincent's University Hospital, Dublin, Ireland.
| | | | | | | |
Collapse
|
154
|
Inflammation and immune response in COPD: where do we stand? Mediators Inflamm 2013; 2013:413735. [PMID: 23956502 PMCID: PMC3728539 DOI: 10.1155/2013/413735] [Citation(s) in RCA: 149] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 07/02/2013] [Indexed: 01/05/2023] Open
Abstract
Increasing evidence indicates that chronic inflammatory and immune responses play key roles in the development and progression of COPD. Recent data provide evidence for a role in the NLRP3 inflammasome in the airway inflammation observed in COPD. Cigarette smoke activates innate immune cells by triggering pattern recognition receptors (PRRs) to release “danger signal”. These signals act as ligands to Toll-like receptors (TLRs), triggering the production of cytokines and inducing innate inflammation. In smokers who develop COPD there appears to be a specific pattern of inflammation in the airways and parenchyma as a result of both innate and adaptive immune responses, with the predominance of CD8+ and CD4+ cells, and in the more severe disease, with the presence of lymphoid follicles containing B lymphocytes and T cells. Furthermore, viral and bacterial infections interfere with the chronic inflammation seen in stable COPD and exacerbations via pathogen-associated molecular patterns (PAMPs). Finally, autoimmunity is another novel aspect that may play a critical role in the pathogenesis of COPD. This review is un update of the currently discussed roles of inflammatory and immune responses in the pathogenesis of COPD.
Collapse
|
155
|
Ma Z, Sun W. The effect of aerosol polyethylenimine/interferon-γ plasmid complexes on expression of inflammatory cytokines in mouse lung. J Aerosol Med Pulm Drug Deliv 2013; 27:117-24. [PMID: 23789706 DOI: 10.1089/jamp.2012.1003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The expression of inflammatory cytokines in lung tissue plays an important role in immune function of the lung. In this study, we tested whether aerosol delivery of the gene of interferon-γ (IFNγ) could affect inflammatory cytokine expression in mouse lung. METHODS Murine IFNγ-expressing plasmids (pcDNA-IFNγ) complexed with polyethylenimine (PEI) (PEI/pcDNA-IFNγ) were constructed, and their transfection efficiency was assessed in vivo using real-time quantitative RT-PCR and enzyme-linked immunosorbent assay. After aerosol administration of the plasmid complexes and confirmation of the IFNγ plasmid location in lung tissue, we measured mRNA levels of the inflammatory cytokines interleukin-1 (IL-1), IL-6, IL-10, tumor necrosis factor-α (TNF-α), and granulocyte-macrophage colony-stimulating factor (GM-CSF) on days 1 to 7 in mouse lung tissues using real-time RT-PCR. RESULTS IFNγ mRNA expression in mouse lung was significantly increased 24 hr after a single aerosol administration of PEI/pcDNA-IFNγ and gradually decreased over the next 5 days, whereas the mRNA expressions of IL-1, IL-6, and GM-CSF were markedly decreased, but not those of IL-10 and TNF-α. CONCLUSIONS PEI/IFNγ gene therapy delivered by aerosol has immune-regulating potential by suppressing lung cytokine mRNA expression, and therefore may alleviate lung disease.
Collapse
Affiliation(s)
- Zhuang Ma
- Department of Respiratory Medicine, General Hospital of Shenyang Military Area Command , Shenyang, 110016, China
| | | |
Collapse
|
156
|
Wang L, Tang Y, Liu S, Mao S, Ling Y, Liu D, He X, Wang X. Metabonomic profiling of serum and urine by (1)H NMR-based spectroscopy discriminates patients with chronic obstructive pulmonary disease and healthy individuals. PLoS One 2013; 8:e65675. [PMID: 23755267 PMCID: PMC3675021 DOI: 10.1371/journal.pone.0065675] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 04/28/2013] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) has seriously impacted the health of individuals and populations. In this study, proton nuclear magnetic resonance (1H NMR)-based metabonomics combined with multivariate pattern recognition analysis was applied to investigate the metabolic signatures of patients with COPD. Serum and urine samples were collected from COPD patients (n = 32) and healthy controls (n = 21), respectively. Samples were analyzed by high resolution 1H NMR (600 MHz), and the obtained spectral profiles were then subjected to multivariate data analysis. Consistent metabolic differences have been found in serum as well as in urine samples from COPD patients and healthy controls. Compared to healthy controls, COPD patients displayed decreased lipoprotein and amino acids, including branched-chain amino acids (BCAAs), and increased glycerolphosphocholine in serum. Moreover, metabolic differences in urine were more significant than in serum. Decreased urinary 1-methylnicotinamide, creatinine and lactate have been discovered in COPD patients in comparison with healthy controls. Conversely, acetate, ketone bodies, carnosine, m-hydroxyphenylacetate, phenylacetyglycine, pyruvate and α-ketoglutarate exhibited enhanced expression levels in COPD patients relative to healthy subjects. Our results illustrate the potential application of NMR-based metabonomics in early diagnosis and understanding the mechanisms of COPD.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yufu Tang
- Department of General Surgery, 202 Hospital of Chinese PLA, Shenyang, People’s Republic of China
| | - Shuo Liu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Shitao Mao
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yuan Ling
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Dan Liu
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Xiaoyu He
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Xiaoge Wang
- Department of Respiratory Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, People’s Republic of China
- * E-mail:
| |
Collapse
|
157
|
Wu J, Yan Z, Schwartz DE, Yu J, Malik AB, Hu G. Activation of NLRP3 inflammasome in alveolar macrophages contributes to mechanical stretch-induced lung inflammation and injury. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:3590-3599. [PMID: 23436933 PMCID: PMC3608749 DOI: 10.4049/jimmunol.1200860] [Citation(s) in RCA: 210] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Mechanical ventilation of lungs is capable of activating the innate immune system and inducing sterile inflammatory response. The proinflammatory cytokine IL-1β is among the definitive markers for accurately identifying ventilator-induced lung inflammation. However, mechanisms of IL-1β release during mechanical ventilation are unknown. In this study, we show that cyclic stretch activates the nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasomes and induces the release of IL-1β in mouse alveolar macrophages via caspase-1- and TLR4-dependent mechanisms. We also observed that NADPH oxidase subunit gp91(phox) was dispensable for stretch-induced cytokine production, whereas mitochondrial generation of reactive oxygen species was required for stretch-induced NLRP3 inflammasome activation and IL-1β release. Further, mechanical ventilation activated the NLRP3 inflammasomes in mouse alveolar macrophages and increased the production of IL-1β in vivo. IL-1β neutralization significantly reduced mechanical ventilation-induced inflammatory lung injury. These findings suggest that the alveolar macrophage NLRP3 inflammasome may sense lung alveolar stretch to induce the release of IL-1β and hence may contribute to the mechanism of lung inflammatory injury during mechanical ventilation.
Collapse
Affiliation(s)
- Jianbo Wu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Zhibo Yan
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - David E. Schwartz
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Jingui Yu
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Asrar B. Malik
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Guochang Hu
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL 60612
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612
| |
Collapse
|
158
|
Kim HJ, Jeong JS, Kim SR, Park SY, Chae HJ, Lee YC. Inhibition of endoplasmic reticulum stress alleviates lipopolysaccharide-induced lung inflammation through modulation of NF-κB/HIF-1α signaling pathway. Sci Rep 2013; 3:1142. [PMID: 23359618 PMCID: PMC3556596 DOI: 10.1038/srep01142] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 11/06/2012] [Indexed: 12/31/2022] Open
Abstract
Lipopolysaccharide (LPS) is involved in a variety of inflammatory disorders. Under stress conditions, endoplasmic reticulum (ER) loses the homeostasis in its functions, which is defined as ER stress. Little is known how ER stress is implicated in LPS-induced lung inflammation. In this study, effects of inhibition of ER stress on LPS-induced lung inflammation and transcriptional regulation were examined. An ER stress regulator, 4-phenylbutyrate (PBA) reduced LPS-induced increases of various ER stress markers in the lung. Furthermore, inhibition of ER stress reduced the LPS-induced lung inflammation. Moreover, LPS-induced increases of NF-κB and HIF-1α activity were lowered by inhibition of ER stress. These results suggest that inhibition of ER stress ameliorates LPS-induced lung inflammation through modulation of NF-κB/IκB and HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Hee Jung Kim
- Department of Internal Medicine, Chonbuk National University Medical School , Jeonju, South Korea
| | | | | | | | | | | |
Collapse
|
159
|
Gharib SA, Altemeier WA, Van Winkle LS, Plopper CG, Schlesinger SY, Buell CA, Brauer R, Lee V, Parks WC, Chen P. Matrix metalloproteinase-7 coordinates airway epithelial injury response and differentiation of ciliated cells. Am J Respir Cell Mol Biol 2012; 48:390-6. [PMID: 23258229 DOI: 10.1165/rcmb.2012-0083oc] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Matrix metalloproteinase-7 (MMP7) expression is quickly up-regulated after injury, and functions to regulate wound repair and various mucosal immune processes. We evaluated the global transcriptional response of airway epithelial cells from wild-type and Mmp7-null mice cultured at an air-liquid interface. The analysis of differentially expressed genes between genotypes after injury revealed an enrichment of functional categories associated with inflammation, cilia, and differentiation. Because these analyses suggested that MMP7 regulated ciliated cell formation, we evaluated the recovery of the airway epithelium in wild-type and Mmp7-null mice in vivo after naphthalene injury, which revealed augmented ciliated cell formation in the absence of MMP7. Moreover, in vitro studies evaluating cell differentiation in air-liquid interface cultures also showed faster ciliated cell production under Mmp7-null conditions compared with wild-type conditions. These studies identified a new role for MMP7 in attenuating ciliated cell differentiation during wound repair.
