851
|
McMillan SJ, Sharma RS, Richards HE, Hegde V, Crocker PR. Siglec-E promotes β2-integrin-dependent NADPH oxidase activation to suppress neutrophil recruitment to the lung. J Biol Chem 2014; 289:20370-6. [PMID: 24895121 PMCID: PMC4106349 DOI: 10.1074/jbc.m114.574624] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Siglec-E is a sialic acid-binding Ig-like lectin expressed on murine myeloid cells. It has recently been shown to function as a negative regulator of β2-integrin-dependent neutrophil recruitment to the lung following exposure to lipopolysaccharide (LPS). Here, we demonstrate that siglec-E promoted neutrophil production of reactive oxygen species (ROS) following CD11b β2-integrin ligation with fibrinogen in a sialic acid-dependent manner, but it had no effect on ROS triggered by a variety of other stimulants. Siglec-E promotion of ROS was likely mediated via Akt activation, because siglec-E-deficient neutrophils plated on fibrinogen exhibited reduced phosphorylation of Akt, and the Akt inhibitor, MK2206, blocked fibrinogen-induced ROS. In vivo imaging showed that siglec-E also promoted ROS in acutely inflamed lungs following exposure of mice to LPS. Importantly, siglec-E-promoted ROS were required for its inhibitory function, as the NADPH oxidase inhibitor, apocynin, reversed the siglec-E-mediated suppression of neutrophil recruitment and blocked neutrophil ROS production in vitro. Taken together, these results demonstrate that siglec-E functions as an inhibitory receptor of neutrophils via positive regulation of NADPH oxidase activation and ROS production. Our findings have implications for the inhibitory role of siglec-9 on human neutrophils in sepsis and acute lung injury.
Collapse
Affiliation(s)
- Sarah J McMillan
- From the Department of Cell Signalling and Immunology, College of Life Sciences, and
| | - Ritu S Sharma
- From the Department of Cell Signalling and Immunology, College of Life Sciences, and
| | - Hannah E Richards
- From the Department of Cell Signalling and Immunology, College of Life Sciences, and
| | - Vikas Hegde
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Paul R Crocker
- From the Department of Cell Signalling and Immunology, College of Life Sciences, and
| |
Collapse
|
852
|
Zhang S, Hwaiz R, Rahman M, Herwald H, Thorlacius H. Ras regulates alveolar macrophage formation of CXC chemokines and neutrophil activation in streptococcal M1 protein-induced lung injury. Eur J Pharmacol 2014; 733:45-53. [DOI: 10.1016/j.ejphar.2014.03.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 03/17/2014] [Accepted: 03/24/2014] [Indexed: 11/16/2022]
|
853
|
Young RW. Prevention of lung injury in cardiac surgery: a review. THE JOURNAL OF EXTRA-CORPOREAL TECHNOLOGY 2014; 46:130-41. [PMID: 25208430 PMCID: PMC4566423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 06/07/2014] [Indexed: 06/03/2023]
Abstract
Inflammatory lung injury is an inevitable consequence of cardiac surgery with cardiopulmonary bypass. The lungs are particularly susceptible to the effects of the systemic inflammatory response to cardiopulmonary bypass. This insult is further exacerbated by a pulmonary ischemia-reperfusion injury after termination of bypass. Older patients and those with pre-existing lung disease will clearly be less tolerant of any lung injury and more likely to develop respiratory failure in the postoperative period. A requirement for prolonged ventilation has implications for morbidity, mortality, and cost of treatment. This review contains a summary of recent interventions and changes of practice that may reduce inflammatory lung injury after cardiac surgery. The review also focuses on a number of general aspects of perioperative management, which may exacerbate such injury, if performed poorly.
Collapse
|
854
|
Zhao L, Wang L, Zhang X, Sun L, Sun D, Sun T. Reduced levels of interleukin‑1 receptor antagonist act as a marker for pneumonia in the elderly. Mol Med Rep 2014; 10:959-64. [PMID: 24888337 DOI: 10.3892/mmr.2014.2284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 04/14/2014] [Indexed: 11/06/2022] Open
Abstract
Pneumonia is a disease causing serious inflammation and infection of the lungs and accounts for >50,000 mortalities annually. The elderly are at an increased risk of developing pneumonia. Pneumonia is more serious in the elderly than in any other age group due to the increased inflammation and risk of community‑acquired pneumonia associated with aging. Interleukin‑1 receptor antagonist (IL‑1RA) is an anti‑inflammatory protein that counteracts the destructive effects of inflammatory proteins. Therefore, the possible association between pneumonia in elderly individuals and reduced levels of IL‑1RA was investigated in the present study. The number of lymphocytes was counted in all subjects and the relative protein expression levels of IL‑1RA were determined using western blot analysis. In addition, the immunological activities of IL‑1RA were measured using ELISA. The results demonstrated that the numbers of lymphocytes in the serum and bronchoalveolar lavage fluid (BALF) were significantly higher in elderly patients than those in young patients. Furthermore, the serum and BALF levels of IL‑1RA in elderly patients were significantly lower than those in young patients (P<0.05 and P<0.01, respectively). Therefore, reduced levels of IL‑1RA in BALF may act as a marker for pneumonia in the elderly and may be a potential adjuvant for the diagnosis of pneumonia in elderly individuals. The results also showed that smoking was associated with significant reductions in the levels of IL‑1RA in the BALF of elderly patients. The association between smokers and non‑smokers found in this study provides support for the hypothesis that smoking may contribute to the pathogenesis of pneumonia by further reducing IL‑IRA levels in certain elderly patients.
Collapse
Affiliation(s)
- Lijie Zhao
- Department of Geriatrics, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163311, P.R. China
| | - Lina Wang
- Department of Geriatrics, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163311, P.R. China
| | - Xiaoyan Zhang
- Department of Geriatrics, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163311, P.R. China
| | - Lili Sun
- Department of Geriatrics, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163311, P.R. China
| | - Dan Sun
- Department of Geriatrics, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163311, P.R. China
| | - Tingli Sun
- Department of Nephrology, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163311, P.R. China
| |
Collapse
|
855
|
Wang H, Xu L, Zhao J, Wang D, Guo R, Wang J, Gong W, Liu T, Zhang Y, Dong L. Regulatory mechanism of pyrrolidine dithiocarbamate is mediated by nuclear factor-κB and inhibits neutrophil accumulation in ARDS mice. Exp Ther Med 2014; 8:614-622. [PMID: 25009629 PMCID: PMC4079437 DOI: 10.3892/etm.2014.1738] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/29/2014] [Indexed: 01/12/2023] Open
Abstract
The aim of the present study was to investigate the regulatory mechanism of nuclear factor (NF)-κB on polymorphonuclear neutrophil (PMN) accumulation and the inflammatory response in lung tissues with acute respiratory distress syndrome (ARDS), as well as the therapeutic effect of pyrrolidine dithiocarbamate (PDTC). Mouse models of ARDS were established by intraperitoneal injection of lipopolysaccharide (LPS). BALB/c mice were divided into control, LPS and PDTC + LPS groups. The expression of PMN adhesion molecules, CD11b/CD18 and intercellular adhesion molecule-1 (ICAM-1), were detected by immunohistochemistry, while the protein expression levels of NF-κB p65 in the lung tissue were analyzed by western blot analysis. In addition, flow cytometry was used to investigate the apoptosis rate of PMNs in the bronchoalveolar fluid, and the expression levels of interleukin (IL)-1β, IL-8 and tumor necrosis factor (TNF)-α and myeloperoxidase (MPO) activity were also determined. Following an intraperitoneal injection of LPS, alveolar septum rupture, pulmonary interstitial hyperemia and PMN infiltration in the alveolar was observed. The protein expression of p65 in the pulmonary cytoplasm decreased, while the expression of p65 in the nucleus increased. The levels of IL-8, IL-1β and TNF-α increased and the high expression status was maintained for 24 h. As the time increased, CD11b/CD18 and ICAM-1 expression increased, as well as MPO activity, while the apoptosis of PMNs was delayed. Compared with the LPS group, the expression of p65 in the pulmonary cytoplasm and the PMN apoptosis rate increased following PDTC intervention, while the expression of p65 in the nucleus decreased, as well as the expression levels of the cytokines and MPO activity. Therefore, PDTC reduced the production of inflammatory cytokines via the NF-κB pathway, which reduced the activation of PMNs in the lung tissue and promoted PMN apoptosis.
Collapse
Affiliation(s)
- Hongman Wang
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, Shangdong 250012, P.R. China ; Department of Pulmonary Medicine, The Thrid Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China ; Department of Pulmonary Medicine, The Fifth Affiliated Hospital of Zunyi Medical University Zhuhai, Zhuhai, Guangdong 519100, P.R. China
| | - Lisheng Xu
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, Shangdong 250012, P.R. China
| | - Jiping Zhao
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, Shangdong 250012, P.R. China
| | - Donghui Wang
- Department of Pulmonary Medicine, The Thrid Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Ranran Guo
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, Shangdong 250012, P.R. China ; Department of Pulmonary Medicine, The Thrid Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Junfei Wang
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, Shangdong 250012, P.R. China
| | - Wenbin Gong
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, Shangdong 250012, P.R. China
| | - Tian Liu
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, Shangdong 250012, P.R. China
| | - Yuanyuan Zhang
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, Shangdong 250012, P.R. China
| | - Liang Dong
- Department of Pulmonary Medicine, Qilu Hospital, Shandong University, Jinan, Shangdong 250012, P.R. China
| |
Collapse
|
856
|
Chrysanthopoulou A, Mitroulis I, Apostolidou E, Arelaki S, Mikroulis D, Konstantinidis T, Sivridis E, Koffa M, Giatromanolaki A, Boumpas DT, Ritis K, Kambas K. Neutrophil extracellular traps promote differentiation and function of fibroblasts. J Pathol 2014; 233:294-307. [PMID: 24740698 DOI: 10.1002/path.4359] [Citation(s) in RCA: 248] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 04/02/2014] [Accepted: 04/07/2014] [Indexed: 12/19/2022]
Abstract
Neutrophil activation by inflammatory stimuli and the release of extracellular chromatin structures (neutrophil extracellular traps - NETs) have been implicated in inflammatory disorders. Herein, we demonstrate that NETs released by neutrophils treated either with fibrosis-related agents, such as cigarette smoke, magnesium silicate, bleomycin, or with generic NET inducers, such as phorbol 12-myristate 13-acetate, induced activation of lung fibroblasts (LFs) and differentiation into myofibroblast (MF) phenotype. Interestingly, the aforementioned agents or IL-17 (a primary initiator of inflammation/fibrosis) had no direct effect on LF activation and differentiation. MFs treated with NETs demonstrated increased connective tissue growth factor expression, collagen production, and proliferation/migration. These fibrotic effects were significantly decreased after degradation of NETs with DNase1, heparin or myeloperoxidase inhibitor, indicating the key role of NET-derived components in LF differentiation and function. Furthermore, IL-17 was expressed in NETs and promoted the fibrotic activity of differentiated LFs but not their differentiation, suggesting that priming by DNA and histones is essential for IL-17-driven fibrosis. Additionally, autophagy was identified as the orchestrator of NET formation, as shown by inhibition studies using bafilomycin A1 or wortmannin. The above findings were further supported by the detection of NETs in close proximity to alpha-smooth muscle actin (α-SMA)-expressing fibroblasts in biopsies from patients with fibrotic interstitial lung disease or from skin scar tissue. Together, these data suggest that both autophagy and NETs are involved not only in inflammation but also in the ensuing fibrosis and thus may represent potential therapeutic targets in human fibrotic diseases.
Collapse
Affiliation(s)
- Akrivi Chrysanthopoulou
- Laboratory of Molecular Hematology, Democritus University of Thrace, Alexandroupolis, Greece
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
857
|
Balakrishna S, Song W, Achanta S, Doran SF, Liu B, Kaelberer MM, Yu Z, Sui A, Cheung M, Leishman E, Eidam HS, Ye G, Willette RN, Thorneloe KS, Bradshaw HB, Matalon S, Jordt SE. TRPV4 inhibition counteracts edema and inflammation and improves pulmonary function and oxygen saturation in chemically induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 2014; 307:L158-72. [PMID: 24838754 DOI: 10.1152/ajplung.00065.2014] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The treatment of acute lung injury caused by exposure to reactive chemicals remains challenging because of the lack of mechanism-based therapeutic approaches. Recent studies have shown that transient receptor potential vanilloid 4 (TRPV4), an ion channel expressed in pulmonary tissues, is a crucial mediator of pressure-induced damage associated with ventilator-induced lung injury, heart failure, and infarction. Here, we examined the effects of two novel TRPV4 inhibitors in mice exposed to hydrochloric acid, mimicking acid exposure and acid aspiration injury, and to chlorine gas, a severe chemical threat with frequent exposures in domestic and occupational environments and in transportation accidents. Postexposure treatment with a TRPV4 inhibitor suppressed acid-induced pulmonary inflammation by diminishing neutrophils, macrophages, and associated chemokines and cytokines, while improving tissue pathology. These effects were recapitulated in TRPV4-deficient mice. TRPV4 inhibitors had similar anti-inflammatory effects in chlorine-exposed mice and inhibited vascular leakage, airway hyperreactivity, and increase in elastance, while improving blood oxygen saturation. In both models of lung injury we detected increased concentrations of N-acylamides, a class of endogenous TRP channel agonists. Taken together, we demonstrate that TRPV4 inhibitors are potent and efficacious countermeasures against severe chemical exposures, acting against exaggerated inflammatory responses, and protecting tissue barriers and cardiovascular function.
