51
|
Li L, Wu W, Huang W, Hu G, Yuan W, Li W. NF-κB RNAi decreases the Bax/Bcl-2 ratio and inhibits TNF-α-induced apoptosis in human alveolar epithelial cells. Inflamm Res 2013; 62:387-97. [PMID: 23334076 DOI: 10.1007/s00011-013-0590-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 12/15/2012] [Accepted: 01/02/2013] [Indexed: 01/31/2023] Open
Abstract
OBJECTIVE Apoptosis of alveolar epithelial cells (AECs) plays a key role in acute lung injury (ALI). Understanding the underlying mechanism is conducive to the treatment of ALI. The goal of this study was to determine the possible involvement of nuclear factor-κB (NF-κB)/p65 and Bax/Bcl-2 in tumor necrosis factor-α (TNF-α)-induced apoptosis in AECs. METHODS Type II AECs, A549, with or without NF-κB/p65 expression silenced by small interfering RNA (siRNA) were challenged with TNF-α. The levels of NF-κB/p65, Bcl-2 and Bax were detected by reverse transcription-polymerase chain reaction, Western blotting, and immunocytochemical staining. The apoptosis rate was measured by flow cytometry. RESULTS TNF-α challenge significantly increased the transcription and translation of NF-κB/p65 and Bax genes, but significantly decreased the Bcl-2 gene level. siRNA silencing of NF-κB/p65 reversed the effect of TNF-α on NF-κB/p65, Bcl-2 and Bax, and significantly decreased the TNF-α-induced apoptosis rate of AECs, as compared to the non-silenced cells. CONCLUSIONS This study indicates that NF-κB plays an important role in the process of TNF-α-induced apoptosis in AECs, via regulation of the expression of Bcl-2 and Bax.
Collapse
Affiliation(s)
- Li Li
- Department of Respiratory Medicine, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010 Guangdong, People's Republic of China
| | | | | | | | | | | |
Collapse
|
52
|
Anti-inflammatory effects of mangiferin on sepsis-induced lung injury in mice via up-regulation of heme oxygenase-1. J Nutr Biochem 2012; 24:1173-81. [PMID: 23266284 DOI: 10.1016/j.jnutbio.2012.09.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 08/27/2012] [Accepted: 09/17/2012] [Indexed: 11/21/2022]
Abstract
Sepsis, a serious unbalanced hyperinflammatory condition, is a tremendous burden for healthcare systems, with a high mortality and limited treatment. Increasing evidences indicated that some active components derived from natural foods have potent anti-inflammatory properties. Here we show that mangiferin (MF), a natural glucosyl xanthone found in both mango and papaya, attenuates cecal ligation and puncture-induced mortality and acute lung injury (ALI), as indicated by reduced systemic and pulmonary inflammatory responses. Moreover, pretreatment with MF inhibits sepsis-activated mitogen-activated protein kinases and nuclear factor kappa-light-chain-enhancer of activated B cells signaling, resulting in inhibiting production of proinflammatory mediators. Notably, MF dose-dependently up-regulates the expression and activity of heme oxygenase (HO)-1 in the lung of septic mice. Further, these beneficial effects of MF on the septic lung injury were eliminated by ZnPP IX, a specific HO-1 inhibitor. Our results suggest that MF attenuates sepsis by up-regulation of HO-1 that protects against sepsis-induced ALI through inhibiting inflammatory signaling and proinflammatory mediators. Thereby, MF may be effective in treating sepsis with ALI.
Collapse
|
53
|
Mantell S, Jones R, Trevethick M. Design and application of locally delivered agonists of the adenosine A(2A) receptor. Expert Rev Clin Pharmacol 2012; 3:55-72. [PMID: 22111533 DOI: 10.1586/ecp.09.57] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The broad spectrum anti-inflammatory actions of adenosine A(2A) receptor agonists are well described. The wide distribution of this receptor, however, suggests that the therapeutic potential of these agents is likely to reside in topical treatments to avoid systemic side effects associated with oral administration. Adenosine A(2A) receptor agonists have been assessed as topical agents: GW328267X (GSK; allergic rhinitis and asthma), UK-432097 (Pfizer; chronic obstructive pulmonary disease [COPD]) and Sonedenoson (MRE0094, King Pharmaceuticals; wound healing). All trials failed to achieve effects against the desired clinical end points. This broad-based review will discuss general principles of chemical design of topically applied agents and potential therapeutic topical applications of current adenosine A(2A) receptor agonists. Potential factors contributing to the lack of efficacy in the above clinical trials will be discussed together with design principles, which may influence efficacy in disease states. Our analysis suggests that adenosine A(2A) receptor agonists have a wide therapeutic potential as topical agents in a wide variety of diseases, such as neutrophil-dependent lung diseases (acute lung injury, exacerbations in asthma and COPD), allergic rhinitis, glaucoma and wound repair. Factors that will influence topical activity include formulation, tissue retention, compound potency, receptor kinetics and pharmacokinetics.