Collapse
Affiliation(s)
- Sina A Gharib
- Center for Lung Biology, University of Washington, 850 Republican Street, Seattle, WA 98109, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Reynier F, de Vos AF, Hoogerwerf JJ, Bresser P, van der Zee JS, Paye M, Pachot A, Mougin B, van der Poll T. Gene expression profiles in alveolar macrophages induced by lipopolysaccharide in humans. Mol Med 2012; 18:1303-11. [PMID: 22952057 DOI: 10.2119/molmed.2012.00230] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/28/2012] [Indexed: 01/13/2023] Open
Abstract
Lipopolysaccharide (LPS) is ubiquitous in the environment. Inhalation of LPS has been implicated in the pathogenesis and/or severity of several lung diseases, including pneumonia, chronic obstructive pulmonary disease and asthma. Alveolar macrophages are the main resident leukocytes exposed to inhaled antigens. To obtain insight into which innate immune pathways become activated within human alveolar macrophages upon exposure to LPS in vivo, we conducted a study in eight healthy humans, in which we instilled sterile saline into a lung segment by bronchoscope, followed by instillation of LPS into the contralateral lung. Six hours later, a bilateral bronchoalveolar lavage was performed and whole-genome transcriptional profiling was done on purified alveolar macrophages, comparing cells exposed to saline or LPS from the same individuals. LPS induced differential expression of 2,932 genes in alveolar macrophages; 1,520 genes were upregulated, whereas 1,440 genes were downregulated. A total of 26 biological functions were overrepresented in LPS-exposed macrophages; 44 canonical pathways affected by LPS were identified, among which the genes associated with the role of pattern recognition receptors in recognition of bacteria and viruses represented the top pathway. Other pathways included cellular immune response, signaling by tumor necrosis factor (receptor) family members, cytokine signaling and glucocorticoid receptor signaling. These results reveal for the first time a large number of functional pathways influenced by the biologically relevant challenge provided by LPS administered into the airways. These data can assist in identifying novel targets for therapeutic intervention in pulmonary diseases associated with LPS exposure, including pneumonia, asthma and chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Frederic Reynier
- Joint Unit Hospices Civils de Lyon - bioMérieux, Hôpital Edouard Herriot, Lyon, France
| | | | | | | | | | | | | | | | | |
Collapse
|
161
|
Achouiti A, Vogl T, Urban CF, Röhm M, Hommes TJ, van Zoelen MAD, Florquin S, Roth J, van 't Veer C, de Vos AF, van der Poll T. Myeloid-related protein-14 contributes to protective immunity in gram-negative pneumonia derived sepsis. PLoS Pathog 2012; 8:e1002987. [PMID: 23133376 PMCID: PMC3486918 DOI: 10.1371/journal.ppat.1002987] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 09/09/2012] [Indexed: 12/30/2022] Open
Abstract
Klebsiella (K.) pneumoniae is a common cause of pneumonia-derived sepsis. Myeloid related protein 8 (MRP8, S100A8) and MRP14 (S100A9) are the most abundant cytoplasmic proteins in neutrophils. They can form MRP8/14 heterodimers that are released upon cell stress stimuli. MRP8/14 reportedly exerts antimicrobial activity, but in acute fulminant sepsis models MRP8/14 has been found to contribute to organ damage and death. We here determined the role of MRP8/14 in K. pneumoniae sepsis originating from the lungs, using an established model characterized by gradual growth of bacteria with subsequent dissemination. Infection resulted in gradually increasing MRP8/14 levels in lungs and plasma. Mrp14 deficient (mrp14−/−) mice, unable to form MRP8/14 heterodimers, showed enhanced bacterial dissemination accompanied by increased organ damage and a reduced survival. Mrp14−/− macrophages were reduced in their capacity to phagocytose Klebsiella. In addition, recombinant MRP8/14 heterodimers, but not MRP8 or MRP14 alone, prevented growth of Klebsiella in vitro through chelation of divalent cations. Neutrophil extracellular traps (NETs) prepared from wildtype but not from mrp14−/− neutrophils inhibited Klebsiella growth; in accordance, the capacity of human NETs to kill Klebsiella was strongly impaired by an anti-MRP14 antibody or the addition of zinc. These results identify MRP8/14 as key player in protective innate immunity during Klebsiella pneumonia. Neutrophils are phagocytes that are well known for their capacity to engulf and kill microbial pathogens. It has become increasingly clear that neutrophils also kill or inhibit growth extracellularly by releasing neutrophil extracellular traps (NETs), chromatin fibers decorated with neutrophil derived proteins. MRP8/14 has been identified as one of the major antimicrobial proteins herein. Previous investigations have shown that endogenously released MRP8/14 is also sensed by the host as a danger signal and able to potentiate the harmful systemic inflammatory response syndrome. Indeed, in the setting of fulminant systemic inflammation, such as induced by endotoxin or Escherichia coli administration, MRP8/14 contributed to organ injury and mortality. The clinical scenario of sepsis however, involves an initial infection at the primary site followed by bacterial spreading to other organs. In the present setting of pneumonia-derived sepsis using the common human respiratory and sepsis pathogen Klebsiella pneumoniae MRP8/14 clearly served a beneficial role in antimicrobial defense. We here provide a likely mechanism by showing that MRP8/14 plays a role in phagocytosis and that its presence is critical in both murine and human NETs to inhibit bacterial growth.
Collapse
Affiliation(s)
- Ahmed Achouiti
- Center for Experimental and Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
van Lieshout MHP, Blok DC, Wieland CW, de Vos AF, van 't Veer C, van der Poll T. Differential roles of MyD88 and TRIF in hematopoietic and resident cells during murine gram-negative pneumonia. J Infect Dis 2012; 206:1415-23. [PMID: 22904341 DOI: 10.1093/infdis/jis505] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Pneumonia is frequently caused by gram-negative pathogens, among which Klebsiella pneumoniae prominently features. Recognition of pathogen-associated molecular patterns by Toll-like receptors (TLRs) is important for an appropriate immune response during infection. TLR signaling can proceed via 2 distinct routes that are dependent on the adaptor proteins Myeloid differentiation primary response gene (88) (MyD88) and TIR-domain-containing adaptor-inducing interferon-β (TRIF). The aim of the study was to determine the relative contribution of MyD88 and TRIF signaling in resident and hematopoietic cells to host defense during pneumonia. METHODS Bone marrow chimeras of MyD88 deficient/wild type and TRIF mutant/wild type mice were created and infected with K. pneumoniae via the airways. RESULTS MyD88 in both resident and hematopoietic cells contributed to survival and antibacterial defense in late-stage infection, whereas only TRIF in hematopoietic cells was protective. On the other hand, resident MyD88 and hematopoietic TRIF contributed to distant cellular injury. Resident MyD88 was pivotal for early chemokine release and neutrophil recruitment in the bronchoalveolar space. CONCLUSIONS MyD88- and TRIF-dependent signaling has a differential contribution to host defense in different cell types that changes from early- to late-stage gram-negative pneumonia.
Collapse
Affiliation(s)
- Miriam H P van Lieshout
- Center of Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
163
|
Matthay MA, Ware LB, Zimmerman GA. The acute respiratory distress syndrome. J Clin Invest 2012; 122:2731-40. [PMID: 22850883 DOI: 10.1172/jci60331] [Citation(s) in RCA: 1373] [Impact Index Per Article: 105.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The acute respiratory distress syndrome (ARDS) is an important cause of acute respiratory failure that is often associated with multiple organ failure. Several clinical disorders can precipitate ARDS, including pneumonia, sepsis, aspiration of gastric contents, and major trauma. Physiologically, ARDS is characterized by increased permeability pulmonary edema, severe arterial hypoxemia, and impaired carbon dioxide excretion. Based on both experimental and clinical studies, progress has been made in understanding the mechanisms responsible for the pathogenesis and the resolution of lung injury, including the contribution of environmental and genetic factors. Improved survival has been achieved with the use of lung-protective ventilation. Future progress will depend on developing novel therapeutics that can facilitate and enhance lung repair.
Collapse
Affiliation(s)
- Michael A Matthay
- Cardiovascular Research Institute and Departments of Medicine and Anesthesia, UCSF, San Francisco, CA, USA.
| | | | | |
Collapse
|
164
|
Patel AA, Lee-Lewis H, Hughes-Hanks J, Lewis CA, Anderson DM. Opposing roles for interferon regulatory factor-3 (IRF-3) and type I interferon signaling during plague. PLoS Pathog 2012; 8:e1002817. [PMID: 22911267 PMCID: PMC3406097 DOI: 10.1371/journal.ppat.1002817] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 06/10/2012] [Indexed: 11/25/2022] Open
Abstract
Type I interferons (IFN-I) broadly control innate immunity and are typically transcriptionally induced by Interferon Regulatory Factors (IRFs) following stimulation of pattern recognition receptors within the cytosol of host cells. For bacterial infection, IFN-I signaling can result in widely variant responses, in some cases contributing to the pathogenesis of disease while in others contributing to host defense. In this work, we addressed the role of type I IFN during Yersinia pestis infection in a murine model of septicemic plague. Transcription of IFN-β was induced in vitro and in vivo and contributed to pathogenesis. Mice lacking the IFN-I receptor, Ifnar, were less sensitive to disease and harbored more neutrophils in the later stage of infection which correlated with protection from lethality. In contrast, IRF-3, a transcription factor commonly involved in inducing IFN-β following bacterial infection, was not necessary for IFN production but instead contributed to host defense. In vitro, phagocytosis of Y. pestis by macrophages and neutrophils was more effective in the presence of IRF-3 and was not affected by IFN-β signaling. This activity correlated with limited bacterial growth in vivo in the presence of IRF-3. Together the data demonstrate that IRF-3 is able to activate pathways of innate immunity against bacterial infection that extend beyond regulation of IFN-β production. Type I interferons (IFN-I) broadly stimulate innate immunity against viral, bacterial and parasitic pathogens. Many bacterial pathogens induce IFN-I through phosphorylation of Interferon Regulatory Factor 3 (IRF-3) allowing it to bind promoters containing Interferon Stimulated Response Elements (ISRE) which include IFN-β and pro-inflammatory cytokines and chemokines. Secreted IFN-β is taken up by the IFN-αβ receptor (IFNAR), triggering activation of the JAK-STAT pathway which also activates ISRE-containing genes. In this work, we have discovered a novel anti-bacterial function of IRF-3. We show that the respiratory pathogen, Yersinia pestis, the causative agent of plague, activates IRF-3 and the IFN-I response and that these two events cause opposite outcomes in the host. While IRF-3 is necessary for an early stage of phagocytosis, IFNAR signaling promotes the infection and may directly contribute to neutrophil depletion during infection. These results demonstrate that an IFN-independent function of IRF-3 is important to host defense against bacterial infection.