Collapse
Affiliation(s)
- Shrilatha Balakrishna
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut
| | - Weifeng Song
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Satyanarayana Achanta
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut
| | - Stephen F Doran
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Boyi Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut
| | - Melanie M Kaelberer
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| | - Zhihong Yu
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Aiwei Sui
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut
| | - Mui Cheung
- Heart Failure Discovery Performance Unit-Metabolic Pathways and Cardiovascular Therapy Unit, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania
| | - Emma Leishman
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana; and
| | - Hilary S Eidam
- Heart Failure Discovery Performance Unit-Metabolic Pathways and Cardiovascular Therapy Unit, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania
| | - Guosen Ye
- Heart Failure Discovery Performance Unit-Metabolic Pathways and Cardiovascular Therapy Unit, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania
| | - Robert N Willette
- Heart Failure Discovery Performance Unit-Metabolic Pathways and Cardiovascular Therapy Unit, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania
| | - Kevin S Thorneloe
- Heart Failure Discovery Performance Unit-Metabolic Pathways and Cardiovascular Therapy Unit, GlaxoSmithKline Pharmaceuticals, King of Prussia, Pennsylvania
| | - Heather B Bradshaw
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana; and
| | - Sadis Matalon
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sven-Eric Jordt
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut;
| |
Collapse
|
858
|
Hästbacka J, Linko R, Tervahartiala T, Varpula T, Hovilehto S, Parviainen I, Vaara ST, Sorsa T, Pettilä V. Serum MMP-8 and TIMP-1 in critically ill patients with acute respiratory failure: TIMP-1 is associated with increased 90-day mortality. Anesth Analg 2014; 118:790-8. [PMID: 24651234 DOI: 10.1213/ane.0000000000000120] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Matrix metalloproteinases (MMPs) likely have an important role in the pathophysiology of acute lung injury. In a recent study, high matrix metalloproteinases (MMP-8) levels in tracheal aspirates of pediatric acute respiratory distress syndrome (ARDS) patients were associated with worse outcome. In patients with sepsis, an imbalance between MMPs and their tissue inhibitors (TIMPs) has been associated with impaired survival. We hypothesized that the elevated systemic MMP-8 and TIMP-1 are associated with worse outcome in acute respiratory failure. METHODS This was a substudy of the observational FINNALI study conducted in 25 Finnish intensive care units over an 8-week period. All patients older than 16 years requiring mechanical ventilation for >6 hours were included. MMP-8 and TIMP-1 levels were analyzed from blood samples taken on enrollment in the study and 48 hours later. Laboratory analyses were performed by using immunofluorometric assay for MMP-8 and ELISA for TIMP-1. MMP-8 and TIMP-1 levels were compared between 90-day survivors and nonsurvivors. Survival was compared in quartiles based on TIMP-1 levels, and ROC analysis was performed to calculate areas under the curves. The relationship between MMP-8 and TIMP-1 levels and degree of hypoxemia was examined. RESULTS The final analyses included 563 patients. Admission TIMP-1 levels were higher in nonsurvivors, median 367 ng/mL (interquartile range 199-562), than survivors, median 240 ng/mL (interquartile range 142-412), WMWodds 1.68 (95% confidence interval [CI], 1.43-2.08). MMP-8 levels may have differed between survivors and nonsurvivors, WMWodds 1.20 (95% CI, 1.01-1.43), but no difference was found in the MMP-8/TIMP-1 molar ratio, WMWodds 0.83 (95% CI, 0.67-1.04). Difference in survival between quartiles based on TIMP-1 was significant (log-rank, P < 0.001). ROC analysis produced an area under the curve 0.63 (95% CI, 0.58-0.69) for TIMP-1. TIMP-1 was associated with severity of hypoxemia. TIMP-1 levels were higher in an ARDS subgroup than in the whole cohort, WMWodds 1.65 (95% CI, 1.15-2.44). CONCLUSIONS MMP-8 levels were possibly higher in 90-day nonsurvivors but performed poorly in predicting outcome. Increased systemic levels of TIMP-1 were associated with more severe hypoxemia and worse outcome in a large cohort of mechanically ventilated critically ill patients and in a subgroup of ARDS patients.
Collapse
Affiliation(s)
- Johanna Hästbacka
- From the *Intensive Care Units, Helsinki University Hospital; †Department of Oral and Maxillofacial Diseases, Helsinki University Hospital and Biomedicum Helsinki, Helsinki; ‡Intensive Care Unit, South Carelia Central Hospital, Lappeenranta; and §Department of Anesthesiology and Intensive Care, Kuopio University Hospital, Kuopio, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
859
|
Wang H, Wang S, Tang A, Gong H, Ma P, Chen L. Combined effects of sivelestat and resveratrol on severe acute pancreatitis-associated lung injury in rats. Exp Lung Res 2014; 40:288-97. [PMID: 24785170 DOI: 10.3109/01902148.2014.908249] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Despite extensive research and clinical efforts made in the management of acute pancre-atitis during the past few decades, to date no effective cure is available and the mortality from severe acute pancre-atitis remains high. Given that lung is the primary cause of early death in acute pancreatitis patients, novel therapeutic approaches aiming to prevent lung injury have become a subject of intensive investigation. In a previous study, we demonstrated that sivelestat, a specific inhibitor of neutrophil elastase, is effective in protecting against lung failure in rats with taurocholate-induced acute pancreatitis. As part of the analyses extended from that study, the present study aimed to evaluate the role of sivelestat and/or resveratrol in the protection against acute pancreatitis-associated lung injury. The extended analyses demonstrated the following: (1) sodium taurocholate induced apparent lung injury and dysfunction manifested by histological anomalies, including vacuolization and apoptosis of the cells in the lung, as well as biochemical aberrations in the blood (an increase in amylase concentration and a decrease in partial arterial oxygen pressure) and increases in activities of reactive oxygen species, interleukin 6, myeloperoxidase, neutrophil elastase, lung edema, bronchotracho alveolar lavage protein concentration, and bronchotracho alveolar lavage cell infiltration in the lung; and (2) in lung tissues, either sivelestat or resveratrol treatment effectively attenuated the taurocholate-induced abnormalities in all parameters analyzed except for serum amylase concentration. In addition, combined treatment with both sivelestat and resveratrol demonstrated additive protective effects on pancreatitis-associated lung injury compared with single treatment.
Collapse
Affiliation(s)
- Houhong Wang
- 1Department of Surgery, Zhejiang University School of Medicine, Second Affiliated Hospital, Hangzhou, China
| | | | | | | | | | | |
Collapse
|
860
|
Nieto-Torres JL, DeDiego ML, Verdiá-Báguena C, Jimenez-Guardeño JM, Regla-Nava JA, Fernandez-Delgado R, Castaño-Rodriguez C, Alcaraz A, Torres J, Aguilella VM, Enjuanes L. Severe acute respiratory syndrome coronavirus envelope protein ion channel activity promotes virus fitness and pathogenesis. PLoS Pathog 2014; 10:e1004077. [PMID: 24788150 PMCID: PMC4006877 DOI: 10.1371/journal.ppat.1004077] [Citation(s) in RCA: 358] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 03/05/2014] [Indexed: 01/12/2023] Open
Abstract
Deletion of Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) envelope (E) gene attenuates the virus. E gene encodes a small multifunctional protein that possesses ion channel (IC) activity, an important function in virus-host interaction. To test the contribution of E protein IC activity in virus pathogenesis, two recombinant mouse-adapted SARS-CoVs, each containing one single amino acid mutation that suppressed ion conductivity, were engineered. After serial infections, mutant viruses, in general, incorporated compensatory mutations within E gene that rendered active ion channels. Furthermore, IC activity conferred better fitness in competition assays, suggesting that ion conductivity represents an advantage for the virus. Interestingly, mice infected with viruses displaying E protein IC activity, either with the wild-type E protein sequence or with the revertants that restored ion transport, rapidly lost weight and died. In contrast, mice infected with mutants lacking IC activity, which did not incorporate mutations within E gene during the experiment, recovered from disease and most survived. Knocking down E protein IC activity did not significantly affect virus growth in infected mice but decreased edema accumulation, the major determinant of acute respiratory distress syndrome (ARDS) leading to death. Reduced edema correlated with lung epithelia integrity and proper localization of Na+/K+ ATPase, which participates in edema resolution. Levels of inflammasome-activated IL-1β were reduced in the lung airways of the animals infected with viruses lacking E protein IC activity, indicating that E protein IC function is required for inflammasome activation. Reduction of IL-1β was accompanied by diminished amounts of TNF and IL-6 in the absence of E protein ion conductivity. All these key cytokines promote the progression of lung damage and ARDS pathology. In conclusion, E protein IC activity represents a new determinant for SARS-CoV virulence. Several highly pathogenic viruses encode small transmembrane proteins with ion-conduction properties named viroporins. Viroporins are generally involved in virus production and maturation processes, which many times are achieved by altering the ion homeostasis of cell organelles. Cells have evolved mechanisms to sense these imbalances in ion concentrations as a danger signal, and consequently trigger the innate immune system. Recently, it has been demonstrated that viroporins are inducers of cytosolic macromolecular complexes named inflammasomes that trigger the activation of key inflammatory cytokines such as IL-1β. The repercussions of this system in viral pathogenesis or disease outcome are currently being explored. SARS-CoV infection induces an uncontrolled inflammatory response leading to pulmonary damage, edema accumulation, severe hypoxemia and eventually death. In this study, we report that SARS-CoV E protein ion channel activity is a determinant of virulence, as the elimination of this function attenuated the virus, reducing the harmful inflammatory cytokine burst produced after infection, in which inflammasome activation plays a critical role. This led to less pulmonary damage and to disease resolution. These novel findings may be of relevance for other viral infections and can possibly be translated in order to find therapies for their associated diseases.
Collapse
Affiliation(s)
- Jose L. Nieto-Torres
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta L. DeDiego
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmina Verdiá-Báguena
- Department of Physics, Laboratory of Molecular Biophysics. Universitat Jaume I, Castellón, Spain
| | - Jose M. Jimenez-Guardeño
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Jose A. Regla-Nava
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Raul Fernandez-Delgado
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Castaño-Rodriguez
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio Alcaraz
- Department of Physics, Laboratory of Molecular Biophysics. Universitat Jaume I, Castellón, Spain
| | - Jaume Torres
- School of Biological Sciences, Division of Structural and Computational Biology, Nanyang Technological University, Singapore, Singapore
| | - Vicente M. Aguilella
- Department of Physics, Laboratory of Molecular Biophysics. Universitat Jaume I, Castellón, Spain
| | - Luis Enjuanes
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail:
| |
Collapse
|
861
|
Jones HD, Crother TR, Gonzalez-Villalobos RA, Jupelli M, Chen S, Dagvadorj J, Arditi M, Shimada K. The NLRP3 inflammasome is required for the development of hypoxemia in LPS/mechanical ventilation acute lung injury. Am J Respir Cell Mol Biol 2014; 50:270-80. [PMID: 24007300 DOI: 10.1165/rcmb.2013-0087oc] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IL-1β is a potent proinflammatory cytokine that is implicated in the pathogenesis of acute respiratory distress syndrome. We hypothesized that LPS and mechanical ventilation (MV) together could lead to IL-1β secretion and the development of acute lung injury (ALI), and that this process would be dependent on caspase-1 and the nucleotide binding domain and leucine-rich repeat (NLR) pyrin domain containing 3 (NLRP3) inflammasome activation. The objectives of this study were to determine the specific role of IL-1β, caspase-1, and the NLRP3 inflammasome in a two-hit model of ALI due to LPS plus MV. We used a two-hit murine model of ALI in which both inhaled LPS and MV were required for the development of hypoxemia, pulmonary neutrophil infiltration, and alveolar leakage. Nlrp3-deficent and Casp1-deficient mice had significantly diminished IL-1β levels in bronchoalveolar lavage fluid, and were specifically protected from hypoxemia, despite similar alveolar neutrophil infiltration and leakage. The IL-1 receptor antagonist, Anakinra, significantly improved the specific development of hypoxemia without significant effects on neutrophil infiltration or alveolar leakage. MV resulted in increased bronchoalveolar lavage extracellular ATP and alveolar macrophage apoptosis as triggers of NLRP3 inflammasome activation. NLRP3 inflammasome activation and IL-1β production play a key role in ALI caused by the combination of LPS and MV, particularly in the hypoxemia associated with acute respiratory distress syndrome. Blocking IL-1 signaling in this model specifically ameliorates hypoxemia, without affecting neutrophil infiltration and alveolar leakage, disassociating these readouts of ALI. MV causes alveolar macrophage apoptosis, a key step in the activation of NLRP3 inflammasome and production of IL-1β.
