Phospholipase A in acute lung injury after trauma and sepsis: Its relation to the inflammatory mediators PMN-elastase, C3a, and neopterin

1989 ◽  
Vol 67 (3) ◽  
pp. 190-195 ◽  
Author(s):  
W. Kellermann ◽  
R. Frentzel-Beyme ◽  
M. Welte ◽  
M. Jochum
2021 ◽  
Vol 12 ◽  
Author(s):  
Chunguang Yan ◽  
Jing Chen ◽  
Yue Ding ◽  
Zetian Zhou ◽  
Bingyu Li ◽  
...  

BackgroundThe ligand-activated transcription factor peroxisome proliferator-activated receptor (PPAR) γ plays crucial roles in diverse biological processes including cellular metabolism, differentiation, development, and immune response. However, during IgG immune complex (IgG-IC)-induced acute lung inflammation, its expression and function in the pulmonary tissue remains unknown.ObjectivesThe study is designed to determine the effect of PPARγ on IgG-IC-triggered acute lung inflammation, and the underlying mechanisms, which might provide theoretical basis for therapy of acute lung inflammation.SettingDepartment of Pathogenic Biology and Immunology, Medical School of Southeast UniversitySubjectsMice with down-regulated/up-regulated PPARγ activity or down-regulation of Early growth response protein 1 (Egr-1) expression, and the corresponding controls.InterventionsAcute lung inflammation is induced in the mice by airway deposition of IgG-IC. Activation of PPARγ is achieved by using its agonist Rosiglitazone or adenoviral vectors that could mediate overexpression of PPARγ. PPARγ activity is suppressed by application of its antagonist GW9662 or shRNA. Egr-1 expression is down-regulated by using the gene specific shRNA.Measures and Main ResultsWe find that during IgG-IC-induced acute lung inflammation, PPARγ expression at both RNA and protein levels is repressed, which is consistent with the results obtained from macrophages treated with IgG-IC. Furthermore, both in vivo and in vitro data show that PPARγ activation reduces IgG-IC-mediated pro-inflammatory mediators’ production, thereby alleviating lung injury. In terms of mechanism, we observe that the generation of Egr-1 elicited by IgG-IC is inhibited by PPARγ. As an important transcription factor, Egr-1 transcription is substantially increased by IgG-IC in both in vivo and in vitro studies, leading to augmented protein expression, thus amplifying IgG-IC-triggered expressions of inflammatory factors via association with their promoters.ConclusionDuring IgG-IC-stimulated acute lung inflammation, PPARγ activation can relieve the inflammatory response by suppressing the expression of its downstream target Egr-1 that directly binds to the promoter regions of several inflammation-associated genes. Therefore, regulation of PPARγ-Egr-1-pro-inflammatory mediators axis by PPARγ agonist Rosiglitazone may represent a novel strategy for blockade of acute lung injury.


1995 ◽  
Vol 23 (6) ◽  
pp. 683-686 ◽  
Author(s):  
J. P. Arnold ◽  
M. Haeger ◽  
J. P. Bengtson ◽  
A. Bengtsson ◽  
B. Lisander

Ten patients undergoing hip replacement surgery were studied regarding activation of complement and leukocytes in association with collection of wound drainage blood. The blood was collected postoperatively but not reinfused due to the possible risks with reinfusion of blood containing inflammatory mediators. Blood samples for analysis of complement activation (TCC), leukocyte activation (PMN elastase) and cytokines (Interleukin-6) were drawn preoperatively from the patients. Blood samples were also drawn intraoperatively from the wound. Samples were also drawn from the collected wound drainage blood, before and after blood was passed through a microporous filter. There were elevated concentrations of TCC, PMN elastase and IL-6 in the collected wound drainage blood before and after the filter. The filtration did not significantly reduce the concentrations of these factors. In the wound blood the concentrations were higher compared to those found in the systemic blood preoperatively, but lower compared to concentrations found in the collected drainage blood. The study demonstrates that the collection of wound drainage whole blood is associated with activation of complement, release of PMN elastase and cytokines.


2017 ◽  
Vol 112 ◽  
pp. 208-209
Author(s):  
Jose Pablo Vazquez-Medina ◽  
Jian-Quin Tao ◽  
Priyal Patel ◽  
Renata Bannitz-Fernandes ◽  
Chandra Dodia ◽  
...  

2016 ◽  
Vol 2016 ◽  
pp. 1-12 ◽  
Author(s):  
Chunguang Yan ◽  
Fuqin Guan ◽  
Yanfei Shen ◽  
Huifang Tang ◽  
Dong Yuan ◽  
...  

Optimal methods are applied to acute lung injury (ALI) and the acute respiratory distress syndrome (ARDS), but the mortality rate is still high. Accordingly, further studies dedicated to identify novel therapeutic approaches to ALI are urgently needed. Bigelovii A is a new natural product and may exhibit anti-inflammatory activity. Therefore, we sought to investigate its effect on lipopolysaccharide- (LPS-) induced ALI and the underlying mechanisms. We found that LPS-induced ALI was significantly alleviated by Bigelovii A treatment, characterized by reduction of proinflammatory mediator production, neutrophil infiltration, and lung permeability. Furthermore, Bigelovii A also downregulated LPS-stimulated inflammatory mediator expressionsin vitro. Moreover, both NF-κB and CCAAT/enhancer-binding proteinδ(C/EBPδ) activation were obviously attenuated by Bigelovii A treatment. Additionally, phosphorylation of both p38 MAPK and ERK1/2 (upstream signals of C/EBPδactivation) in response to LPS challenge was also inhibited by Bigelovii A. Therefore, Bigelovii A could attenuate LPS-induced inflammation by suppression of NF-κB, inflammatory mediators, and p38 MAPK/ERK1/2—C/EBPδ, inflammatory mediators signaling pathways, which provide a novel theoretical basis for the possible application of Bigelovii A in clinic.


Lung ◽  
2005 ◽  
Vol 183 (2) ◽  
pp. 87-100 ◽  
Author(s):  
Xia Zhao ◽  
Marwan Dib ◽  
Ellen Andersson ◽  
Changbin Shi ◽  
Bengt Widegren ◽  
...  

2014 ◽  
Vol 116 (12) ◽  
pp. 1521-1530 ◽  
Author(s):  
Aron B. Fisher

The Comroe lecture on which this review is based described my research path during the past 45 years, beginning with studies of oxidant stress (hyperoxia) and eventuating in the discovery of a synthetic inhibitor of phospholipase A2activity (called MJ33) that prevents acute lung injury in mice exposed to lipopolysaccharide. In between were studies of lung ischemia, lung surfactant metabolism, the protein peroxiredoxin 6 and its phospholipase A2activity, and mechanisms for NADPH oxidase activation. These seemingly unrelated research activities provided the nexus for identification of a novel target and a potentially novel therapeutic agent for prevention or treatment of acute lung injury.


2015 ◽  
Vol 765 ◽  
pp. 447-456 ◽  
Author(s):  
Soumen Choudhury ◽  
Kannan Kandasamy ◽  
Bhojane Somnath Maruti ◽  
M. Pule Addison ◽  
Jaya Kiran Kasa ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document