Collapse
Affiliation(s)
- Ami A. Patel
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri, United States of America
| | - Hanni Lee-Lewis
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri, United States of America
| | - Jennifer Hughes-Hanks
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Craig A. Lewis
- Starling Enterprise, LLC, Columbia, Missouri, United States of America
| | - Deborah M. Anderson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
165
|
Eddens T, Kolls JK. Host defenses against bacterial lower respiratory tract infection. Curr Opin Immunol 2012; 24:424-30. [PMID: 22841348 DOI: 10.1016/j.coi.2012.07.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 07/06/2012] [Accepted: 07/12/2012] [Indexed: 02/08/2023]
Abstract
Bacterial pneumonia continues to be a significant cause of morbidity and mortality worldwide. Recent studies have shown that lung epithelia signal through pattern recognition receptors to initiate the innate immune response. Other mediators of innate immunity against bacterial pneumonia include transepithelial dendritic cells, alveolar macrophages, and innate produces of IL-17. CD4+ T cells and B cells play a key role in eliminating and preventing the development of bacterial pneumonias. B cell development and maturation can be modulated by the lung epithelia through BAFF and APRIL, furthering our current understanding of the role of epithelial cells in the immune response.
Collapse
Affiliation(s)
- Taylor Eddens
- Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | | |
Collapse
|
166
|
Erdely A, Antonini JM, Salmen-Muniz R, Liston A, Hulderman T, Simeonova PP, Kashon ML, Li S, Gu JK, Stone S, Chen BT, Frazer DG, Zeidler-Erdely PC. Type I interferon and pattern recognition receptor signaling following particulate matter inhalation. Part Fibre Toxicol 2012; 9:25. [PMID: 22776377 PMCID: PMC3537608 DOI: 10.1186/1743-8977-9-25] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 06/21/2012] [Indexed: 12/02/2022] Open
Abstract
Background Welding, a process that generates an aerosol containing gases and metal-rich particulates, induces adverse physiological effects including inflammation, immunosuppression and cardiovascular dysfunction. This study utilized microarray technology and subsequent pathway analysis as an exploratory search for markers/mechanisms of in vivo systemic effects following inhalation. Mice were exposed by inhalation to gas metal arc – stainless steel (GMA-SS) welding fume at 40 mg/m3 for 3 hr/d for 10 d and sacrificed 4 hr, 14 d and 28 d post-exposure. Whole blood cells, aorta and lung were harvested for global gene expression analysis with subsequent Ingenuity Pathway Analysis and confirmatory qRT-PCR. Serum was collected for protein profiling. Results The novel finding was a dominant type I interferon signaling network with the transcription factor Irf7 as a central component maintained through 28 d. Remarkably, these effects showed consistency across all tissues indicating a systemic type I interferon response that was complemented by changes in serum proteins (decreased MMP-9, CRP and increased VCAM1, oncostatin M, IP-10). In addition, pulmonary expression of interferon α and β and Irf7 specific pattern recognition receptors (PRR) and signaling molecules (Ddx58, Ifih1, Dhx58, ISGF3) were induced, an effect that showed specificity when compared to other inflammatory exposures. Also, a canonical pathway indicated a coordinated response of multiple PRR and associated signaling molecules (Tlr7, Tlr2, Clec7a, Nlrp3, Myd88) to inhalation of GMA-SS. Conclusion This methodological approach has the potential to identify consistent, prominent and/or novel pathways and provides insight into mechanisms that contribute to pulmonary and systemic effects following toxicant exposure.
Collapse
Affiliation(s)
- Aaron Erdely
- Pathology and Physiology Research Branch, Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Hoppstädter J, Diesel B, Eifler LK, Schmid T, Brüne B, Kiemer AK. Glucocorticoid-induced leucine zipper is downregulated in human alveolar macrophages upon Toll-like receptor activation. Eur J Immunol 2012; 42:1282-93. [PMID: 22539300 DOI: 10.1002/eji.201142081] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Induction of the glucocorticoid-induced leucine zipper (GILZ) by glucocorticoids plays a role in their antiinflammatory action, whereas GILZ expression is reduced under inflammatory conditions. The mechanisms regulating GILZ expression during inflammation, however, have not yet been characterized. Here, we investigated GILZ expression in human alveolar macrophages (AMs) following Toll-like receptor (TLR) activation. Macrophages were shown to predominantly express GILZ transcript variant 2. Lipopolysaccharide-treated AMs, THP-1 cells, and lungs of lipopolysaccharide-exposed mice displayed decreased GILZ protein and mRNA levels. The effect was strictly dependent on the adapter molecule MyD88, as shown by using specific ligands or a knockdown strategy. Investigations on the functional significance of GILZ downregulation performed by GILZ knockdown revealed a proinflammatory response, as indicated by increased cytokine expression and NF-κB activity. We found that TLR activation reduced GILZ mRNA stability, which was mediated via the GILZ 3'-untranslated region. Finally, involvement of the mRNA-binding protein tristetraprolin (TTP) is suggested, since TTP overexpression or knockdown modulated GILZ expression and TTP was induced in a MyD88-dependent fashion. Taken together, our data show a MyD88- and TTP-dependent GILZ downreg-ulation in human macrophages upon TLR activation. Suppression of GILZ is mediated by mRNA destabilization, which might represent a regulatory mechanism in macrophage activation.
Collapse
Affiliation(s)
- Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Saarbrücken, Germany
| | | | | | | | | | | |
Collapse
|
168
|
Barna BP, Culver DA, Kanchwala A, Singh RJ, Huizar I, Abraham S, Malur A, Marshall I, Kavuru MS, Thomassen MJ. Alveolar macrophage cathelicidin deficiency in severe sarcoidosis. J Innate Immun 2012; 4:569-78. [PMID: 22759465 DOI: 10.1159/000339149] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 04/27/2012] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Dysfunctional immune responses characterize sarcoidosis, but the status of cathelicidin, a potent immunoregulatory and antimicrobial molecule, has not been established in clinical disease activity. METHODS Alveolar macrophage cathelicidin expression was determined in biopsy-proven sarcoidosis patients classified clinically as 'severe' (requiring systemic treatment) or 'non-severe' (never requiring treatment). Bronchoalveolar lavage (BAL) cells from sarcoidosis patients and healthy controls were analyzed for mRNA expression of cathelicidin, vitamin D receptor (VDR) and the VDR coactivator steroid receptor coactivator-3 (SRC3) by quantitative PCR. Cathelicidin-derived peptide LL-37 was determined by immunocytochemistry. Serum calcidiol (25-hydroxyvitamin D2; vitD2) and calcitriol (1,25-dihydroxyvitamin D3; vitD3) were quantified. RESULTS The results indicated reduced BAL cell expression of cathelicidin and SRC3 in severe but not non-severe sarcoidosis compared to controls. Serum levels of biologically active vitD3 in both severe and non-severe patients were within the control range even though vitD2 levels in both groups were below the recommended level (30 ng/ml). Sarcoidosis and control alveolar macrophages were studied in vitro to determine cathelicidin responses to vitD3 and tumor necrosis factor-α (TNFα), a vitD3 antagonist elevated in active sarcoidosis. Alveolar macrophage cathelicidin was stimulated by vitD3 but repressed by TNFα, which also repressed SRC3. CONCLUSIONS These findings suggest that TNFα-mediated repression of SRC3 contributes to alveolar macrophage cathelicidin deficiency in severe sarcoidosis despite healthy vitD3 levels. Deficiency of cathelicidin, a multifunctional regulator of immune cells and proinflammatory cytokines, may impede resolution of inflammation in the lungs of patients with severe sarcoidosis.