Collapse
|
862
|
Mercer PF, Williams AE, Scotton CJ, José RJ, Sulikowski M, Moffatt JD, Murray LA, Chambers RC. Proteinase-activated receptor-1, CCL2, and CCL7 regulate acute neutrophilic lung inflammation. Am J Respir Cell Mol Biol 2014; 50:144-57. [PMID: 23972264 DOI: 10.1165/rcmb.2013-0142oc] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
PAR1 plays a central role in mediating the interplay between coagulation and inflammation, but its role in regulating acute neutrophilic inflammation is unknown. We report that antagonism of PAR1 was highly effective at reducing acute neutrophil accumulation in a mouse model of LPS-induced lung inflammation. PAR1 antagonism also reduced alveolar-capillary barrier disruption in these mice. This protection was associated with a reduction in the expression of the chemokines, CCL2 and CCL7, but not the proinflammatory cytokines, TNF and IL-6, or the classic neutrophil chemoattractants, CXCL1 and CXCL2. Antibody neutralization of CCL2 and CCL7 significantly reduced LPS-induced total leukocyte and neutrophil accumulation, recovered from the bronchoalveolar lavage fluid of challenged mice. Immunohistochemical analysis revealed that CCL2 predominantly localized to alveolar macrophages and pulmonary epithelial cells, whereas CCL7 was restricted to the pulmonary epithelium. In keeping with these observations, the intranasal administration of recombinant CCL2 (rCCL2) and rCCL7 led to the accumulation of neutrophils within the lung airspaces of naive mice in the absence of any underlying inflammation. Flow cytometry analysis further demonstrated an increase in Ly6G(hi) neutrophils expressing the chemokine receptors, CCR1 and CCR2, isolated from mouse lungs compared with circulating neutrophils. Conversely, the expression of CXCR2 decreased on neutrophils isolated from the lung compared with circulating neutrophils. Furthermore, this switch in chemokine receptor expression was accentuated after acute LPS-induced lung inflammation. Collectively, these findings reveal a novel role for PAR1 and the chemokines, CCL2 and CCL7, during the early events of acute neutrophilic inflammation.
Collapse
Affiliation(s)
- Paul F Mercer
- 1 Centre for Inflammation and Tissue Repair, University College London, London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
863
|
Shu YS, Tao W, Miao QB, Lu SC, Zhu YB. Galangin Dampens Mice Lipopolysaccharide-Induced Acute Lung Injury. Inflammation 2014; 37:1661-8. [DOI: 10.1007/s10753-014-9894-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
864
|
Baluk P, Phillips K, Yao LC, Adams A, Nitschké M, McDonald DM. Neutrophil dependence of vascular remodeling after Mycoplasma infection of mouse airways. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1877-89. [PMID: 24726646 DOI: 10.1016/j.ajpath.2014.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 02/20/2014] [Accepted: 02/25/2014] [Indexed: 12/22/2022]
Abstract
Vascular remodeling is a feature of sustained inflammation in which capillaries enlarge and acquire the phenotype of venules specialized for plasma leakage and leukocyte recruitment. We sought to determine whether neutrophils are required for vascular remodeling in the respiratory tract by using Mycoplasma pulmonis infection as a model of sustained inflammation in mice. The time course of vascular remodeling coincided with the influx of neutrophils during the first few days after infection and peaked at day 5. Depletion of neutrophils with antibody RB6-8C5 or 1A8 reduced neutrophil influx and vascular remodeling after infection by about 90%. Similarly, vascular remodeling after infection was suppressed in Cxcr2(-/-) mice, in which neutrophils adhered to the endothelium of venules but did not extravasate into the tissue. Expression of the venular adhesion molecule P-selectin increased in endothelial cells from day 1 to day 3 after infection, as did expression of the Cxcr2-receptor ligands Cxcl1 and Cxcl2. Tumor necrosis factor α (TNFα) expression increased more than sixfold in the trachea of wild-type and Cxcr2(-/-) mice, but intratracheal administration of TNFα did not induce vascular remodeling similar to that seen in infection. We conclude that neutrophil influx is required for remodeling of capillaries into venules in the airways of mice with Mycoplasma infection and that TNFα signaling is necessary but not sufficient for vascular remodeling.
Collapse
Affiliation(s)
- Peter Baluk
- Cardiovascular Research Institute, Department of Anatomy, and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.
| | - Keeley Phillips
- Cardiovascular Research Institute, Department of Anatomy, and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Li-Chin Yao
- Cardiovascular Research Institute, Department of Anatomy, and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Alicia Adams
- Cardiovascular Research Institute, Department of Anatomy, and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Maximilian Nitschké
- Cardiovascular Research Institute, Department of Anatomy, and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Donald M McDonald
- Cardiovascular Research Institute, Department of Anatomy, and the Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| |
Collapse
|
865
|
Tsai CL, Lin YC, Wang HM, Chou TC. Baicalein, an active component of Scutellaria baicalensis, protects against lipopolysaccharide-induced acute lung injury in rats. JOURNAL OF ETHNOPHARMACOLOGY 2014; 153:197-206. [PMID: 24534526 DOI: 10.1016/j.jep.2014.02.010] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 02/05/2014] [Accepted: 02/08/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Baicalein (BE), a phenolic flavonoid extracted mainly from the root of Scutellaria baicalensis Georgi, a Chinese herb, is traditionally used in oriental medicine. Several studies have demonstrated that BE exerts many beneficial effects including anti-inflammatory and antioxidant activities. However, its effect on acute lung injury (ALI) and the molecular mechanisms involved remain unclear and warrant further investigation. The aim of the study is to investigate whether BE improves lipopolysaccharide (LPS, intratracheally, i.t.)-induced ALI in rats, and further study the underlying mechanisms of its activity. MATERIAL AND METHODS Rats were administrated with LPS (5mg/kg/body weight, i.t.) through a 24-gauge catheter to establish the ALI model. The effects of BE on the levels of pro-inflammatory cytokines, nitrite/nitrate in bronchoalveolar lavage fluid, and the expression of nuclear factor-erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1) and nuclear factor-kappa B (NF-κB) activation as well as the histopathological changes were evaluated. RESULTS Results showed that BE (20mg/kg, i.p.) treatment markedly attenuated LPS-induced lung edema, elevation of the levels of IL-1β, TNF-α, IL-6, CINC-3, and nitrite/nitrate in bronchoalveolar lavage fluid accompanied by a remarkable improvement of lung histopathological symptoms. The LPS-enhanced inflammatory cell infiltration and myeloperoxidase activity, O2(-) formation and the expression of inducible nitric oxide synthase and nitrotyrosin in lungs were all attenuated by BE. Notably, BE could augment Nrf2/HO-1 cascade, but inhibited NF-κB activation in LPS-instilled lungs that was strongly reversed by blocking HO-1 activity. CONCLUSION This study is the first to demonstrate that BE protects against LPS-induced ALI in rats. The underlying mechanisms may include inhibition of NF-κB-mediated inflammatory responses and upregulation of Nrf2/HO-1 pathway, which ultimately alleviates the pathological symptoms of ALI.
Collapse
Affiliation(s)
- Chen-Liang Tsai
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan
| | - Yu-Chieh Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hui-Min Wang
- Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tz-Chong Chou
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan; China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
866
|
Li LF, Lai YT, Chang CH, Lin MC, Liu YY, Kao KC, Tsai YH. Neutrophil elastase inhibitor reduces ventilation-induced lung injury via nuclear factor-κB and NF-κB repressing factor in mice. Exp Biol Med (Maywood) 2014; 239:1045-1057. [PMID: 24728725 DOI: 10.1177/1535370214529393] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Mechanical ventilation used in patients with acute lung injury can damage pulmonary epithelial cells through production of inflammatory cytokines, oxygen radicals, and neutrophil infiltration, termed ventilator-induced lung injury. Neutrophil elastase, nuclear factor-κB (NF-κB), and NF-κB repressing factor (NRF) have previously been shown to participate in the regulation of macrophage inflammatory protein-2 (MIP-2) during airway inflammation. However, the mechanisms regulating interactions among mechanical ventilation, neutrophil influx, and NF-κB/NRF remain unclear. Thus, we hypothesized that neutrophil elastase inhibitor attenuated ventilation-induced neutrophil recruitment and MIP-2 production through inhibition of the NF-κB/NRF pathway. Male C57BL/6 mice were exposed to low-tidal-volume (6 mL/kg) or high-tidal-volume (30 mL/kg) mechanical ventilation using room air with or without 2 µg/g NF-κB inhibitor SN50 or 6 µg/g NRF short interfering RNA or 100 µg/g neutrophil elastase inhibitor administration. Nonventilated mice served as a control group. Evan blue dye, lung wet-to-dry weight ratio, free radicals, myeloperoxidase, histopathologic grading of lung tissue, inflammatory cytokines, Western blot of NF-κB and NRF, and gene expression of NRF were measured to establish the extent of lung injury. Neutrophil elastase inhibitor ameliorated high-tidal-volume ventilation-induced lung injury, neutrophil influx, production of MIP-2 and malondialdehyde, activation of NF-κB and NRF, apoptotic epithelial cell death, and disruption of bronchial microstructure in mice. Mechanical stretch-augmented acute lung injury was also attenuated through pharmacological inhibition of NF-κB activity by SN50 and NRF expression by NRF short interfering RNA. Our data suggest that neutrophil elastase inhibitor attenuates high-tidal-volume mechanical ventilation-induced neutrophil influx, oxidative stress, and production of MIP-2, at least partly, through inhibition of NF-κB/NRF pathway. Understanding the protective effects of neutrophil elastase inhibitor associated with the reduction of MIP-2 allow clarification of the pathophysiological mechanisms regulating severe lung inflammation and development of possible therapeutic strategies involved in acute lung injury.
Collapse
Affiliation(s)
- Li-Fu Li
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Yi-Ting Lai
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan Graduate Institute of Clinical Medical Sciences and Department of Respiratory Care, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chih-Hao Chang
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan
| | - Meng-Chih Lin
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Yung-Yang Liu
- Chest Department, Taipei Veterans General Hospital, Taipei 112, Taiwan Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Kuo-Chin Kao
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Ying-Huang Tsai
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan Department of Respiratory Therapy, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| |
Collapse
|
867
|
Jiang S, Park DW, Tadie JM, Gregoire M, Deshane J, Pittet JF, Abraham E, Zmijewski JW. Human resistin promotes neutrophil proinflammatory activation and neutrophil extracellular trap formation and increases severity of acute lung injury. THE JOURNAL OF IMMUNOLOGY 2014; 192:4795-803. [PMID: 24719460 DOI: 10.4049/jimmunol.1302764] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although resistin was recently found to modulate insulin resistance in preclinical models of type II diabetes and obesity, recent studies also suggested that resistin has proinflammatory properties. We examined whether the human-specific variant of resistin affects neutrophil activation and the severity of LPS-induced acute lung injury. Because human and mouse resistin have distinct patterns of tissue distribution, experiments were performed using humanized resistin mice that exclusively express human resistin (hRTN(+/-)(/-)) but are deficient in mouse resistin. Enhanced production of TNF-α or MIP-2 was found in LPS-treated hRtn(+/-/-) neutrophils compared with control Rtn(-/-/-) neutrophils. Expression of human resistin inhibited the activation of AMP-activated protein kinase, a major sensor and regulator of cellular bioenergetics that also is implicated in inhibiting inflammatory activity of neutrophils and macrophages. In addition to the ability of resistin to sensitize neutrophils to LPS stimulation, human resistin enhanced neutrophil extracellular trap formation. In LPS-induced acute lung injury, humanized resistin mice demonstrated enhanced production of proinflammatory cytokines, more severe pulmonary edema, increased neutrophil extracellular trap formation, and elevated concentration of the alarmins HMGB1 and histone 3 in the lungs. Our results suggest that human resistin may play an important contributory role in enhancing TLR4-induced inflammatory responses, and it may be a target for future therapies aimed at reducing the severity of acute lung injury and other inflammatory situations in which neutrophils play a major role.
Collapse
Affiliation(s)
- Shaoning Jiang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294
| | | | | | | | | | | | | | | |
Collapse
|
868
|
Yeh CH, Yang JJ, Yang ML, Li YC, Kuan YH. Rutin decreases lipopolysaccharide-induced acute lung injury via inhibition of oxidative stress and the MAPK-NF-κB pathway. Free Radic Biol Med 2014; 69:249-57. [PMID: 24486341 DOI: 10.1016/j.freeradbiomed.2014.01.028] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 01/17/2014] [Accepted: 01/22/2014] [Indexed: 12/17/2022]
Abstract
Acute lung injury (ALI) is a serious disease with unacceptably high mortality and morbidity rates. Up to now, no effective therapeutic strategy for ALI has been established. Rutin, quercetin-3-rhamnosyl glucoside, expresses a wide range of biological activities and pharmacological effects, such as anti-inflammatory, antihypertensive, anticarcinogenic, vasoprotective, and cardioprotective activities. Pretreatment with rutin inhibited not only histopathological changes in lung tissues but also infiltration of polymorphonuclear granulocytes into bronchoalveolar lavage fluid in lipopolysaccharide (LPS)-induced ALI. In addition, LPS-induced inflammatory responses, including increased secretion of proinflammatory cytokines and lipid peroxidation, were inhibited by rutin in a concentration-dependent manner. Furthermore, rutin suppressed phosphorylation of NF-κB and MAPK and degradation of IκB, an NF-κB inhibitor. Decreased activities of antioxidative enzymes such as superoxide dismutase, catalase, glutathione peroxidase, and heme oxygenase-1 caused by LPS were reversed by rutin. At the same time, we found that ALI amelioration by chelation of extracellular metal ions with rutin is more efficacious than with deferoxamine. These results indicate that the protective mechanism of rutin is through inhibition of MAPK-NF-κB activation and upregulation of antioxidative enzymes.