Collapse
Affiliation(s)
- Barbara P Barna
- Division of Pulmonary and Critical Care Medicine, East Carolina University, Greenville, NC 27834, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Hoogendijk AJ, Pinhanços SS, van der Poll T, Wieland CW. Intrapulmonary administration of a p38 mitogen activated protein kinase inhibitor partially prevents pulmonary inflammation. Immunobiology 2012; 218:435-42. [PMID: 22727776 DOI: 10.1016/j.imbio.2012.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 05/30/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Gram-positive and gram-negative bacteria are common causative agents of respiratory tract infection. Lipopolysaccharide (LPS) is a component of the gram-negative cell wall and a strong inducer of inflammation. The main proinflammatory component of the gram-positive bacterial cell wall is lipoteichoic acid (LTA). The protein kinase p38 mitogen activated protein kinase (MAPK) plays an important role in the inflammatory process induced by these two bacterial structures. AIM We here sought to establish the impact of local p38 MAPK inhibition on lung inflammatory responses induced by LPS and LTA. We investigated the effects of direct intrapulmonary delivery of a p38 MAPK inhibitor on local LPS and LTA induced airway inflammation in mice. RESULTS In vitro, BIRB 796 reduced LPS induced p38 MAPK phosphorylation in alveolar macrophage and respiratory epithelial cell lines and diminished cytokine/chemokine release. In vivo, BIRB 796 circumvented p38 MAPK phosphorylation in both LPS and LTA induced inflammation. Cellular influx was not affected. Lung TNFα, IL-6, MIP-2 and LIX production was reduced in LPS induced inflammation but not in lung inflammation by LTA. BIRB 796 reduced total protein and IgM in bronchoalveolar lavage fluid after LTA instillation, while enhancing TATc and d-dimers in LPS- and LTA induced inflammation. CONCLUSION These results taken together with earlier studies on systemic administration of p38 MAPK inhibitors in rodents and humans suggest that direct intrapulmonary delivery of a p38 MAPK inhibitor is less effective in inhibiting inflammation and is associated with unexpected procoagulant effects in the bronchoalveolar space.
Collapse
Affiliation(s)
- Arie Johan Hoogendijk
- Center for Infection and Immunity Amsterdam, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
170
|
Li HH, Li Q, Liu P, Liu Y, Li J, Wasserloos K, Chao W, You M, Oury TD, Chhinder S, Hackam DJ, Billiar TR, Leikauf GD, Pitt BR, Zhang LM. WNT1-inducible signaling pathway protein 1 contributes to ventilator-induced lung injury. Am J Respir Cell Mol Biol 2012; 47:528-35. [PMID: 22700866 DOI: 10.1165/rcmb.2012-0127oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Although strides have been made to reduce ventilator-induced lung injury (VILI), critically ill patients can vary in sensitivity to VILI, suggesting gene-environment interactions could contribute to individual susceptibility. This study sought to uncover candidate genes associated with VILI using a genome-wide approach followed by functional analysis of the leading candidate in mice. Alveolar-capillary permeability after high tidal volume (HTV) ventilation was measured in 23 mouse strains, and haplotype association mapping was performed. A locus was identified on chromosome 15 that contained ArfGAP with SH3 domain, ankyrin repeat and PH domain 1 (Asap1), adenylate cyclase 8 (Adcy8), WNT1-inducible signaling pathway protein 1 (Wisp1), and N-myc downstream regulated 1 (Ndrg1). Information from published studies guided initial assessment to Wisp1. After HTV, lung WISP1 protein increased in sensitive A/J mice, but was unchanged in resistant CBA/J mice. Anti-WISP1 antibody decreased HTV-induced alveolar-capillary permeability in sensitive A/J mice, and recombinant WISP1 protein increased HTV-induced alveolar-capillary permeability in resistant CBA/J mice. HTV-induced WISP1 coimmunoprecipitated with glycosylated Toll-like receptor (TLR) 4 in A/J lung homogenates. After HTV, WISP1 increased in strain-matched control lungs, but was unchanged in TLR4 gene-targeted lungs. In peritoneal macrophages from strain-matched mice, WISP1 augmented LPS-induced TNF release that was inhibited in macrophages from TLR4 or CD14 antigen gene-targeted mice, and was attenuated in macrophages from myeloid differentiation primary response gene 88 gene-targeted or TLR adaptor molecule 1 mutant mice. These findings support a role for WISP1 as an endogenous signal that acts through TLR4 signaling to increase alveolar-capillary permeability in VILI.
Collapse
Affiliation(s)
- Hui-Hua Li
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Tanaka S, Saito Y, Kunisawa J, Kurashima Y, Wake T, Suzuki N, Shultz LD, Kiyono H, Ishikawa F. Development of mature and functional human myeloid subsets in hematopoietic stem cell-engrafted NOD/SCID/IL2rγKO mice. THE JOURNAL OF IMMUNOLOGY 2012; 188:6145-55. [PMID: 22611244 DOI: 10.4049/jimmunol.1103660] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Although physiological development of human lymphoid subsets has become well documented in humanized mice, in vivo development of human myeloid subsets in a xenotransplantation setting has remained unevaluated. Therefore, we investigated in vivo differentiation and function of human myeloid subsets in NOD/SCID/IL2rγ(null) (NSG) mouse recipients transplanted with purified lineage(-)CD34(+)CD38(-) cord blood hematopoietic stem cells. At 4-6 mo posttransplantation, we identified the development of human neutrophils, basophils, mast cells, monocytes, and conventional and plasmacytoid dendritic cells in the recipient hematopoietic organs. The tissue distribution and morphology of these human myeloid cells were similar to those identified in humans. After cytokine stimulation in vitro, phosphorylation of STAT molecules was observed in neutrophils and monocytes. In vivo administration of human G-CSF resulted in the recruitment of human myeloid cells into the recipient circulation. Flow cytometry and confocal imaging demonstrated that human bone marrow monocytes and alveolar macrophages in the recipients displayed intact phagocytic function. Human bone marrow-derived monocytes/macrophages were further confirmed to exhibit phagocytosis and killing of Salmonella typhimurium upon IFN-γ stimulation. These findings demonstrate the development of mature and functionally intact human myeloid subsets in vivo in the NSG recipients. In vivo human myelopoiesis established in the NSG humanized mouse system may facilitate the investigation of human myeloid cell biology including in vivo analyses of infectious diseases and therapeutic interventions.
Collapse
Affiliation(s)
- Satoshi Tanaka
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
172
|
Fuschillo S, Martucci M, Donner CF, Balzano G. Airway bacterial colonization: the missing link between COPD and cardiovascular events? Respir Med 2012; 106:915-23. [PMID: 22546638 DOI: 10.1016/j.rmed.2012.03.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 03/28/2012] [Accepted: 03/29/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death worldwide and, according to the World Health Organization, its prevalence will double by 2020. COPD is a chronic inflammatory disease of the lung characterized by poorly reversible airflow limitation and, frequently, by extrapulmonary manifestations. In particular, the cardiovascular manifestations are responsible for high morbidity and mortality. METHODS AND RESULTS A systematic literature search was performed of studies published in Medline until December 2010, using the key-words: COPD, bacterial colonization, COPD exacerbation, atherosclerosis, systemic inflammation, cardiovascular event and risk factors. In addition to the studies identified in the primary search, reference lists of included articles were analyzed for additional papers related to the topic. The pathogenetic mechanisms underlying atherosclerosis - namely inflammation, oxidative stress and endothelial dysfunction - are in common with COPD. Moreover, they are increased in the presence of COPD, especially in patients who present airway bacterial colonization, increased rate of exacerbations and elevated levels of both airway and systemic inflammation. CONCLUSION COPD is associated with an increased burden of atherosclerotic disease. Systemic inflammation and oxidative stress play key roles in this association. COPD patients with airway bacterial colonization, as compared to patients without airway colonization, generally present more frequent exacerbations and higher levels of both airway and systemic inflammation. This COPD subgroup should be considered at particularly increased risk of developing cardiovascular complications and receive more attention concerning diagnosis, treatment, prevention and research.
Collapse
Affiliation(s)
- Salvatore Fuschillo
- Pulmonary Rehabilitation Unit, Salvatore Maugeri Foundation, Scientific Institute of Telese, Bagni Vecchi 1, 82037 Telese Terme, BN, Italy.
| | | | | | | |
Collapse
|
173
|
Ci X, Chu X, Wei M, Yang X, Cai Q, Deng X. Different effects of farrerol on an OVA-induced allergic asthma and LPS-induced acute lung injury. PLoS One 2012; 7:e34634. [PMID: 22563373 PMCID: PMC3338508 DOI: 10.1371/journal.pone.0034634] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 03/02/2012] [Indexed: 01/05/2023] Open
Abstract
Background Farrerol, isolated from rhododendron, has been shown to have the anti-bacterial activity, but no details on the anti-inflammatory activity. We further evaluated the effects of this compound in two experimental models of lung diseases. Methodology/Principal Findings For the asthma model, female BALB/c mice were challenged with ovalbumin (OVA), and then treated daily with farrerol (20 and 40 mg/kg, ip) as a therapeutic treatment from day 22 to day 26 post immunization. To induce acute lung injury, female BALB/c mice were injected intranasally with LPS and treated with farrerol (20 and 40 mg/kg, i.p.) 1 h prior to LPS stimulation. Inflammation in the two different models was determined using ELISA, histology, real-time PCR and western blot. Farrerol significantly regulated the phenotype challenged by OVA, like cell number, Th1 and Th2 cytokines levels in the BALF, the OVA-specific IgE level in the serum, goblet cell hyperplasia in the airway, airway hyperresponsiveness to inhaled methacholine and mRNA expression of chemokines and their receptors. Furthermore, farrerol markedly attenuated the activation of phosphorylation of Akt and nuclear factor-κB (NF-κB) subunit p65 both in vivo and in vitro. However, farrerol has no effect on the acute lung injury model. Conclusion/Significance Our finding demonstrates that the distinct anti-inflammatory effect of farrerol in the treatment of asthma acts by inhibiting the PI3K and NF-κB pathway.
Collapse
Affiliation(s)
- Xinxin Ci
- Institute of Zoonoses, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Xiao Chu
- Institute of Zoonoses, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Miaomiao Wei
- Institute of Zoonoses, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Xiaofeng Yang
- Institute of Zoonoses, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Qinren Cai
- Institute of Zoonoses, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Xuming Deng
- Institute of Zoonoses, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, People’s Republic of China
- * E-mail:
| |
Collapse
|
174
|
Abstract
Chronic obstructive pulmonary disease (COPD) is characterised by progressive airflow obstruction that is only partly reversible, inflammation in the airways, and systemic effects or comorbities. The main cause is smoking tobacco, but other factors have been identified. Several pathobiological processes interact on a complex background of genetic determinants, lung growth, and environmental stimuli. The disease is further aggravated by exacerbations, particularly in patients with severe disease, up to 78% of which are due to bacterial infections, viral infections, or both. Comorbidities include ischaemic heart disease, diabetes, and lung cancer. Bronchodilators constitute the mainstay of treatment: β(2) agonists and long-acting anticholinergic agents are frequently used (the former often with inhaled corticosteroids). Besides improving symptoms, these treatments are also thought to lead to some degree of disease modification. Future research should be directed towards the development of agents that notably affect the course of disease.
Collapse
Affiliation(s)
- Marc Decramer
- Respiratory Division, University Hospital, University of Leuven, Leuven, Belgium.
| | | | | |
Collapse
|
175
|
Napolitano JR, Liu MJ, Bao S, Crawford M, Nana-Sinkam P, Cormet-Boyaka E, Knoell DL. Cadmium-mediated toxicity of lung epithelia is enhanced through NF-κB-mediated transcriptional activation of the human zinc transporter ZIP8. Am J Physiol Lung Cell Mol Physiol 2012; 302:L909-18. [PMID: 22345571 DOI: 10.1152/ajplung.00351.2011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cadmium (Cd), a toxic heavy metal and carcinogen that is abundantly present in cigarette smoke, is a cause of smoking-induced lung disease. SLC39A8 (ZIP8), a zinc transporter, is a major portal for Cd uptake into cells. We have recently identified that ZIP8 expression is under the transcriptional control of the NF-κB pathway. On the basis of this, we hypothesized that cigarette-smoke induced inflammation would increase ZIP8 expression in lung epithelia, thereby enhancing Cd uptake and cell toxicity. Herein we report that ZIP8 is a central mediator of Cd-mediated toxicity. TNF-α treatment of primary human lung epithelia and A549 cells induced ZIP8 expression, resulting in significantly higher cell death attributable to both apoptosis and necrosis following Cd exposure. Inhibition of the NF-κB pathway and ZIP8 expression significantly reduced cell toxicity. Zinc (Zn), a known cytoprotectant, prevented Cd-mediated cell toxicity via ZIP8 uptake. Consistent with cell culture findings, a significant increase in ZIP8 mRNA and protein expression was observed in the lung of chronic smokers compared with nonsmokers. From these studies, we conclude that ZIP8 expression is induced in lung epithelia in an NF-κB-dependent manner, thereby resulting in increased cell death in the presence of Cd. From this we contend that ZIP8 plays a critical role at the interface between micronutrient (Zn) metabolism and toxic metal exposure (Cd) in the lung microenvironment following cigarette smoke exposure. Furthermore, dietary Zn intake, or a lack thereof, may be a contributing factor in smoking-induced lung disease.
Collapse
Affiliation(s)
- Jessica R Napolitano
- Integrated Biomedical Science Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
176
|
Xu W, Chen M, Ge N, Xu J. Hemagglutinin from the H5N1 virus activates Janus kinase 3 to dysregulate innate immunity. PLoS One 2012; 7:e31721. [PMID: 22359619 PMCID: PMC3280993 DOI: 10.1371/journal.pone.0031721] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 01/11/2012] [Indexed: 11/19/2022] Open
Abstract
Highly pathogenic avian influenza viruses (HPAIVs) cause severe disease in humans. There are no effective vaccines or antiviral therapies currently available to control fatal outbreaks due in part to the lack of understanding of virus-mediated immunopathology. In our study, we used hemagglutinin (HA) of H5N1 virus to investigate the related signaling pathways and their relationship to dysregulated innate immune reaction. We found the HA of H5N1 avian influenza triggered an abnormal innate immune signalling in the pulmonary epithelial cells, through an unusual process involving activation of Janus kinase 3 (JAK3) that is exclusively associated with γc chain and is essential for signaling via all γc cytokine receptors. By using a selective JAK3 inhibitor and JAK3 knockout mice, we have, for the first time, demonstrated the ability to target active JAK3 to counteract injury to the lungs and protect immunocytes from acute hypercytokinemia -induced destruction following the challenge of H5N1 HA in vitro and in vivo. On the basis of the present data, it appears that the efficacy of selective JAK3 inhibition is likely based on its ability to block multiple cytokines and protect against a superinflammatory response to pathogen-associated molecular patterns (PAMPs) attack. Our findings highlight the potential value of selective JAK3 inhibitor in treating the fatal immunopathology caused by H5N1 challenge.
Collapse
Affiliation(s)
- Wei Xu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Minhui Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Nanhai Ge
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jun Xu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China
- * E-mail:
| |
Collapse
|
177
|
Koppe U, Suttorp N, Opitz B. Recognition of Streptococcus pneumoniae by the innate immune system. Cell Microbiol 2012; 14:460-6. [DOI: 10.1111/j.1462-5822.2011.01746.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
178
|
Xie T, Liang J, Liu N, Wang Q, Li Y, Noble PW, Jiang D. MicroRNA-127 inhibits lung inflammation by targeting IgG Fcγ receptor I. THE JOURNAL OF IMMUNOLOGY 2012; 188:2437-44. [PMID: 22287715 DOI: 10.4049/jimmunol.1101070] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The molecular mechanisms of acute lung injury are incompletely understood. MicroRNAs (miRNAs) are crucial biological regulators that act by suppressing their target genes and are involved in a variety of pathophysiologic processes. miR-127 appears to be downregulated during lung injury. We set out to investigate the role of miR-127 in lung injury and inflammation. Expression of miR-127 significantly reduced cytokine release by macrophages. Looking into the mechanisms of regulation of inflammation by miR-127, we found that IgG FcγRI (CD64) was a target of miR-127, as evidenced by reduced CD64 protein expression in macrophages overexpressing miR-127. Furthermore, miR-127 significantly reduced the luciferase activity with a reporter construct containing the native 3' untranslated region of CD64. Importantly, we demonstrated that miR-127 attenuated lung inflammation in an IgG immune complex model in vivo. Collectively, these data show that miR-127 targets macrophage CD64 expression and promotes the reduction of lung inflammation. Understanding how miRNAs regulate lung inflammation may represent an attractive way to control inflammation induced by infectious or noninfectious lung injury.
Collapse
Affiliation(s)
- Ting Xie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
179
|
Eitel J, Meixenberger K, van Laak C, Orlovski C, Hocke A, Schmeck B, Hippenstiel S, N'Guessan PD, Suttorp N, Opitz B. Rac1 regulates the NLRP3 inflammasome which mediates IL-1beta production in Chlamydophila pneumoniae infected human mononuclear cells. PLoS One 2012; 7:e30379. [PMID: 22276187 PMCID: PMC3262829 DOI: 10.1371/journal.pone.0030379] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 12/19/2011] [Indexed: 01/19/2023] Open
Abstract
Chlamydophila pneumoniae causes acute respiratory tract infections and has been associated with development of asthma and atherosclerosis. The production of IL-1β, a key mediator of acute and chronic inflammation, is regulated on a transcriptional level and additionally on a posttranslational level by inflammasomes. In the present study we show that C. pneumoniae-infected human mononuclear cells produce IL-1β protein depending on an inflammasome consisting of NLRP3, the adapter protein ASC and caspase-1. We further found that the small GTPase Rac1 is activated in C. pneumoniae-infected cells. Importantly, studies with specific inhibitors as well as siRNA show that Rac1 regulates inflammasome activation in C. pneumoniae-infected cells. In conclusion, C. pneumoniae infection of mononuclear cells stimulates IL-1β production dependent on a NLRP3 inflammasome-mediated processing of proIL-1β which is controlled by Rac1.
Collapse
Affiliation(s)
- Julia Eitel
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Karolin Meixenberger
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia van Laak
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Christine Orlovski
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Hocke
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Bernd Schmeck
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Philippe Dje N'Guessan
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Norbert Suttorp
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Bastian Opitz
- Department of Internal Medicine, Infectious Diseases and Pulmonary Medicine, Charité – Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
180
|
von Bubnoff D, Sell U, Arriens S, Specht S, Hoerauf A, Bieber T. Differential Expression of Toll-Like Receptor 2 on Dendritic Cells from Asymptomatic and Symptomatic Atopic Donors. Int Arch Allergy Immunol 2012; 159:41-50. [DOI: 10.1159/000335234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 11/14/2011] [Indexed: 11/19/2022] Open
|
181
|
Abstract
Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS), have high mortality rates with few treatment options. An important regulatory factor in the pathology observed in ALI/ARDS is a disruption of the pulmonary endothelial barrier which, in combination with epithelial barrier disruption, causes leakage of fluid, protein and cells into lung airspaces. Degradation of the glycosaminoglycan, hyaluronan (HA), is involved in reduction of the endothelial glycocalyx, disruption of endothelial cell-cell contacts and activation of HA binding proteins upregulated in ALI/ARDS which promote a loss of pulmonary vascular integrity. In contrast, exogenous administration of high molecular weight HA has been shown to be protective in several models of ALI. This review focuses on the dichotomous role of HA to both promote and inhibit ALI based on its size and the HA binding proteins present. Further, potential therapeutic applications of high molecular weight HA in treating ALI/ARDS are discussed.