Collapse
Affiliation(s)
- Chung-Hsin Yeh
- Department of Neurology, Show Chwan Memorial Hospital, Changhua, Taiwan; Department of Nursing, College of Medicine & Nursing, Hung Kuang University, Taichung, Taiwan
| | - Jiann-Jou Yang
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Ming-Ling Yang
- Department of Anatomy, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Ching Li
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Hsiang Kuan
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Pharmacy, Chung Shan Medical University Hospital, Taichung 402, Taiwan.
| |
Collapse
|
869
|
ZOU YINGGANG, DONG CHUNLING, YUAN MINGZHEN, GAO GUANGYUAN, WANG SIYI, LIU XIAODING, HAN HUIQIAO, LI BO. Instilled air promotes lipopolysaccharide-induced acute lung injury. Exp Ther Med 2014; 7:816-820. [PMID: 24660029 PMCID: PMC3961132 DOI: 10.3892/etm.2014.1523] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 01/23/2014] [Indexed: 01/11/2023] Open
Abstract
Optimization of intratracheal instillation is necessary to establish an ideal animal model of acute lung injury (ALI) in order to further reveal the cellular and molecular pathogenesis of ALI. It is possible that instilling air from a prefilled syringe may promote the delivery of reagents into the alveolar spaces, resulting in different pulmonary responses. In the present study, the influence of instilling air by trans-tracheal intratracheal instillation in a lipopolysaccharide (LPS)-induced mouse model of ALI was investigated. The bronchoalveolar lavage (BAL) fluid biochemical index, BAL fluid differential cell counts, lung wet/dry weight ratio, lung histology and BAL fluid interleukin-8 (IL-8) levels were assessed 24 h subsequent to intratracheal instillation. Instilled air promoted LPS-induced ALI, as indicated by the severity of acute pulmonary inflammation and increased IL-8 release. In conclusion, this study indicates that instilled air may be used to improve the intratracheal instillation procedure and to establish a more reliable animal model of ALI.
Collapse
Affiliation(s)
- YINGGANG ZOU
- Department of Human Anatomy, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
- Department of Obstetrics and Gynecology, Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - CHUNLING DONG
- Department of Respiratory Medicine, Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - MINGZHEN YUAN
- Department of Human Anatomy, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - GUANGYUAN GAO
- Department of Human Anatomy, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - SIYI WANG
- Department of Human Anatomy, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - XIAODING LIU
- Department of Human Anatomy, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - HUIQIAO HAN
- Department of Human Anatomy, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - BO LI
- Department of Human Anatomy, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
870
|
Interleukin 17A promotes pneumococcal clearance by recruiting neutrophils and inducing apoptosis through a p38 mitogen-activated protein kinase-dependent mechanism in acute otitis media. Infect Immun 2014; 82:2368-77. [PMID: 24664502 DOI: 10.1128/iai.00006-14] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Streptococcus pneumoniae is a Gram-positive and human-restricted pathogen colonizing the nasopharynx with an absence of clinical symptoms as well as a major pathogen causing otitis media (OM), one of the most common childhood infections. Upon bacterial infection, neutrophils are rapidly activated and recruited to the infected site, acting as the frontline defender against emerging microbial pathogens via different ways. Evidence shows that interleukin 17A (IL-17A), a neutrophil-inducing factor, plays important roles in the immune responses in several diseases. However, its function in response to S. pneumoniae OM remains unclear. In this study, the function of IL-17A in response to S. pneumoniae OM was examined using an in vivo model. We developed a model of acute OM (AOM) in C57BL/6 mice and found that neutrophils were the dominant immune cells that infiltrated to the middle ear cavity (MEC) and contributed to bacterial clearance. Using IL-17A knockout (KO) mice, we found that IL-17A boosted neutrophil recruitment to the MEC and afterwards induced apoptosis, which was identified to be conducive to bacterial clearance. In addition, our observation suggested that the p38 mitogen-activated protein kinase (MAPK) signaling pathway was involved in the recruitment and apoptosis of neutrophils mediated by IL-17A. These data support the conclusion that IL-17A contributes to the host immune response against S. pneumoniae by promoting neutrophil recruitment and apoptosis through the p38 MAPK signaling pathway.
Collapse
|
871
|
Dynamics of pulmonary endothelial barrier function in acute inflammation: mechanisms and therapeutic perspectives. Cell Tissue Res 2014; 355:657-73. [PMID: 24599335 PMCID: PMC7102256 DOI: 10.1007/s00441-014-1821-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/16/2014] [Indexed: 12/11/2022]
Abstract
The lungs provide a large inner surface to guarantee respiration. In lung alveoli, a delicate membrane formed by endo- and epithelial cells with their fused basal lamina ensures rapid and effective gas exchange between alveolar and vascular compartments while concurrently forming a robust barrier against inhaled particles and microbes. However, upon infectious or sterile inflammatory stimulation, tightly regulated endothelial barrier leakiness is required for leukocyte transmigration. Further, endothelial barrier disruption may result in uncontrolled extravasation of protein-rich fluids. This brief review summarizes some important mechanisms of pulmonary endothelial barrier regulation and disruption, focusing on the role of specific cell populations, coagulation and complement cascades and mediators including angiopoietins, specific sphingolipids, adrenomedullin and reactive oxygen and nitrogen species for the regulation of pulmonary endothelial barrier function. Further, current therapeutic perspectives against development of lung injury are discussed.
Collapse
|
872
|
Wagner JG, Birmingham NP, Jackson-Humbles D, Jiang Q, Harkema JR, Peden DB. Supplementation with γ-tocopherol attenuates endotoxin-induced airway neutrophil and mucous cell responses in rats. Free Radic Biol Med 2014; 68:101-9. [PMID: 24333275 PMCID: PMC3961823 DOI: 10.1016/j.freeradbiomed.2013.11.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 11/13/2013] [Accepted: 11/25/2013] [Indexed: 12/31/2022]
Abstract
Neutrophil-mediated tissue injury is a shared pathogenesis of both chronic pulmonary diseases and acute responses to pathogens, allergens, and airborne pollutants. Interventions to minimize toxic effects of neutrophil-derived oxidants and proteases are usually limited to corticosteroids, which can have adverse side effects. We used a rodent model of endotoxin-induced lung injury to test the hypothesis that the dietary supplement γ-tocopherol (γT), a natural form of vitamin E with antioxidant and novel anti-inflammatory properties, will protect from adverse nasal and pulmonary inflammatory responses induced by endotoxin (lipopolysaccharide; LPS). Male Fisher F344 rats were intranasally (i.n.) instilled with LPS for 2 consecutive days. Beginning 2 days before i.n. LPS, the rats were gavaged daily with 30mg/kg γT. Twenty-four hours after the last i.n. LPS, bronchoalveolar lavage fluid (BALF) was collected, and pulmonary and nasal tissues were analyzed for gene expression and morphometric analyses of neutrophils and intraepithelial mucosubstances (IM). LPS caused increased BALF total cells (70% increase), neutrophils (300%), protein (35%), PGE2 (500%), and secreted mucins (75%). Robust increases in neutrophils and IM were detected in conducting airways. Pulmonary expression of MUC5AC, MIP-2, CINC-1, and MCP-1 was elevated three- to eightfold by LPS. Treatment with γT inhibited LPS-induced increases in BALF total cells, neutrophils, protein, PGE2, and secreted mucins, as well as IM and tissue neutrophil influx. Furthermore γT induced the expression of the regulatory cytokines IL-10 and IFN-γ while decreasing MUC5AC, MIP-2, CINC-1, and MCP-1. These data demonstrate novel therapeutic effects of the dietary vitamin E γT promoting anti-inflammatory pathways to protect from neutrophil-mediated lung injury.
Collapse
Affiliation(s)
- James G Wagner
- Center for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA; Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, USA.
| | - Neil P Birmingham
- Center for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Daven Jackson-Humbles
- Center for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA; Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, USA
| | - Qing Jiang
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Jack R Harkema
- Center for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA; Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI 48824, USA
| | - David B Peden
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
873
|
Zosky GR, Iosifidis T, Perks K, Ditcham WGF, Devadason SG, Siah WS, Devine B, Maley F, Cook A. The concentration of iron in real-world geogenic PM₁₀ is associated with increased inflammation and deficits in lung function in mice. PLoS One 2014; 9:e90609. [PMID: 24587402 PMCID: PMC3938778 DOI: 10.1371/journal.pone.0090609] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/02/2014] [Indexed: 11/19/2022] Open
Abstract
Background There are many communities around the world that are exposed to high levels of particulate matter <10 µm (PM10) of geogenic (earth derived) origin. Mineral dusts in the occupational setting are associated with poor lung health, however very little is known about the impact of heterogeneous community derived particles. We have preliminary evidence to suggest that the concentration of iron (Fe) may be associated with the lung inflammatory response to geogenic PM10. We aimed to determine which physico-chemical characteristics of community sampled geogenic PM10 are associated with adverse lung responses. Methods We collected geogenic PM10 from four towns in the arid regions of Western Australia. Adult female BALB/c mice were exposed to 100 µg of particles and assessed for inflammatory and lung function responses 6 hours, 24 hours and 7 days post-exposure. We assessed the physico-chemical characteristics of the particles and correlated these with lung outcomes in the mice using principal components analysis and multivariate linear regression. Results Geogenic particles induced an acute inflammatory response that peaked 6 hours post-exposure and a deficit in lung mechanics 7 days post-exposure. This deficit in lung mechanics was positively associated with the concentration of Fe and particle size variability and inversely associated with the concentration of Si. Conclusions The lung response to geogenic PM10 is complex and highly dependent on the physico-chemical characteristics of the particles. In particular, the concentration of Fe in the particles may be a key indicator of the potential population health consequences for inhaling geogenic PM10.
Collapse
Affiliation(s)
- Graeme R. Zosky
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- * E-mail:
| | - Thomas Iosifidis
- Telethon Institute for Child Health Research, Subiaco, Western Australia, Australia
| | - Kara Perks
- Telethon Institute for Child Health Research, Subiaco, Western Australia, Australia
| | - Will G. F. Ditcham
- School of Paediatrics and Child Health, The University of Western Australia, Subiaco, Western Australia, Australia
| | - Sunalene G. Devadason
- School of Paediatrics and Child Health, The University of Western Australia, Subiaco, Western Australia, Australia
| | - W. Shan Siah
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- School of Population Health, The University of Western Australia, Crawley, Western Australia, Australia
| | - Brian Devine
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- School of Population Health, The University of Western Australia, Crawley, Western Australia, Australia
| | - Fiona Maley
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- School of Population Health, The University of Western Australia, Crawley, Western Australia, Australia
| | - Angus Cook
- Cooperative Research Centre for Asthma and Airways, Sydney, New South Wales, Australia
- School of Population Health, The University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
874
|
Shalaby SM, El-Shal AS, Abd-Allah SH, Selim AO, Selim SA, Gouda ZA, Abd El Motteleb DM, Zanfaly HE, El-Assar HM, Abdelazim S. Mesenchymal stromal cell injection protects against oxidative stress in Escherichia coli-induced acute lung injury in mice. Cytotherapy 2014; 16:764-75. [PMID: 24525173 DOI: 10.1016/j.jcyt.2013.12.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/07/2013] [Accepted: 12/12/2013] [Indexed: 01/08/2023]
Abstract
BACKGROUND AIMS Stem cells may be a promising therapy for acute respiratory distress syndrome. Recent in vivo and in vitro studies suggested that the mesenchymal stromal cells (MSCs) have anti-oxidative stress properties. We hypothesized that intravenous injection of bone marrow-derived mesenchymal stem cells (MSCs) could attenuate Escherichia coli-induced acute lung injury (ALI) in mice by controlling the oxidative stress status. METHODS Eighty mice were randomly divided into four groups: group 1 (control group) received 25 μL of saline as a vehicle; group 2 contained E coli-induced ALI mice; group 3 included mice that received MSCs before induction of ALI; group 4 included mice that received MSCs after induction of ALI. Lung samples were isolated and assayed for oxidative stress variables and histopathologic analysis. Total anti-oxidant capacity was measured in broncho-alveolar lavage. RESULTS Pre- and post-injury MSC injection increased survival, reduced pulmonary edema and attenuated lung injuries in ALI mice. Histologically, MSCs exhibited a considerable degree of preservation of the pulmonary alveolar architecture. An increase of anti-oxidant enzyme activities and a decrease of myeloperoxidase activity and malondialdehyde levels in the MSC recipient groups versus the ALI group were found. Furthermore, the total anti-oxidant capacity and reduced glutathione levels were significantly increased in MSCs recipient groups versus the ALI group. Weak +ve inducible nitric oxide synthase immuno-expression in groups that received MSCs was detected. Pre-injury MSC injection showed better effects than did post-injury MSC injection. CONCLUSIONS Systemic bone marrow-derived MSC injection was effective in modulating the oxidative stress status in E coli-induced acute lung injury in mice.