Collapse
Affiliation(s)
- Patrick A Singleton
- Department of Medicine, Section of Pulmonary and Critical Care, The University of Chicago, Chicago, IL 60637, USA ; Department of Anesthesia and Critical Care, Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
182
|
Lunov O, Syrovets T, Loos C, Nienhaus GU, Mailänder V, Landfester K, Rouis M, Simmet T. Amino-functionalized polystyrene nanoparticles activate the NLRP3 inflammasome in human macrophages. ACS NANO 2011; 5:9648-57. [PMID: 22111911 DOI: 10.1021/nn203596e] [Citation(s) in RCA: 198] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Specifically designed and functionalized nanoparticles hold great promise for biomedical applications. Yet, the applicability of nanoparticles is critically predetermined by their surface functionalization. Here we demonstrate that amino-functionalized polystyrene nanoparticles (PS-NH(2)) of ∼100 nm in diameter, but not carboxyl- or nonfunctionalized particles, trigger NLRP3 inflammasome activation and subsequent release of proinflammatory interleukin 1β (IL-1β) by human macrophages. PS-NH(2) induced time-dependent proton accumulation in lysosomes associated with lysosomal destabilization, release of cathepsin B, and damage of the mitochondrial membrane. Accumulation of mitochondrial reactive oxygen species was accompanied by oxidation of thioredoxin, a protein playing a central role in maintaining the cellular redox balance. Upon oxidation, thioredoxin dissociated from the thioredoxin-interacting protein (TXNIP). Liberated TXNIP, in turn, interacted with the NLRP3 protein, resulting in a conformational change of the pyrin domain of the NLRP3 protein, as was predicted by molecular modeling. Consequently, this prompted assembly of the NLRP3 inflammasome complex with recruitment and activation of caspase-1, inducing IL-1β release by cleavage of pro-IL-1β. The central role of the NLRP3 inflammasome for cytokine production was confirmed by in vitro knockdown of NLRP3 and of the adaptor protein ASC, confirming that other inflammasomes were not activated by PS-NH(2). The PS-NH(2)-mediated proinflammatory macrophage activation could be antagonized by the radical scavenger N-acetyl-L-cysteine, which prevented mitochondrial damage, caspase-1 activation, and the subsequent release of IL-1β. Our study reveals the molecular mechanism of NLRP3 inflammasome activation by amino-functionalized nanoparticles and suggests a strategy as to how such adverse effects could be antagonized.
Collapse
Affiliation(s)
- Oleg Lunov
- Institute of Pharmacology of Natural Products & Clinical Pharmacology, Ulm University, Ulm, Germany
| | | | | | | | | | | | | | | |
Collapse
|
183
|
Menéndez R, Sahuquillo-Arce JM, Reyes S, Martínez R, Polverino E, Cillóniz C, Córdoba JG, Montull B, Torres A. Cytokine activation patterns and biomarkers are influenced by microorganisms in community-acquired pneumonia. Chest 2011; 141:1537-1545. [PMID: 22194589 PMCID: PMC7094498 DOI: 10.1378/chest.11-1446] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background The inflammatory response in community-acquired pneumonia (CAP) depends on the host and on the challenge of the causal microorganism. Here, we analyze the patterns of inflammatory cytokines, procalcitonin (PCT), and C-reactive protein (CRP) in order to determine their diagnostic value. Methods This was a prospective study of 658 patients admitted with CAP. PCT and CRP were analyzed by immunoluminometric and immunoturbidimetric assays. Cytokines (tumor necrosis factor-α [TNF-α], IL-1β, IL-6, IL-8, and IL-10) were measured using enzyme immunoassay. Results The lowest medians of CRP, PCT, TNF-α, and IL-6 were found in CAP of unknown cause, and the highest were found in patients with positive blood cultures. Different cytokine profiles and biomarkers were found depending on cause: atypical bacteria (lower PCT and IL-6), viruses (lower PCT and higher IL-10), Enterobacteriaceae (higher IL-8), Streptococcus pneumoniae (high PCT), and Legionella pneumophila (higher CRP and TNF-α). PCT ≥ 0.36 mg/dL to predict positive blood cultures showed sensitivity of 85%, specificity of 42%, and negative predictive value (NPV) of 98%, whereas a cutoff of ≤ 0.5 mg/dL to predict viruses or atypicals vs bacteria showed sensitivity of 89%/81%, specificity of 68%/68%, positive predictive value of 12%/22%, and NPV of 99%/97%. In a multivariate Euclidean distance model, the lowest inflammatory expression was found in unknown cause and the highest was found in L pneumophila, S pneumoniae, and Enterobacteriaceae. Atypical bacteria exhibit an inflammatory pattern closer to that of viruses. Conclusions Different inflammatory patterns elicited by different microorganisms may provide a useful tool for diagnosis. Recognizing these patterns provides additional information that may facilitate a broader understanding of host inflammatory response to microorganisms.
Collapse
Affiliation(s)
- Rosario Menéndez
- Servicio de Neumología, Hospital Universitari i Politècnic La Fe, Valencia, Spain.
| | | | - Soledad Reyes
- Servicio de Neumología, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Raquel Martínez
- Servicio de Neumología, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Eva Polverino
- Servei de Pneumologia, Institut del Tòrax, Hospital Clinic, IDIBAPS, CIBERES, Universitat de Barcelona, Barcelona, Spain
| | - Catia Cillóniz
- Servei de Pneumologia, Institut del Tòrax, Hospital Clinic, IDIBAPS, CIBERES, Universitat de Barcelona, Barcelona, Spain
| | - Juan Ginés Córdoba
- Servicio de Microbiología, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Beatriz Montull
- Servicio de Neumología, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Antoni Torres
- Servei de Pneumologia, Institut del Tòrax, Hospital Clinic, IDIBAPS, CIBERES, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
184
|
Koppe U, Högner K, Doehn JM, Müller HC, Witzenrath M, Gutbier B, Bauer S, Pribyl T, Hammerschmidt S, Lohmeyer J, Suttorp N, Herold S, Opitz B. Streptococcus pneumoniae stimulates a STING- and IFN regulatory factor 3-dependent type I IFN production in macrophages, which regulates RANTES production in macrophages, cocultured alveolar epithelial cells, and mouse lungs. THE JOURNAL OF IMMUNOLOGY 2011; 188:811-7. [PMID: 22156592 DOI: 10.4049/jimmunol.1004143] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Streptococcus pneumoniae is the leading cause of community-acquired pneumonia. In this study, we examine an innate immune recognition pathway that senses pneumococcal infection, triggers type I IFN production, and regulates RANTES production. We found that human and murine alveolar macrophages as well as murine bone marrow macrophages, but not alveolar epithelial cells, produced type I IFNs upon infection with S. pneumoniae. This response was dependent on the pore-forming toxin pneumolysin and appeared to be mediated by a cytosolic DNA-sensing pathway involving the adapter molecule STING and the transcription factor IFN regulatory factor 3. Indeed, DNA was present in the cytosol during pneumococcal infection as indicated by the activation of the AIM2 inflammasome, which is known to sense microbial DNA. Type I IFNs produced by S. pneumoniae-infected macrophages positively regulated gene expression and RANTES production in macrophages and cocultured alveolar epithelial cells in vitro. Moreover, type I IFNs controlled RANTES production during pneumococcal pneumonia in vivo. In conclusion, we identified an immune sensing pathway detecting S. pneumoniae that triggers a type I IFN response and positively regulates RANTES production.
Collapse
Affiliation(s)
- Uwe Koppe
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité University Medicine Berlin, 13353 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Heidemann SM, Sandhu H, Kovacevic N, Phumeetham S, Solomon R. Detection of tumor necrosis factor-α and interleukin-6 in exhaled breath condensate of rats with pneumonia due to staphylococcal enterotoxin B. Exp Lung Res 2011; 37:563-7. [PMID: 21967195 DOI: 10.3109/01902148.2011.611963] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Exhaled breath condensate (EBC) may contain mediators of acute lung injury. The objectives were to determine if EBC could be collected in a mechanically ventilated rat, to measure tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in the EBC after staphylococcal enterotoxin B administration (SEB) and to find out if the concentrations of TNF-α and IL-6 correlated with those in lung lavage. Four hours after SEB instillation, rats were placed on mechanical ventilation and EBC was collected over 90 minutes. Lung lavage was collected and white cell count was determined. TNF-α and IL-6 were measured in the EBC and lavage. EBC was available in a sufficient quantity (250-400 μL) for the measurement of cytokines. The rats that received SEB had an inflammatory response when compared to control rats as shown by an increase in white cell count. TNF-α and IL-6 were detected in the EBC. Concentration of TNF-α correlated with that in the lavage (r = .497, P = .021), whereas IL-6 did not. EBC can be collected in rats in sufficient quantities to study acute lung injury. TNF-α and IL-6 can be measured in the EBC. Correlation between TNF-α in the EBC and lavage was demonstrated in this rat model of lung injury.
Collapse
|
186
|
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous syndrome associated with abnormal inflammatory immune responses of the lung to noxious particles and gases. Cigarette smoke activates innate immune cells such as epithelial cells and macrophages by triggering pattern recognition receptors, either directly or indirectly via the release of damage-associated molecular patterns from stressed or dying cells. Activated dendritic cells induce adaptive immune responses encompassing T helper (Th1 and Th17) CD4+ T cells, CD8+ cytotoxicity, and B-cell responses, which lead to the development of lymphoid follicles on chronic inflammation. Viral and bacterial infections not only cause acute exacerbations of COPD, but also amplify and perpetuate chronic inflammation in stable COPD via pathogen-associated molecular patterns. We discuss the role of autoimmunity (autoantibodies), remodelling, extracellular matrix-derived fragments, impaired innate lung defences, oxidative stress, hypoxia, and dysregulation of microRNAs in the persistence of the pulmonary inflammation despite smoking cessation.
Collapse
Affiliation(s)
- Guy G Brusselle
- Laboratory for Translational Research of Obstructive Pulmonary Disease, Department of Respiratory Medicine, Ghent University Hospital and Ghent University, Ghent, Belgium.
| | | | | |
Collapse
|
187
|
Thaikoottathil J, Chu HW. MAPK/AP-1 activation mediates TLR2 agonist-induced SPLUNC1 expression in human lung epithelial cells. Mol Immunol 2011; 49:415-22. [PMID: 21899893 DOI: 10.1016/j.molimm.2011.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 08/02/2011] [Accepted: 08/06/2011] [Indexed: 12/12/2022]
Abstract
BACKGROUND Short Palate Lung and Nasal epithelium Clone 1 (SPLUNC1) is a newly described host defense protein, primarily expressed in large airway epithelial cells. Reduced SPLUNC1 has been reported in allergic and cigarette smoke-exposed airways. We found that Mycoplasma pneumoniae increases SPLUNC1 in airway epithelium in part via activating TLR2-NF-κB pathway. However, the contribution of additional signaling pathways to TLR2-mediated SPLUNC1 expression remains unclear. In the present study, we investigated if TLR2-induced mitogen-activated protein kinase (MAPK)/activator protein-1 (AP-1) signaling regulates SPLUNC1 expression in human lung epithelial cells. METHODS Human lung epithelial NCI-H292 cells were stimulated with a TLR2 agonist Palmitoyl (3)-Cys-Ser-Lys (4)-OH (Pam(3)CSK(4)). MAPK/AP-1 activation and its role in SPLUNC1 regulation were investigated by Western blot, c-Jun activation assay, chromatin immunoprecipitation (ChIP) and real-time PCR. SPLUNC1 promoter activity was assessed by a luciferase reporter assay. RESULTS Pam(3)CSK(4) increased SPLUNC1 expression in NCI-H292 cells in a dose- and time-dependent manner, and enhanced SPLUNC1 promoter activity. Pam(3)CSK(4)-treated cells demonstrated activated MAPK and c-Jun compared to untreated cells. ChIP assay indicated increased c-Jun binding to the SPLUNC1 promoter following Pam(3)CSK(4) stimulation. Inhibition of ERK1/2 significantly reduced Pam(3)CSK(4)-mediated c-Jun activation and SPLUNC1 expression. CONCLUSIONS Our results for the first time demonstrate that TLR2-mediated MAPK/AP-1 activation up-regulates lung epithelial SPLUNC1 expression at the transcriptional level. Understanding SPLUNC1 gene regulation should provide more specific therapeutic targets to restore deficient SPLUNC1 production in diseased airways.
Collapse
Affiliation(s)
- Jyoti Thaikoottathil
- Department of Medicine, National Jewish Health, University of Colorado Denver, Denver, CO 80206, USA
| | | |
Collapse
|
188
|
Lippmann J, Müller HC, Naujoks J, Tabeling C, Shin S, Witzenrath M, Hellwig K, Kirschning CJ, Taylor GA, Barchet W, Bauer S, Suttorp N, Roy CR, Opitz B. Dissection of a type I interferon pathway in controlling bacterial intracellular infection in mice. Cell Microbiol 2011; 13:1668-82. [PMID: 21790939 DOI: 10.1111/j.1462-5822.2011.01646.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Defence mechanisms against intracellular bacterial pathogens are incompletely understood. Our study characterizes a type I IFN-dependent cell-autonomous defence pathway directed against Legionella pneumophila, an intracellular model organism and frequent cause of pneumonia. We show that macrophages infected with L. pneumophila produced IFNβ in a STING- and IRF3- dependent manner. Paracrine type I IFNs stimulated upregulation of IFN-stimulated genes and a cell-autonomous defence pathway acting on replicating and non-replicating Legionella within their specialized vacuole. Our infection experiments in mice lacking receptors for type I and/or II IFNs show that type I IFNs contribute to expression of IFN-stimulated genes and to bacterial clearance as well as resistance in L. pneumophila pneumonia in addition to type II IFN. Overall, our study shows that paracrine type I IFNs mediate defence against L. pneumophila, and demonstrates a protective role of type I IFNs in in vivo infections with intracellular bacteria.
Collapse
Affiliation(s)
- Juliane Lippmann
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Kim ES, Chang YS, Choi SJ, Kim JK, Yoo HS, Ahn SY, Sung DK, Kim SY, Park YR, Park WS. Intratracheal transplantation of human umbilical cord blood-derived mesenchymal stem cells attenuates Escherichia coli-induced acute lung injury in mice. Respir Res 2011; 12:108. [PMID: 21843339 PMCID: PMC3166924 DOI: 10.1186/1465-9921-12-108] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 08/15/2011] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Human umbilical cord blood (UCB)-derived mesenchymal stem cells (MSCs) attenuate hyperoxic neonatal lung injury primarily through anti-inflammatory effects. We hypothesized that intratracheal transplantation of human UCB-derived MSCs could attenuate Escherichia coli (E. coli)-induced acute lung injury (ALI) in mice by suppressing the inflammatory response. METHODS Eight-week-old male ICR mice were randomized to control or ALI groups. ALI was induced by intratracheal E. coli instillation. Three-hours after E. coli instillation, MSCs, fibroblasts or phosphate-buffered saline were intratracheally administered randomly and survival was analyzed for 7 days post-injury. Lung histology including injury scores, myeloperoxidase (MPO) activity, and protein levels of interleukin (IL)-1α, IL-1β, IL-6, tumor necrosis factor (TNF)-α, and macrophage inflammatory protein (MIP)-2 as well as the wet-dry lung ratio and bacterial counts from blood and bronchoalveolar lavage (BAL) were evaluated at 1, 3, and 7 days post-injury. Levels of inflammatory cytokines in the lung were also profiled using protein macroarrays at day 3 post-injury which showed peak inflammation. RESULTS MSC transplantation increased survival and attenuated lung injuries in ALI mice, as evidenced by decreased injury scores on day 3 post-injury and reduced lung inflammation including increased MPO activity and protein levels of IL-1α, IL-1β, IL-6, TNF-α, and MIP-2 on day 3 and 7 post-injury. Inflammatory cytokine profiles in the lungs at day 3 post-injury were attenuated by MSC transplantation. MSCs also reduced the elevated lung water content at day 3 post-injury and bacterial counts in blood and BAL on day 7 post-injury. CONCLUSIONS Intratracheal transplantation of UCB-derived MSCs attenuates E. coli-induced ALI primarily by down-modulating the inflammatory process and enhancing bacterial clearance.
Collapse
Affiliation(s)
- Eun Sun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Current world literature. Curr Opin Allergy Clin Immunol 2011; 11:150-6. [PMID: 21368622 DOI: 10.1097/aci.0b013e3283457ab0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
191
|
Role of innate immunity in the development of allergy and asthma. Curr Opin Allergy Clin Immunol 2011; 11:127-31. [PMID: 21325945 DOI: 10.1097/aci.0b013e32834487c6] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Asthma is essentially a developmental disease, in which the normal growth and development of the respiratory and immune systems are affected by environmental exposures acting on underlying genetic predispositions. The purpose of this review is to examine the role of innate immunity in the lungs in the development of allergy and asthma. RECENT FINDINGS Both the innate and adaptive arms of the immune system are immature at birth and undergo prolonged periods of postnatal maturation. As such, they are vulnerable to adverse environmental exposures, both before and after birth. Both genetic predispositions and environmentally induced epigenetic changes in gene expression are likely to contribute to the risk of asthma; however, the relative contributions are unclear. Increasing interest is focused on deficient innate responses of the respiratory epithelium to viral infections and how these may increase the risk of asthma. However, definitive proof that these are primary and not secondary effects is lacking. Although most research has concentrated on the role of respiratory viral infections in increasing the asthma risk, the recent suggestion that the lung has a resident bacteriome and potentially important viral-bacterial interactions in the lungs broadens research scope in this area. SUMMARY Classic risk factors for asthma include a family history of asthma and allergies, early and persistent allergic sensitization and viral lower-respiratory infections in early life. However, these factors do not fully explain the risk. Perhaps, the resident pulmonary microbiome and the immune response that this generates during respiratory viral infections will provide the 'missing link' in the epidemiology.
Collapse
|
192
|
Li Y, Wang J, He HY, Ma LJ, Zeng J, Deng GC, Liu X, Engelhardt JF, Wang Y. Immunohistochemical demonstration of airway epithelial cell markers of guinea pig. Tissue Cell 2011; 43:283-90. [PMID: 21705035 DOI: 10.1016/j.tice.2011.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 05/15/2011] [Accepted: 05/24/2011] [Indexed: 11/25/2022]
Abstract
The guinea pig (Cavea porcellus) is a mammalian non-rodent species in the Caviidae family. The sensitivity of the respiratory system and the susceptibility to infectious diseases allows the guinea pig to be a useful model for both infectious and non-infectious lung diseases such as asthma and tuberculosis. In this report, we demonstrated for the first time, the major cell types and composition in the guinea pig airway epithelium, using cell type-specific markers by immunohistochemical staining using the commercial available immunological reagents that cross-react with guinea pig. Our results revealed the availability of antibodies cross-reacting with airway epithelial cell types of basal, non-ciliated columnar, ciliated, Clara, goblet and alveolar type II cells, as well as those cells expressing Mucin 5AC, Mucin 2, Aquaporin 4 and Calcitonin Gene Related Peptide. The distribution of these various cell types were quantified in the guinea pig airway by immunohistochemical staining and were comparable with morphometric studies using an electron microscopy assay. Moreover, this study also demonstrated that goblet cells are the main secretory cell type in the guinea pig's airway, distinguishing this species from rats and mice. These results provide useful information for the understanding of airway epithelial cell biology and mechanisms of epithelial-immune integration in guinea pig models.