Collapse
Affiliation(s)
- Sally M Shalaby
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Amal S El-Shal
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Somia H Abd-Allah
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Assmaa O Selim
- Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sally A Selim
- Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Zienab A Gouda
- Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Hala E Zanfaly
- Anesthesia and Intensive Care Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Heba M El-Assar
- Anesthesia and Intensive Care Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Shymaa Abdelazim
- Microbiology and Immunology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
875
|
Si-Tahar M, Blanc F, Furio L, Chopy D, Balloy V, Lafon M, Chignard M, Fiette L, Langa F, Charneau P, Pothlichet J. Protective role of LGP2 in influenza virus pathogenesis. J Infect Dis 2014; 210:214-23. [PMID: 24493823 DOI: 10.1093/infdis/jiu076] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Influenza A virus triggers a contagious respiratory disease that can cause considerable morbidity and mortality. Using an in vitro approach, we previously demonstrated that the pattern recognition receptor retinoic acid-inducible gene I (RIG-I) plays a key role in influenza A virus-mediated immune response. However, the importance of RIG-I signaling in vivo has not been thoroughly examined, because of the lack of an appropriate mouse models. To circumvent this issue, we generated a new transgenic mouse overexpressing LGP2 (hereafter, "LGP2 TG mice"), a major regulator of the RIG-I signaling pathway. The time course of several parameters was compared in infected wild-type and LGP2 TG mice. We found that LGP2 TG mice displayed significantly reduced inflammatory mediators and a lower leukocyte infiltration into the bronchoalveolar airspace. More importantly, LGP2 TG mice had a significant survival advantage. Hence, our in vivo study reveals that LGP2 is a major downregulator of the influenza A virus-triggered detrimental inflammatory response.
Collapse
Affiliation(s)
- Mustapha Si-Tahar
- Institut Pasteur, Unité de Défense Innée et Inflammation Inserm, U874
| | - Fany Blanc
- Institut Pasteur, Unité de Défense Innée et Inflammation Inserm, U874
| | - Laetitia Furio
- Institut Pasteur, Unité de Défense Innée et Inflammation Inserm, U874
| | - Damien Chopy
- Institut Pasteur, Unité de Neuroimmunologie Virale CNRS UMR3569
| | - Viviane Balloy
- Institut Pasteur, Unité de Défense Innée et Inflammation Inserm, U874
| | - Monique Lafon
- Institut Pasteur, Unité de Neuroimmunologie Virale CNRS UMR3569
| | - Michel Chignard
- Institut Pasteur, Unité de Défense Innée et Inflammation Inserm, U874
| | | | - Francina Langa
- Institut Pasteur, Plate-Forme Technologique Centre d'Ingenierie Genetique Murine
| | - Pierre Charneau
- Laboratoire de Virologie moléculaire et vectorologie, Institut Pasteur, Paris, France
| | - Julien Pothlichet
- Institut Pasteur, Unité de Défense Innée et Inflammation Inserm, U874
| |
Collapse
|
876
|
Inhibition of inflammatory injure by polysaccharides from Bupleurum chinense through antagonizing P-selectin. Carbohydr Polym 2014; 105:20-5. [PMID: 24708947 DOI: 10.1016/j.carbpol.2014.01.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 01/10/2014] [Accepted: 01/12/2014] [Indexed: 12/21/2022]
Abstract
P-selectin-mediated adhesion between endothelium and neutrophils is a crucial process leading to acute inflammatory injure. Thus, P-selectin has been considered as promising target for therapeutics of acute inflammatory-related diseases. In the present study, the water-soluble polysaccharides (BCPs) were isolated from Bupleurum chinense, and we evaluated their therapeutical effects on acute inflammatory injure and antagonistic function against P-selectin-mediated neutrophil adhesion. Our results showed that BCPs significantly impaired the leukocyte infiltration and relieve lung injury in LPS-induced acute pneumonia model. BCPs significantly blocked the binding of P-selectin to neutrophils and inhibited P-selectin-mediated neutrophils rolling along CHO-P cell monolayer. The result from in vitro protein binding assay showed a direct evidence indicating that BCPs-treatment significantly eliminated the interaction between rhP-Fc and its physiological ligand PSGL-1 at protein level. Together, these results provide a novel therapeutical strategy for amelioration of inflammation-related disease processes by polysaccharides from B. chinense.
Collapse
|
877
|
Inhibition of extracellular HMGB1 attenuates hyperoxia-induced inflammatory acute lung injury. Redox Biol 2014; 2:314-22. [PMID: 24563849 PMCID: PMC3926109 DOI: 10.1016/j.redox.2014.01.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 01/14/2014] [Accepted: 01/14/2014] [Indexed: 01/07/2023] Open
Abstract
Prolonged exposure to hyperoxia results in acute lung injury (ALI), accompanied by a significant elevation in the levels of proinflammatory cytokines and leukocyte infiltration in the lungs. However, the mechanisms underlying hyperoxia-induced proinflammatory ALI remain to be elucidated. In this study, we investigated the role of the proinflammatory cytokine high mobility group box protein 1 (HMGB1) in hyperoxic inflammatory lung injury, using an adult mouse model. The exposure of C57BL/6 mice to ≥99% O2 (hyperoxia) significantly increased the accumulation of HMGB1 in the bronchoalveolar lavage fluids (BALF) prior to the onset of severe inflammatory lung injury. In the airways of hyperoxic mice, HMGB1 was hyperacetylated and existed in various redox forms. Intratracheal administration of recombinant HMGB1 (rHMGB1) caused a significant increase in leukocyte infiltration into the lungs compared to animal treated with a non-specific peptide. Neutralizing anti-HMGB1 antibodies, administrated before hyperoxia significantly attenuated pulmonary edema and inflammatory responses, as indicated by decreased total protein content, wet/dry weight ratio, and numbers of leukocytes in the airways. This protection was also observed when HMGB1 inhibitors were administered after the onset of the hyperoxic exposure. The aliphatic antioxidant, ethyl pyruvate (EP), inhibited HMGB1 secretion from hyperoxic macrophages and attenuated hyperoxic lung injury. Overall, our data suggest that HMGB1 plays a critical role in mediating hyperoxic ALI through the recruitment of leukocytes into the lungs. If these results can be translated to humans, they suggest that HMGB1 inhibitors provide treatment regimens for oxidative inflammatory lung injury in patients receiving hyperoxia through mechanical ventilation. Exposure to hyperoxia results in accumulation of high levels of airway HMGB1 that precede inflammatory acute lung injury (ALI). Airway HMGB1 is critical in mediating hyperoxia-induced inflammatory ALI via recruiting leukocytes including neutrophils. Extracellular HMGB1-accumulated upon prolonged exposure to hyperoxia is hyperacetylated, existing in different redox states. Small molecule EP, administrated even after the onset of hyperoxic exposure, can mitigate hyperoxia-induced inflammatory ALI by inhibiting HMGB1 release into the extracellular milieu.
Collapse
Key Words
- ALI, acute lung injury
- BALF, bronchoalveolar lavage fluids
- EP, ethyl pyruvate
- GST, gluthatione-s-transferase
- HMGB1
- HMGB1, high mobility group box protein 1
- Hyperacetylation
- Hyperoxia
- MV, mechanical ventilation
- Macrophage
- NLS, nuclear localization signal
- PMNs, polymorphonuclear neutrophils
- RA, room air
- ROS, reactive oxygen species
- Redox state
- rHMGB1, recombinant HMGB1
Collapse
|
878
|
Gonzales JN, Gorshkov B, Varn MN, Zemskova MA, Zemskov EA, Sridhar S, Lucas R, Verin AD. Protective effect of adenosine receptors against lipopolysaccharide-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 2014; 306:L497-507. [PMID: 24414256 DOI: 10.1152/ajplung.00086.2013] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Acute lung injury and acute respiratory distress syndrome (ALI/ARDS) affect 200,000 people a year in the USA. Pulmonary vascular and specifically endothelial cell (EC) barrier compromise is a hallmark of these diseases. We have recently shown that extracellular adenosine enhances human pulmonary (EC) barrier via activation of adenosine receptors (ARs) in cell cultures. On the basis of these data, we hypothesized that activation of ARs might exert barrier-protective effects in a model of ALI/ARDS in mice. To test this hypothesis, we examined the effects of pre- and posttreatment of adenosine and 5'-N-ethylcarboxamidoadenosine (NECA), a nonselective stable AR agonist, on LPS-induced lung injury. Mice were given vehicle or LPS intratracheally followed by adenosine, NECA, or vehicle instilled via the internal jugular vein. Postexperiment cell counts, Evans Blue Dye albumin (EBDA) extravasation, levels of proteins, and inflammatory cytokines were analyzed. Harvested lungs were used for histology and myeloperoxidase studies. Mice challenged with LPS alone demonstrated an inflammatory response typical of ALI. Cell counts, EBDA extravasation, as well as levels of proteins and inflammatory cytokines were decreased in adenosine-treated mice. Histology displayed reduced infiltration of neutrophils. NECA had a similar effect on LPS-induced vascular barrier compromise. Importantly, posttreatment with adenosine or NECA recovers lung vascular barrier and reduces inflammation induced by LPS challenge. Furthermore, adenosine significantly attenuated protein degradation of A2A and A3 receptors induced by LPS. Collectively, our results demonstrate that activation of ARs protects and restores vascular barrier functions and reduces inflammation in LPS-induced ALI.
Collapse
Affiliation(s)
- Joyce N Gonzales
- Assistant Prof. of Medicine, Div. of Pulmonary and Critical Care Medicine, Georgia Regents Univ., Rm. BBR-5513, 1120 15th St., Augusta, GA 30912.
| | | | | | | | | | | | | | | |
Collapse
|
879
|
Han D, Hu Y, Li L, Tian H, Chen Z, Wang L, Ma H, Yang H, Teng K. Highly pathogenic porcine reproductive and respiratory syndrome virus infection results in acute lung injury of the infected pigs. Vet Microbiol 2014; 169:135-46. [PMID: 24472226 PMCID: PMC7127595 DOI: 10.1016/j.vetmic.2013.12.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 12/19/2013] [Accepted: 12/23/2013] [Indexed: 01/11/2023]
Abstract
Highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) was firstly characterized in 2006 in China. The virus has caused great economic loss to the Chinese swine production during the past years. Herein, we experimentally infected SPF pigs using two strains of PRRSV with different pathogenicity and observed the lung pathological changes looking for new sights on the possible pathogenesis associated with the virulence of HP-PRRSV. The results indicated that the HP-PRRSV-infected pigs died and exhibited severe pathological changes of lungs featuring increased neutrophils, mast cells and mononuclear macrophages, compared with the pigs inoculated with low pathogenic (LP-) PRRSV. Furthermore, the pigs infected with HP-PRRSV showed the higher levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, interleukin (IL)-8 and histamine, leukotriene B4 (LTB4), platelet activation factor (PAF) in sera than those inoculated with LP-PRRSV. Additionally, the fibrosis of lung was observed in the HP-PRRSV-infected pigs. At present, our findings suggest that the aberrant immune responses triggered by HP-PRRSV infection are closely related to acute lung injury (ALI), and especially the pathological changes in lung vascular system are of particular significance. These associated pathological changes of lung are in part responsible for the additional morbidity and mortality observed in HP-PRRSV infection.
Collapse
Affiliation(s)
- Deping Han
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Yanxin Hu
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Limin Li
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Haiyan Tian
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Zhi Chen
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Lin Wang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Haiyan Ma
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Hanchun Yang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China.
| | - Kedao Teng
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and Key state Laboratory of Agrobiotechnology, China Agricultural University, No.2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China.
| |
Collapse
|
880
|
Kusek ME, Pazos MA, Pirzai W, Hurley BP. In vitro coculture assay to assess pathogen induced neutrophil trans-epithelial migration. J Vis Exp 2014:e50823. [PMID: 24430378 DOI: 10.3791/50823] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Mucosal surfaces serve as protective barriers against pathogenic organisms. Innate immune responses are activated upon sensing pathogen leading to the infiltration of tissues with migrating inflammatory cells, primarily neutrophils. This process has the potential to be destructive to tissues if excessive or held in an unresolved state. Cocultured in vitro models can be utilized to study the unique molecular mechanisms involved in pathogen induced neutrophil trans-epithelial migration. This type of model provides versatility in experimental design with opportunity for controlled manipulation of the pathogen, epithelial barrier, or neutrophil. Pathogenic infection of the apical surface of polarized epithelial monolayers grown on permeable transwell filters instigates physiologically relevant basolateral to apical trans-epithelial migration of neutrophils applied to the basolateral surface. The in vitro model described herein demonstrates the multiple steps necessary for demonstrating neutrophil migration across a polarized lung epithelial monolayer that has been infected with pathogenic P. aeruginosa (PAO1). Seeding and culturing of permeable transwells with human derived lung epithelial cells is described, along with isolation of neutrophils from whole human blood and culturing of PAO1 and nonpathogenic K12 E. coli (MC1000). The emigrational process and quantitative analysis of successfully migrated neutrophils that have been mobilized in response to pathogenic infection is shown with representative data, including positive and negative controls. This in vitro model system can be manipulated and applied to other mucosal surfaces. Inflammatory responses that involve excessive neutrophil infiltration can be destructive to host tissues and can occur in the absence of pathogenic infections. A better understanding of the molecular mechanisms that promote neutrophil trans-epithelial migration through experimental manipulation of the in vitro coculture assay system described herein has significant potential to identify novel therapeutic targets for a range of mucosal infectious as well as inflammatory diseases.