Collapse
Affiliation(s)
- Yong Li
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources of Western China, Yinchuan, Ningxia 750021, China
| | | | | | | | | | | | | | | | | |
Collapse
|
193
|
Witzenrath M, Pache F, Lorenz D, Koppe U, Gutbier B, Tabeling C, Reppe K, Meixenberger K, Dorhoi A, Ma J, Holmes A, Trendelenburg G, Heimesaat MM, Bereswill S, van der Linden M, Tschopp J, Mitchell TJ, Suttorp N, Opitz B. The NLRP3 Inflammasome Is Differentially Activated by Pneumolysin Variants and Contributes to Host Defense in Pneumococcal Pneumonia. THE JOURNAL OF IMMUNOLOGY 2011; 187:434-40. [DOI: 10.4049/jimmunol.1003143] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
194
|
Lennon FE, Singleton PA. Role of hyaluronan and hyaluronan-binding proteins in lung pathobiology. Am J Physiol Lung Cell Mol Physiol 2011; 301:L137-47. [PMID: 21571904 DOI: 10.1152/ajplung.00071.2010] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Hyaluronan (HA) has diverse functions in normal lung homeostasis and pulmonary disease. HA constitutes the major glycosaminoglycan in lung tissue, with HA degradation products, produced by hyaluronidase enzymes and reactive oxygen species, being implicated in several lung diseases, including acute lung injury, asthma, chronic obstructive pulmonary disease, and pulmonary hypertension. The differential activities of HA and its degradation products are due, in part, to regulation of multiple HA-binding proteins, including cluster of differentiation 44 (CD44), Toll-like receptor 4 (TLR4), HA-binding protein 2 (HABP2), and receptor for HA-mediated motility (RHAMM). Recent research indicates that exogenous administration of high-molecular-weight HA can serve as a novel therapeutic intervention for lung diseases, including lipopolysaccharide (LPS)-induced acute lung injury, sepsis/ventilator-induced lung injury, and airway hyperreactivity. This review focuses on the regulatory role of HA and HA-binding proteins in lung pathology and discusses the capacity of HA to augment and inhibit various lung diseases.
Collapse
Affiliation(s)
- Frances E Lennon
- Section of Pulmonary and Critical Care, Department of Medicine, Pritzker School of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | | |
Collapse
|
195
|
Reisetter AC, Stebounova LV, Baltrusaitis J, Powers L, Gupta A, Grassian VH, Monick MM. Induction of inflammasome-dependent pyroptosis by carbon black nanoparticles. J Biol Chem 2011; 286:21844-52. [PMID: 21525001 DOI: 10.1074/jbc.m111.238519] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Inhalation of nanoparticles has been implicated in respiratory morbidity and mortality. In particular, carbon black nanoparticles are found in many different environmental exposures. Macrophages take up inhaled nanoparticles and respond via release of inflammatory mediators and in some cases cell death. Based on new data, we propose that exposure of macrophages (both a macrophage cell line and primary human alveolar macrophages) to carbon black nanoparticles induces pyroptosis, an inflammasome-dependent form of cell death. Exposure of macrophages to carbon black nanoparticles resulted in inflammasome activation as defined by cleavage of caspase 1 to its active form and downstream IL-1β release. The cell death that occurred with carbon black nanoparticle exposure was identified as pyroptosis by the protective effect of a caspase 1 inhibitor and a pyroptosis inhibitor. These data demonstrate that carbon black nanoparticle exposure activates caspase 1, increases IL-1β release after LPS priming, and induces the proinflammatory cell death, pyroptosis. The identification of pyroptosis as a cellular response to carbon nanoparticle exposure is novel and relates to environmental and health impacts of carbon-based particulates.
Collapse
Affiliation(s)
- Anna C Reisetter
- Department of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | |
Collapse
|
196
|
Masuko H, Sakamoto T, Kaneko Y, Iijima H, Naito T, Noguchi E, Hirota T, Tamari M, Hizawa N. Lower FEV1 in non-COPD, nonasthmatic subjects: association with smoking, annual decline in FEV1, total IgE levels, and TSLP genotypes. Int J Chron Obstruct Pulmon Dis 2011; 6:181-9. [PMID: 21468164 PMCID: PMC3064418 DOI: 10.2147/copd.s16383] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Indexed: 12/02/2022] Open
Abstract
Few studies have investigated the significance of decreased FEV(1) in non-COPD, nonasthmatic healthy subjects. We hypothesized that a lower FEV(1) in these subjects is a potential marker of an increased susceptibility to obstructive lung disease such as asthma and COPD. This was a cross-sectional analysis of 1505 Japanese adults. We divided the population of healthy adults with no respiratory diseases whose FEV(1)/FVC ratio was ≥ 70% (n = 1369) into 2 groups according to their prebronchodilator FEV(1) (% predicted) measurements: < 80% (n = 217) and ≥ 80% (n = 1152). We compared clinical data - including gender, age, smoking habits, total IgE levels, and annual decline of FEV(1) - between these 2 groups. In addition, as our group recently found that TSLP variants are associated with asthma and reduced lung function, we assessed whether TSLP single nucleotide polymorphisms (SNPs) were associated with baseline lung function in non-COPD, nonasthmatic healthy subjects (n = 1368). Although about half of the subjects with lower FEV(1) had never smoked, smoking was the main risk factor for the decreased FEV(1) in non-COPD, nonasthmatic subjects. However, the subjects with lower FEV(1) had a significantly higher annual decline in FEV(1) independent of smoking status. Airflow obstruction was associated with increased levels of total serum IgE (P = 0.029) and with 2 functional TSLP SNPs (corrected P = 0.027-0.058 for FEV(1)% predicted, corrected P = 0.015-0.033 for FEV(1)/FVC). This study highlights the importance of early recognition of a decreased FEV(1) in healthy subjects without evident pulmonary diseases because it predicts a rapid decline in FEV(1) irrespective of smoking status. Our series of studies identified TSLP variants as a potential susceptibility locus to asthma and to lower lung function in non-COPD, nonasthmatic healthy subjects, which may support the contention that genetic determinants of lung function influence susceptibility to asthma.
Collapse
Affiliation(s)
- Hironori Masuko
- Division of Respiratory Medicine, Institute of Clinical Medicine, University of Tsukuba, Ibaraki, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Lafferty EI, Qureshi ST, Schnare M. The role of toll-like receptors in acute and chronic lung inflammation. JOURNAL OF INFLAMMATION-LONDON 2010; 7:57. [PMID: 21108806 PMCID: PMC3003652 DOI: 10.1186/1476-9255-7-57] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 11/25/2010] [Indexed: 12/13/2022]
Abstract
By virtue of its direct contact with the environment, the lung is constantly challenged by infectious and non-infectious stimuli that necessitate a robust yet highly controlled host response coordinated by the innate and adaptive arms of the immune system. Mammalian Toll-like receptors (TLRs) function as crucial sentinels of microbial and non-infectious antigens throughout the respiratory tract and mediate host innate immunity. Selective induction of inflammatory responses to harmful environmental exposures and tolerance to innocuous antigens are required to maintain tissue homeostasis and integrity. Conversely, dysregulated innate immune responses manifest as sustained and self-perpetuating tissue damage rather than controlled tissue repair. In this article we review aspects of Toll-like receptor function that are relevant to the development of acute lung injury and chronic obstructive lung diseases as well as resistance to frequently associated microbial infections.
Collapse
Affiliation(s)
- Erin I Lafferty
- Division of Experimental Medicine, McGill University, Montréal, Québec H3A 1A3, Canada.
| | | | | |
Collapse
|
198
|
Rohmann K, Tschernig T, Pabst R, Goldmann T, Drömann D. Innate immunity in the human lung: pathogen recognition and lung disease. Cell Tissue Res 2010; 343:167-74. [PMID: 20936302 PMCID: PMC7087881 DOI: 10.1007/s00441-010-1048-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Accepted: 09/01/2010] [Indexed: 11/27/2022]
Abstract
As the human lung is exposed to a variety of microbial pathogens in the environment, a first line of defense is built up by pulmonary cells like bronchial/alveolar epithelial cells and alveolar macrophages. These cells express several pattern recognition receptors (PRRs) recognizing highly conserved microbial motifs and initiating the production of chemokines and pro- and anti-inflammatory cytokines acting as transmembrane or intracellular receptors. This might not only lead to acute but also to chronic inflammation which is discussed as an underlying mechanism in the pathogenesis of different lung diseases.
Collapse
Affiliation(s)
- Kristina Rohmann
- Medical Clinic III, University of Schleswig-Holstein, Campus Lübeck, 23538 Lübeck, Germany
| | | | | | | | | |
Collapse
|
199
|
Hussell T, Goulding J. Structured regulation of inflammation during respiratory viral infection. THE LANCET. INFECTIOUS DISEASES 2010; 10:360-6. [PMID: 20417418 DOI: 10.1016/s1473-3099(10)70067-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Innate immune cells including macrophages, dendritic cells, and granulocytes are resident within or patrol very different microenvironments in the host. Their activity or responsiveness to antigen is dictated by site-specific factors. Because of the constant exposure to environmental antigens and commensal microorganisms, mucosal immunity needs to be more constrained than peripheral counterparts to prevent unnecessary inflammation. The epithelial surfaces that dominate all mucosal tissues provide an ideal regulator since innate immune cells are often in intimate contact with, or lie immediately beneath, them and a breach in epithelial integrity would signal a damaging event and release innate immunity from their influence. We discuss the role of the respiratory epithelium in raising the threshold of innate immune cell activation at homoeostasis, how its absence triggers innate immunity, and how inflammatory resolution often produces an altered homoeostatic environment that can affect the next inflammatory event at this site.
Collapse
Affiliation(s)
- Tracy Hussell
- Imperial College London, Leukocyte Biology Section, National Heart and Lung Institute, London, UK.
| | | |
Collapse
|