Collapse
Affiliation(s)
- Mark E Kusek
- Department of Pediatrics, Harvard Medical School
| | | | | | | |
Collapse
|
881
|
Bai J, Tang L, Lomas-Neira J, Chen Y, McLeish KR, Uriarte SM, Chung CS, Ayala A. TAT-SNAP-23 treatment inhibits the priming of neutrophil functions contributing to shock and/or sepsis-induced extra-pulmonary acute lung injury. Innate Immun 2014; 21:42-54. [PMID: 24391146 DOI: 10.1177/1753425913516524] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Respiratory burst function of neutrophils is thought to play a pivotal role in the development of pathologies such as indirect (extra-pulmonary) acute lung injury (iALI), as well as sepsis. The current study was conducted to determine the effect of an HIV transactivator of transcription (TAT)-fusion protein containing a soluble N-ethylmaleimide-sensitive factor attachment protein receptor domain from synaptosome-associated protein-23 (SNAP-23) on the shock/sepsis- and sepsis-enhanced neutrophil burst capacity using the clinical relevant two-hit iALI mouse model and the classical cecal ligation and puncture (CLP) septic model. TAT-SNAP-23 significantly decreased the blood neutrophil respiratory burst in vitro, and also in vivo in CLP and hemorrhaged mice. We found that the neutrophil influx to the lung tissue, as measured by myeloperoxidase levels and neutrophil-specific esterase(+) cells, was also decreased in the TAT-SNAP-23-treated group. Consistent with this, treatment of TAT-SNAP-23 significantly reduced the disruption of lung tissue architecture and protein concentration of bronchoalveolar lavage fluid in iALI mice compared with vehicle-treated iALI mice. In addition, although TAT-SNAP-23 did not alter the extent of local cytokine/chemokine expression, the in vitro migration capacity of neutrophils was blunted from septic and hemorrhagic mice. These data support our hypothesis that TAT-SNAP-23 reduces neutrophil dysfunction in iALI and sepsis by inhibiting neutrophil respiratory burst.
Collapse
Affiliation(s)
- Jianwen Bai
- Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Tong Ji University, Shanghai, PR China
| | - Lunxian Tang
- Department of Emergency Medicine and Critical Care, Shanghai East Hospital, Tong Ji University, Shanghai, PR China
| | - Joanne Lomas-Neira
- Department of Surgery, Division of Surgical Research, the Alpert School of Medicine at Brown University/Rhode Island Hospital, Providence, RI, USA
| | - Yaping Chen
- Department of Surgery, Division of Surgical Research, the Alpert School of Medicine at Brown University/Rhode Island Hospital, Providence, RI, USA
| | - Kenneth R McLeish
- Department of Medicine, University of Louisville, Louisville, KY, USA Robley Rx VAMC, Louisville, KY, USA
| | - Silvia M Uriarte
- Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Chun-Shiang Chung
- Department of Surgery, Division of Surgical Research, the Alpert School of Medicine at Brown University/Rhode Island Hospital, Providence, RI, USA
| | - Alfred Ayala
- Department of Surgery, Division of Surgical Research, the Alpert School of Medicine at Brown University/Rhode Island Hospital, Providence, RI, USA
| |
Collapse
|
882
|
Tamai K, Tachikawa R, Tomii K, Nagata K, Otsuka K, Nakagawa A, Otsuka K, Matsumoto T, Monden K, Takeshita J, Tanaka K, Kawamura T, Otoshi T, Fujimoto D. Prognostic value of bronchoalveolar lavage in patients with non-HIV pneumocystis pneumonia. Intern Med 2014; 53:1113-7. [PMID: 24881733 DOI: 10.2169/internalmedicine.53.0520] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Non-HIV patients with pneumocystis pneumonia (PCP) have a poor prognosis. We aimed to evaluate the prognostic factors for in-hospital mortality in terms of the clinical findings, including the results of bronchoalveolar lavage fluid (BALF)-analyses, in non-HIV PCP patients. METHODS We retrospectively reviewed non-HIV PCP patients diagnosed using bronchoalveolar lavage between April 2006 and July 2012. For patients with a poor respiratory status, noninvasive positive pressure ventilation (NPPV) was used during the bronchoalveolar lavage (BAL) procedure. Data regarding demographics, laboratory findings and the prognosis were evaluated. RESULTS A total of 29 non-HIV PCP patients were analyzed. NPPV was carried out safely and successfully in 12 patients during the BAL procedure. Twelve patients (41%) died. The multivariate logistic regression analysis identified only BALF neutrophilia to be a significant prognostic factor determining in-hospital mortality. The log-rank test showed that the patients with BALF neutrophilia (≥ 31%) had a significantly lower survival rate than the other patients (p=0.001). CONCLUSION Only BALF neutrophilia was found to be a significant predictor of survival in patients with non-HIV PCP. Our data also emphasize the significance of performing BAL in such patients, as it provides both diagnostic and prognostic information.
Collapse
Affiliation(s)
- Koji Tamai
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
883
|
[Ventilator-induced immune dysfunction]. MEDECINE INTENSIVE REANIMATION 2014; 23:9-16. [PMID: 32288738 PMCID: PMC7117828 DOI: 10.1007/s13546-014-0846-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 12/26/2013] [Indexed: 10/30/2022]
Abstract
Although mechanical ventilation is an essential support in patients admitted to the intensive care unit, clinical and experimental studies have shown that it could be harmful and could induce lung injury. Pulmonary and immune cells can convert mechanical stimuli into biological signals that will lead to inflammation. This sterile inflammation both locally and systemically will cause immunosuppression.
Collapse
|
884
|
Abstract
The acute respiratory distress syndrome (ARDS) is a major public health problem and a leading source of morbidity in intensive care units. Lung tissue in patients with ARDS is characterized by inflammation, with exuberant neutrophil infiltration, activation, and degranulation that is thought to initiate tissue injury through the release of proteases and oxygen radicals. Treatment of ARDS is supportive primarily because the underlying pathophysiology is poorly understood. This gap in knowledge must be addressed to identify urgently needed therapies. Recent research efforts in anti-inflammatory drug development have focused on identifying common control points in multiple signaling pathways. The protein kinase C (PKC) serine-threonine kinases are master regulators of proinflammatory signaling hubs, making them attractive therapeutic targets. Pharmacological inhibition of broad-spectrum PKC activity and, more importantly, of specific PKC isoforms (as well as deletion of PKCs in mice) exerts protective effects in various experimental models of lung injury. Furthermore, PKC isoforms have been implicated in inflammatory processes that may be involved in the pathophysiologic changes that result in ARDS, including activation of innate immune and endothelial cells, neutrophil trafficking to the lung, regulation of alveolar epithelial barrier functions, and control of neutrophil proinflammatory and prosurvival signaling. This review focuses on the mechanistic involvement of PKC isoforms in the pathogenesis of ARDS and highlights the potential of developing new therapeutic paradigms based on the selective inhibition (or activation) of specific PKC isoforms.
Collapse
|
885
|
Li W, Wu X, Yan F, Liu J, Tang Y, Ma K, Li S. Effects of pulmonary artery perfusion with urinary trypsin inhibitor as a lung protective strategy under hypothermic low-flow cardiopulmonary bypass in an infant piglet model. Perfusion 2013; 29:434-42. [PMID: 24335190 DOI: 10.1177/0267659113517286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE This study aimed to evaluate the effects of pulmonary artery perfusion with a urinary trypsin inhibitor (UTI) as a lung protective strategy in order to provide an experimental basis for immature lung clinical protective strategies on deep hypothermia with low-flow (DHLF) cardiopulmonary bypass (CPB)-induced pulmonary injury in an infant piglet model. METHODS The piglets (n=15), aged 18.7±0.3 days, weight 4.48±0.21kg, were randomly divided into 3 groups, with 5 piglets in each group: the control group, the pulmonary artery perfusion without UTI group (Group P) and the pulmonary artery perfusion with UTI group (Group U). The levels of the cytokines tumour necrosis factor-α, myeloperoxidase, malondialdehyde and interleukin-10 (TNF-α, MPO, MDA and IL-10) in pulmonary venous serum and lung tissue and the activity of NF-kappa B in lung tissue were measured by enzyme-linked immunosorbent assay (ELISA) and electrophoresis mobility shift assay (EMSA), respectively. RESULTS After DHLF-CPB, all of the piglets demonstrated a state of lung injury as a deterioration of lung function indices, lung injury scores, pulmonary ultrastructure changes, expression of TNF-α, MPO, MDA and IL-10 and the activities of nuclear factor-kappa B (NF-κB), while pulmonary artery perfusion with UTI significantly ameliorated lung function and histopathological changes, with greatly decreased serum levels of TNF-α and MPO compared to the other two groups. Also, we found an increase in the level of IL-10 in Group U lungs compared with that in Group P lungs, which correlated with a strong inhibition in the activity of NF-κB. CONCLUSION Pulmonary artery perfusion with UTI ameliorated the DHLF-induced immature pulmonary injury in the lungs via a reduction of pro-inflammatory cytokine expression and up-regulated levels of IL-10 by inhibiting the activity of NF-κB.
Collapse
Affiliation(s)
- W Li
- Center of Pediatric Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - X Wu
- Center of Pediatric Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - F Yan
- Center of Pediatric Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - J Liu
- Center of Pediatric Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - Y Tang
- Center of Animals for Experiment, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - K Ma
- Center of Pediatric Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| | - S Li
- Center of Pediatric Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
886
|
Williams AE, Chambers RC. The mercurial nature of neutrophils: still an enigma in ARDS? Am J Physiol Lung Cell Mol Physiol 2013; 306:L217-30. [PMID: 24318116 DOI: 10.1152/ajplung.00311.2013] [Citation(s) in RCA: 287] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The acute respiratory distress syndrome (ARDS) is a life-threatening lung condition resulting from direct and indirect insults to the lung. It is characterized by disruption of the endothelial-epithelial barrier, alveolar damage, pulmonary edema, and respiratory failure. A key feature of ARDS is the accumulation of neutrophils in the lung microvasculature, interstitium, and alveolar space. Despite a clear association between neutrophil influx into the lung and disease severity, there is some debate as to whether neutrophils directly contribute to disease pathogenesis. The primary function of neutrophils is to provide immediate host defense against pathogenic microorganisms. Neutrophils release numerous antimicrobial factors such as reactive oxygen species, proteinases, and neutrophil extracellular traps. However, these factors are also toxic to host cells and can result in bystander tissue damage. The excessive accumulation of neutrophils in ARDS may therefore contribute to disease progression. Central to neutrophil recruitment is the release of chemokines, including the archetypal neutrophil chemoattractant IL-8, from resident pulmonary cells. However, the chemokine network in the inflamed lung is complex and may involve several other chemokines, including CXCL10, CCL2, and CCL7. This review will therefore focus on the experimental and clinical evidence supporting neutrophils as key players in ARDS and the chemokines involved in recruiting them into the lung.
Collapse
Affiliation(s)
- Andrew E Williams
- Centre for Inflammation and Tissue Repair, Univ. College London, Rayne Institute, 5 Univ. St., London WC1E 6JF, UK.
| | | |
Collapse
|
887
|
|
888
|
Neto-Neves EM, Sousa-Santos O, Ferraz KC, Rizzi E, Ceron CS, Romano MMD, Gali LG, Maciel BC, Schulz R, Gerlach RF, Tanus-Santos JE. Matrix metalloproteinase inhibition attenuates right ventricular dysfunction and improves responses to dobutamine during acute pulmonary thromboembolism. J Cell Mol Med 2013; 17:1588-97. [PMID: 24199964 PMCID: PMC3914650 DOI: 10.1111/jcmm.12163] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 09/16/2013] [Indexed: 02/06/2023] Open
Abstract
Activated matrix metalloproteinases (MMPs) cause cardiomyocyte injury during acute pulmonary thromboembolism (APT). However, the functional consequences of this alteration are not known. We examined whether doxycycline (a MMP inhibitor) improves right ventricle function and the cardiac responses to dobutamine during APT. APT was induced with autologous blood clots (350 mg/kg) in anaesthetized male lambs pre-treated with doxycycline (Doxy, 10 mg/kg/day, intravenously) or saline. Non-embolized control lambs received doxycycline pre-treatment or saline. The responses to intravenous dobutamine (Dob, 1, 5, 10 μg/kg/min.) or saline infusions at 30 and 120 min. after APT induction were evaluated by echocardiography. APT increased mean pulmonary artery pressure and pulmonary vascular resistance index by ∼185%. Doxycycline partially prevented APT-induced pulmonary hypertension (P < 0.05). RV diameter increased in the APT group (from 10.7 ± 0.8 to 18.3 ± 1.6 mm, P < 0.05), but not in the Doxy+APT group (from 13.3 ± 0.9 to 14.4 ± 1.0 mm, P > 0.05). RV dysfunction on stress echocardiography was observed in embolized lambs (APT+Dob group) but not in embolized animals pre-treated with doxycycline (Doxy+APT+Dob). APT increased MMP-9 activity, oxidative stress and gelatinolytic activity in the RV. Although doxycycline had no effects on RV MMP-9 activity, it prevented the increases in RV oxidative stress and gelatinolytic activity (P < 0.05). APT increased serum cardiac troponin I concentrations (P < 0.05), doxycycline partially prevented this alteration (P < 0.05). We found evidence to support that doxycycline prevents RV dysfunction and improves the cardiac responses to dobutamine during APT.
Collapse
Affiliation(s)
- Evandro M Neto-Neves
- Department of Pharmacology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
889
|
Saha D, Takahashi K, de Prost N, Winkler T, Pinilla-Vera M, Baron RM, Vidal Melo MF. Micro-autoradiographic assessment of cell types contributing to 2-deoxy-2-[(18)F]fluoro-D-glucose uptake during ventilator-induced and endotoxemic lung injury. Mol Imaging Biol 2013; 15:19-27. [PMID: 22752654 DOI: 10.1007/s11307-012-0575-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of the study was to use micro-autoradiography to investigate the lung cell types responsible for 2-deoxy-2-[(18)F]fluoro-D-glucose (FDG) uptake in murine models of acute lung injury (ALI). PROCEDURES C57/BL6 mice were studied in three groups: controls, ventilator-induced lung injury (VILI), and endotoxin. VILI was produced by high tidal volumes and zero end-expiratory pressure and endotoxin ALI, by intranasal administration. Following FDG injection, the lungs were processed and exposed to autoradiographic emulsion. Grain density over cells was used to quantify FDG uptake. RESULTS Neutrophils, macrophages, and type 2 epithelial cells presented higher grain densities during VILI and endotoxin ALI than controls. Remarkably, cell grain density in specific cell types was dependent on the injury mechanism. Whereas macrophages showed high grain densities during endotoxin ALI, similar to those exhibited by neutrophils, type 2 epithelial cells demonstrated the second highest grain density (with neutrophils as the highest) during VILI. CONCLUSIONS In murine models of VILI and endotoxin ALI, FDG uptake occurs not only in neutrophils but also in macrophages and type 2 epithelial cells. FDG uptake by individual cell types depends on the mechanism underlying ALI.
Collapse
Affiliation(s)
- Dalia Saha
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St., Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
890
|
Hirota JA, Hiebert PR, Gold M, Wu D, Graydon C, Smith JA, Ask K, McNagny K, Granville DJ, Knight DA. Granzyme B deficiency exacerbates lung inflammation in mice after acute lung injury. Am J Respir Cell Mol Biol 2013; 49:453-62. [PMID: 23642129 DOI: 10.1165/rcmb.2012-0512oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Granzyme B (GzmB) is a serine protease with intracellular and extracellular activities capable of regulating inflammation through cytokine processing and the apoptosis of effector cells. We tested the hypothesis that GzmB expression in T regulatory cells (Tregs) is required for the control of inflammatory responses and pathology during acute lung injury. To substantiate the clinical relevance of GzmB during lung injury, we performed GzmB immunohistochemistry on lung tissue from patients with acute respiratory distress syndrome (ARDS) and healthy control subjects. We also performed in vivo experiments with wild-type (WT) C57BL/6 and GzmB(-/-) mice exposed to a single intranasal instillation of bleomycin to model lung injury. Our results demonstrate that the expression of GzmB was elevated in ARDS lung sections, relative to healthy control samples. Bleomycin-exposed GzmB(-/-) mice exhibited greater morbidity and mortality, which was associated with increased numbers of lung lymphocytes. Bleomycin induced an equal increase in CD4(+)/CD25(+)/FoxP3(+) Treg populations in WT and GzmB(-/-) mice. GzmB expression was not significant in Tregs, with the majority of the expression localized to natural killer (NK)-1.1(+) cells. The expression of GzmB in NK cells of bleomycin-exposed WT mice was associated with greater lymphocyte apoptosis, reduced total lymphocyte numbers, and reduced pathology relative to GzmB(-/-) mice. Our data demonstrate that GzmB deficiency results in the exacerbation of lymphocytic inflammation during bleomycin-induced acute lung injury, which is associated with pathology, morbidity, and mortality.
Collapse
Affiliation(s)
- Jeremy A Hirota
- University of British Columbia James Hogg Research Centre, St. Paul's Hospital, 1081 Burrard St., Vancouver, BC, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
891
|
Monoacylglycerol lipase (MAGL) inhibition attenuates acute lung injury in mice. PLoS One 2013; 8:e77706. [PMID: 24204926 PMCID: PMC3808422 DOI: 10.1371/journal.pone.0077706] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 09/13/2013] [Indexed: 02/06/2023] Open
Abstract
Endocannabinoid signaling is terminated by enzymatic hydrolysis, a process that, for 2-Arachidonoylglycerol (2-AG), is mediated by monoacylglycerol lipase (MAGL). The piperidine carbamate, 4-nitrophenyl- 4-(dibenzo[d] [1,3]dioxol-5-yl (hydroxy) methyl) piperidine- 1-carboxylate (JZL184), is a drug that inhibits MAGL and presents high potency and selectivity. Thus, JZL184 increases the levels of 2-AG, an endocannabinoid that acts on the CB1 and CB2 cannabinoid receptors. Here, we investigated the effects of MAGL inhibition, with a single dose (16 mg/kg, intraperitoneally (i.p.)) of JZL184, in a murine model of lipopolysaccharide (LPS) -induced acute lung injury (ALI) 6, 24 and 48 hours after the inflammatory insult. Treatment with JZL184 decreased the leukocyte migration into the lungs as well as the vascular permeability measured through the bronchoalveolar lavage fluid (BAL) and histological analysis. JZL184 also reduced the cytokine and chemokine levels in the BAL and adhesion molecule expression in the blood and BAL. The CB1 and CB2 receptors were considered involved in the anti-inflammatory effects of JZL184 because the AM281 selective CB1 receptor antagonist (1-(2,4-dichlorophenyl)-5-(4-iodophenyl)-4-methyl-N-4-morpholinyl-1H-pyrazole-3-carboxamide) and the AM630 selective CB2 receptor antagonist ([6-iodo-2-methyl-1-[2-(4-morpholinyl)ethyl]-1H-indol-3-yl](4-methoxyphenyl)-methanone) blocked the anti-inflammatory effects previously described for JZL184. It was concluded that MAGL inhibition, and consequently the increase in 2-AG levels, produced anti-inflammatory effects in a murine model of LPS-induced ALI, a finding that was considered a consequence of the activation of the CB1 and CB2 receptors.
Collapse
|
892
|
Lewis SM, Khan N, Beale R, Treacher DF, Brown KA. Depletion of blood neutrophils from patients with sepsis: treatment for the future? Int Immunopharmacol 2013; 17:1226-32. [PMID: 24144812 DOI: 10.1016/j.intimp.2013.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Organ failure arising from severe sepsis accounts for nearly 6 million deaths worldwide per annum. At present there are no specific pharmacological agents available for its treatment and identifying a suitable therapeutic target is urgently needed. Neutrophils appear to be contributing directly to pulmonary damage in severe forms of lung injury and indirectly to the failure of other organs. Blood neutrophils from patients with sepsis possess a phenotype that is indicative of activation and our results show that neutrophils isolated from patients with sepsis exhibit a supranormal adherence to endothelial monolayers treated with pro-inflammatory cytokines. Additional studies reveal that the patients' cells are highly efficient at releasing IL-8. We also demonstrate that organ function is improved upon removing neutrophils from the circulation. In this article we propose that in severe sepsis there is a subpopulation of neutrophils which is actively engaged in pathological insult. The phenotypic characterisation of this subset may provide a novel therapeutic strategy for sepsis that could lead to patient benefit.
Collapse
Affiliation(s)
- Sion M Lewis
- Intensive Care Unit, Guy's and St. Thomas' NHS Foundation Trust, London, UK; Vascular Immunology, King's College London, UK
| | | | | | | | | |
Collapse
|
893
|
Fullerton JN, O'Brien AJ, Gilroy DW. Pathways mediating resolution of inflammation: when enough is too much. J Pathol 2013; 231:8-20. [PMID: 23794437 DOI: 10.1002/path.4232] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 06/11/2013] [Accepted: 06/17/2013] [Indexed: 01/22/2023]
Abstract
Patients with critical illness, and in particular sepsis, are now recognized to undergo unifying, pathogenic disturbances of immune function. Whilst scientific and therapeutic focus has traditionally been on understanding and modulating the initial pro-inflammatory limb, recent years have witnessed a refocusing on the development and importance of immunosuppressive 'anti-inflammatory' pathways. Several mechanisms are known to drive this phenomenon; however, no overriding conceptual framework justifies them. In this article we review the contribution of pro-resolution pathways to this phenotype, describing the observed immune alterations in terms of either a failure of resolution of inflammation or the persistence of pro-resolution processes causing inappropriate 'injurious resolution'-a novel hypothesis. The dysregulation of key processes in critical illness, including apoptosis of infiltrating neutrophils and their efferocytosis by macrophages, are discussed, along with the emerging role of specialized cell subtypes Gr1(+) CD11b(+) myeloid-derived suppressor cells and CD4(+) CD25(+) FoxP3(+) T-regulatory cells.
Collapse
Affiliation(s)
- James N Fullerton
- Centre for Clinical Pharmacology, Division of Medicine, University College London, London, UK.
| | | | | |
Collapse
|
894
|
Chen J, Mo Y, Schlueter CF, Hoyle GW. Inhibition of chlorine-induced pulmonary inflammation and edema by mometasone and budesonide. Toxicol Appl Pharmacol 2013; 272:408-13. [PMID: 23800689 PMCID: PMC4005342 DOI: 10.1016/j.taap.2013.06.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/29/2013] [Accepted: 06/11/2013] [Indexed: 12/21/2022]
Abstract
Chlorine gas is a widely used industrial compound that is highly toxic by inhalation and is considered a chemical threat agent. Inhalation of high levels of chlorine results in acute lung injury characterized by pneumonitis, pulmonary edema, and decrements in lung function. Because inflammatory processes can promote damage in the injured lung, anti-inflammatory therapy may be of potential benefit for treating chemical-induced acute lung injury. We previously developed a chlorine inhalation model in which mice develop epithelial injury, neutrophilic inflammation, pulmonary edema, and impaired pulmonary function. This model was used to evaluate nine corticosteroids for the ability to inhibit chlorine-induced neutrophilic inflammation. Two of the most potent corticosteroids in this assay, mometasone and budesonide, were investigated further. Mometasone or budesonide administered intraperitoneally 1h after chlorine inhalation caused a dose-dependent inhibition of neutrophil influx in lung tissue sections and in the number of neutrophils in lung lavage fluid. Budesonide, but not mometasone, reduced the levels of the neutrophil attractant CXCL1 in lavage fluid 6h after exposure. Mometasone or budesonide also significantly inhibited pulmonary edema assessed 1 day after chlorine exposure. Chlorine inhalation resulted in airway hyperreactivity to inhaled methacholine, but neither mometasone nor budesonide significantly affected this parameter. The results suggest that mometasone and budesonide may represent potential treatments for chemical-induced lung injury.
Collapse
Affiliation(s)
- Jing Chen
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yiqun Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Connie F. Schlueter
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Gary W. Hoyle
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| |
Collapse
|
895
|
Cornélio Favarin D, Robison de Oliveira J, Jose Freire de Oliveira C, de Paula Rogerio A. Potential effects of medicinal plants and secondary metabolites on acute lung injury. BIOMED RESEARCH INTERNATIONAL 2013; 2013:576479. [PMID: 24224172 PMCID: PMC3810192 DOI: 10.1155/2013/576479] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 08/16/2013] [Accepted: 08/23/2013] [Indexed: 12/20/2022]
Abstract
Acute lung injury (ALI) is a life-threatening syndrome that causes high morbidity and mortality worldwide. ALI is characterized by increased permeability of the alveolar-capillary membrane, edema, uncontrolled neutrophils migration to the lung, and diffuse alveolar damage, leading to acute hypoxemic respiratory failure. Although corticosteroids remain the mainstay of ALI treatment, they cause significant side effects. Agents of natural origin, such as medicinal plants and their secondary metabolites, mainly those with very few side effects, could be excellent alternatives for ALI treatment. Several studies, including our own, have demonstrated that plant extracts and/or secondary metabolites isolated from them reduce most ALI phenotypes in experimental animal models, including neutrophil recruitment to the lung, the production of pro-inflammatory cytokines and chemokines, edema, and vascular permeability. In this review, we summarized these studies and described the anti-inflammatory activity of various plant extracts, such as Ginkgo biloba and Punica granatum, and such secondary metabolites as epigallocatechin-3-gallate and ellagic acid. In addition, we highlight the medical potential of these extracts and plant-derived compounds for treating of ALI.
Collapse
Affiliation(s)
- Daniely Cornélio Favarin
- Departamento de Clínica Médica, Laboratório de ImunoFarmacologia Experimental, Instituto de Ciências da Saúde, Universidade Federal do Triângulo Mineiro, Rua Manoel Carlos 162, 38025-380 Uberaba, MG, Brazil
| | - Jhony Robison de Oliveira
- Departamento de Clínica Médica, Laboratório de ImunoFarmacologia Experimental, Instituto de Ciências da Saúde, Universidade Federal do Triângulo Mineiro, Rua Manoel Carlos 162, 38025-380 Uberaba, MG, Brazil
| | | | - Alexandre de Paula Rogerio
- Departamento de Clínica Médica, Laboratório de ImunoFarmacologia Experimental, Instituto de Ciências da Saúde, Universidade Federal do Triângulo Mineiro, Rua Manoel Carlos 162, 38025-380 Uberaba, MG, Brazil
| |
Collapse
|
896
|
Ulm C, Saffarzadeh M, Mahavadi P, Müller S, Prem G, Saboor F, Simon P, Middendorff R, Geyer H, Henneke I, Bayer N, Rinné S, Lütteke T, Böttcher-Friebertshäuser E, Gerardy-Schahn R, Schwarzer D, Mühlenhoff M, Preissner KT, Günther A, Geyer R, Galuska SP. Soluble polysialylated NCAM: a novel player of the innate immune system in the lung. Cell Mol Life Sci 2013; 70:3695-708. [PMID: 23619613 PMCID: PMC11113884 DOI: 10.1007/s00018-013-1342-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 04/10/2013] [Accepted: 04/11/2013] [Indexed: 11/27/2022]
Abstract
Posttranslational modification of the neural cell adhesion molecule (NCAM) by polysialic acid (polySia) is well studied in the nervous system and described as a dynamic modulator of plastic processes like precursor cell migration, axon fasciculation, and synaptic plasticity. Here, we describe a novel function of polysialylated NCAM (polySia-NCAM) in innate immunity of the lung. In mature lung tissue of healthy donors, polySia was exclusively attached to the transmembrane isoform NCAM-140 and located to intracellular compartments of epithelial cells. In patients with chronic obstructive pulmonary disease, however, increased polySia levels and processing of the NCAM carrier were observed. Processing of polysialylated NCAM was reproduced in a mouse model by bleomycin administration leading to an activation of the inflammasome and secretion of interleukin (IL)-1β. As shown in a cell culture model, polySia-NCAM-140 was kept in the late trans-Golgi apparatus of lung epithelial cells and stimulation by IL-1β or lipopolysaccharide induced metalloprotease-mediated ectodomain shedding, resulting in the secretion of soluble polySia-NCAM. Interestingly, polySia chains of secreted NCAM neutralized the cytotoxic activity of extracellular histones as well as DNA/histone-network-containing "neutrophil extracellular traps", which are formed during invasion of microorganisms. Thus, shedding of polySia-NCAM by lung epithelial cells may provide a host-protective mechanism to reduce tissue damage during inflammatory processes.
Collapse
Affiliation(s)
- Christina Ulm
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany
| | - Mona Saffarzadeh
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany
| | - Poornima Mahavadi
- Department of Internal Medicine II, Justus-Liebig-University, Giessen, Germany
| | - Sandra Müller
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany
| | - Gerlinde Prem
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany
| | - Farhan Saboor
- Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, Giessen, Germany
| | - Peter Simon
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany
| | - Ralf Middendorff
- Institute of Anatomy and Cell Biology, Faculty of Medicine, Justus-Liebig-University, Giessen, Germany
| | - Hildegard Geyer
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany
| | - Ingrid Henneke
- Department of Internal Medicine II, Justus-Liebig-University, Giessen, Germany
| | - Nils Bayer
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany
| | - Susanne Rinné
- Institute of Physiology and Pathophysiology, Philipps-University, Marburg, Germany
| | - Thomas Lütteke
- Institute of Veterinary Physiology and Biochemistry, Justus-Liebig-University, Giessen, Germany
| | | | | | - David Schwarzer
- Institute of Cellular Chemistry, Medical School, Hannover, Germany
| | | | | | - Andreas Günther
- Department of Internal Medicine II, Justus-Liebig-University, Giessen, Germany
| | - Rudolf Geyer
- Institute of Biochemistry, Justus-Liebig-University, Giessen, Germany
| | | |
Collapse
|
897
|
Frevert U, Nacer A, Cabrera M, Movila A, Leberl M. Imaging Plasmodium immunobiology in the liver, brain, and lung. Parasitol Int 2013; 63:171-86. [PMID: 24076429 DOI: 10.1016/j.parint.2013.09.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 08/28/2013] [Accepted: 09/18/2013] [Indexed: 01/10/2023]
Abstract
Plasmodium falciparum malaria is responsible for the deaths of over half a million African children annually. Until a decade ago, dynamic analysis of the malaria parasite was limited to in vitro systems with the typical limitations associated with 2D monocultures or entirely artificial surfaces. Due to extremely low parasite densities, the liver was considered a black box in terms of Plasmodium sporozoite invasion, liver stage development, and merozoite release into the blood. Further, nothing was known about the behavior of blood stage parasites in organs such as the brain where clinical signs manifest and the ensuing immune response of the host that may ultimately result in a fatal outcome. The advent of fluorescent parasites, advances in imaging technology, and availability of an ever-increasing number of cellular and molecular probes have helped illuminate many steps along the pathogenetic cascade of this deadly tropical parasite.
Collapse
Affiliation(s)
- Ute Frevert
- Division of Medical Parasitology, Department of Microbiology, New York University School of Medicine, 341 E 25 Street, New York, NY 10010, USA.
| | | | | | | | | |
Collapse
|
898
|
Ban K, Peng Z, Kozar RA. Inhibition of ERK1/2 worsens intestinal ischemia/reperfusion injury. PLoS One 2013; 8:e76790. [PMID: 24073294 PMCID: PMC3779170 DOI: 10.1371/journal.pone.0076790] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 09/03/2013] [Indexed: 01/09/2023] Open
Abstract
Background The role of extracellular signal-regulated protein kinase (ERK) in intestinal ischemia/reperfusion (I/R) injury has not been well investigated. The aim of the current study was to examine the effect of inhibition of the ERK pathway in an in vitro and in vivo model of intestinal I/R injury. Methods ERK1/2 activity was inhibited using the specific inhibitor, U0126, in intestinal epithelial cells under hypoxia/reoxygenation conditions and in mice subjected to 1 hour of intestinal ischemia followed by 6 hours reperfusion. In vitro, cell proliferation was assessed by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay, apoptosis by DNA fragmentation, and migration using an in vitro model of intestinal wound healing. Cells were also transfected with a p70S6K plasmid and the effects of overexpression similarly analyzed. In vivo, the effects of U0126 on intestinal cell proliferation and apoptosis, intestinal permeability, lung and intestinal neutrophil infiltration and injury, and plasma cytokine levels were measured. Survival was also assessed after U0126. Activity of p70S6 kinase (p70S6K) was measured by Western blot. Results In vitro, inhibition of ERK1/2 by U0126 significantly decreased cell proliferation and migration but enhanced cell apoptosis. Overexpression of p70S6K promoted cell proliferation and decreased cell apoptosis. In vivo, U0126 significantly increased cell apoptosis and decreased cell proliferation in the intestine, increased intestinal permeability, intestinal and lung neutrophil infiltration, and injury, as well as systemic pro-inflammatory cytokines, TNF-α, IL-6 and IL-1β. Mortality was also significantly increased by U0126. Inhibition of ERK1/2 by U0126 also abolished activity of p70S6K both in vitro and in vivo models. Conclusion Pharmacologic inhibition of ERK1/2 by U0126 worsens intestinal IR injury. The detrimental effects are mediated, at least in part, by inhibition of p70S6K, the major effector of mammalian target of rapamycin pathway.
Collapse
Affiliation(s)
- Kechen Ban
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- * E-mail:
| | - Zhanglong Peng
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Rosemary A. Kozar
- Department of Surgery, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
899
|
Zhang S, Song L, Wang Y, Herwald H, Thorlacius H. Targeting CD162 protects against streptococcal M1 protein-evoked neutrophil recruitment and lung injury. Am J Physiol Lung Cell Mol Physiol 2013; 305:L756-63. [PMID: 24039252 DOI: 10.1152/ajplung.00220.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Streptococcus pyogenes of the M1 serotype can cause streptococcal toxic shock syndrome and acute lung damage. CD162 is an adhesion molecule that has been reported to mediate neutrophil recruitment in acute inflammatory reactions. In this study, the purpose was to investigate the role of CD162 in M1 protein-provoked lung injury. Male C57BL/6 mice were treated with monoclonal antibody directed against CD162 or a control antibody before M1 protein challenge. Edema, neutrophil infiltration, and CXC chemokines were determined in the lung, 4 h after M1 protein administration. Fluorescence intravital microscopy was used to analyze leukocyte-endothelium interactions in the pulmonary microcirculation. Inhibition of CD162 reduced M1 protein-provoked accumulation of neutrophils, edema, and CXC chemokine formation in the lung by >54%. Moreover, immunoneutralization of CD162 abolished leukocyte rolling and firm adhesion in pulmonary venules of M1 protein-treated animals. In addition, inhibition of CD162 decreased M1 protein-induced capillary trapping of leukocytes in the lung microvasculature and improved microvascular perfusion in the lungs of M1 protein-treated animals. Our findings suggest that CD162 plays an important role in M1 protein-induced lung damage by regulating leukocyte rolling in pulmonary venules. Consequently, inhibition of CD162 attenuates M1 protein-evoked leukocyte adhesion and extravasation in the lung. Thus, our results suggest that targeting the CD162 might pave the way for novel opportunities to protect against pulmonary damage in streptococcal infections.
Collapse
Affiliation(s)
- Songen Zhang
- Dept. of Clinical Sciences, Malmö, Section of Surgery, Skåne Univ. Hospital, Lund Univ., 205 02 Malmö, Sweden.
| | | | | | | | | |
Collapse
|
900
|
Wan LM, Tan L, Wang ZR, Liu SX, Wang YL, Liang SY, Zhong JB, Lin HS. Preventive and therapeutic effects of Danhong injection on lipopolysaccharide induced acute lung injury in mice. JOURNAL OF ETHNOPHARMACOLOGY 2013; 149:352-359. [PMID: 23850708 DOI: 10.1016/j.jep.2013.06.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 05/10/2013] [Accepted: 06/24/2013] [Indexed: 06/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Danhong injection (DHI), a Chinese Materia Medica standardized product extracted from Radix et Rhizoma Salviae Miltiorrhizae (Salvia miltiorrhiza Bge., Labiatae, Danshen in Chinese) and Flos Carthami (Carthamus tinctorius L., Compositae, Honghua in Chinese), has been reported to have anti-inflammatory, anti-oxidative and anti-fibrinolytic properties, which is used extensively for the treatment of cardiovascular diseases in clinic. AIM OF THIS STUDY The present study aimed to investigate the preventive and therapeutic effects of DHI on lipopolysaccharide (LPS) induced acute lung injury (ALI) in mice. MATERIALS AND METHODS Lung injury was induced by intranasal instillation with 10 μg LPS. Mice were randomly divided into four groups: Control group; LPS group; LPS+5 ml/kg DHI group and LPS+10 ml/kg DHI group. The effects of DHI on LPS-induced neutrophils influx, inflammatory cytokines release, protein leakage, myeloperoxidase (MPO) and superoxide dismutase (SOD) activities, malondialdehyde (MDA) level were examined. In addition, the NF-κB activation in lung tissues was detected by Western blot. RESULTS In LPS challenged mice, DHI significantly reduced the infiltration of activated neutrophils and decreased the levels of TNF-α and IL-6 in bronchoalveolar lavage fluid (BALF). DHI also inhibited protein extravasation in BALF, attenuated edema and the pathological changes in the lung. In addition, DHI markedly prevented LPS-induced elevation of MDA and MPO levels, as well as reduction of SOD activity. Further study demonstrated that DHI effectively inhibited the NF-κB activation in lung tissues. CONCLUSION DHI has been demonstrated to protect mice from LPS induced acute lung injury by its anti-inflammatory and anti-oxidant activities.
Collapse
Affiliation(s)
- Li-Mei Wan
- Department of Respiratory Medicine, Zengcheng people's hospital (Boji-affiliated hospital of Sun Yat-Sen university), Guangzhou 511300, China
| | | | | | | | | | | | | | | |
Collapse
|