Collapse
Affiliation(s)
- Simon Mantell
- PC 675, Pfizer Global R&D, Sandwich, Kent, CT13 9NJ, UK.
| | | | | |
Collapse
|
54
|
Abstract
More than 50% of severely injured patients have chest trauma. Second insults frequently result in acute lung injury (ALI), with sepsis being the main underlying condition. We aimed to develop a standardized, reproducible, and clinically relevant double-hit mouse model of ALI induced by chest trauma and polymicrobial sepsis and to investigate the pathophysiologic role of activated neutrophils. Lung contusion was applied to C57Bl/6 mice via a focused blast wave. Twenty-four hours later, sepsis was induced by cecal ligation and puncture. For polymorphonuclear leukocyte (PMN) depletion, animals received intravenous injections of PMN-depleting antibody. In response to blunt chest trauma followed by sepsis as well as after sepsis alone, a significant local and systemic inflammatory response with increased cytokine/chemokine levels in lung and plasma was observed. In contrast, lung apoptosis was markedly elevated only after a double hit. Intra-alveolar neutrophils and total bronchoalveolar lavage protein concentrations were markedly increased following isolated chest trauma or the combined insult, but not after sepsis alone. Lung myeloperoxidase activity was enhanced only in response to the double hit accompanied by histological disruption of the alveolar architecture, lung congestion, and marked cellular infiltrates. Neutrophil depletion significantly diminished lung interleukin 1β and interleukin 6 concentrations and reduced the degree of septic ALI. Here we have established a novel and highly reproducible mouse model of chest trauma-induced septic ALI characterizing a clinical relevant double-hit scenario. In particular, the depletion of neutrophils substantially mitigated the extent of lung injury, indicating a pathomechanistic role for neutrophils in chest trauma-induced septic ALI.
Collapse
|
55
|
Calisto KL, Camacho AC, Mittestainer FC, Carvalho BM, Guadagnini D, Carvalheira JB, Saad MJ. Diacerhein attenuates the inflammatory response and improves survival in a model of severe sepsis. Crit Care 2012; 16:R158. [PMID: 22897821 PMCID: PMC3580748 DOI: 10.1186/cc11478] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 06/21/2012] [Accepted: 08/13/2012] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Hyperglycemia and insulin resistance have been associated with a worse outcome in sepsis. Although tight glycemic control through insulin therapy has been shown to reduce morbidity and mortality rates, the effect of intensive insulin therapy in patients with severe sepsis is controversial because of the increased risk of serious adverse events related to hypoglycemia. Recently, knowledge about diacerhein, an anthraquinone drug with powerful antiinflammatory properties, revealed that this drug improves insulin sensitivity, mediated by the reversal of chronic subclinical inflammation. The aim of the present study was to evaluate whether the antiinflammatory effects of diacerhein after onset of sepsis-induced glycemic alterations is beneficial and whether the survival rate is prolonged in this situation. METHODS Diffuse sepsis was induced by cecal ligation and puncture surgery (CLP) in male Wistar rats. Blood glucose and inflammatory cytokine levels were assessed 24 hours after CLP. The effect of diacerhein on survival of septic animals was investigated in parallel with insulin signaling and its modulators in liver, muscle, and adipose tissue. RESULTS Here we demonstrated that diacerhein treatment improves survival during peritoneal-induced sepsis and inhibits sepsis-induced insulin resistance by improving insulin signaling via increased insulin-receptor substrate-1-associated phosphatidylinositol 3-kinase activity and Akt phosphorylation. Diacerhein also decreases the activation of endoplasmic reticulum stress signaling that involves upregulation of proinflammatory pathways, such as the I kappa B kinase and c-Jun NH2-terminal kinase, which blunts insulin-induced insulin signaling in liver, muscle, and adipose tissue. Additionally, our data show that this drug promoted downregulation of proinflammatory signaling cascades that culminate in transcription of immunomodulatory factors such interleukin (IL)-1β, IL-6, and tumor necrosis factor-α. CONCLUSIONS This study demonstrated that diacerhein treatment increases survival and attenuates the inflammatory response with a significant effect on insulin sensitivity. On the basis of efficacy and safety profile, diacerhein represents a novel antiinflammatory therapy for management of insulin resistance in sepsis and a potential approach for future clinical trials.
Collapse
Affiliation(s)
- Kelly L Calisto
- Department of Internal Medicine, FCM, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil
| | - Angélica C Camacho
- Department of Internal Medicine, FCM, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil
| | - Francine C Mittestainer
- Department of Internal Medicine, FCM, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil
| | - Bruno M Carvalho
- Department of Internal Medicine, FCM, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil
| | - Dioze Guadagnini
- Department of Internal Medicine, FCM, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil
| | - José B Carvalheira
- Department of Internal Medicine, FCM, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil
| | - Mario J Saad
- Department of Internal Medicine, FCM, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, SP, Brazil
| |
Collapse
|
56
|
Hudock KM, Liu Y, Mei J, Marino RC, Hale JE, Dai N, Worthen GS. Delayed resolution of lung inflammation in Il-1rn-/- mice reflects elevated IL-17A/granulocyte colony-stimulating factor expression. Am J Respir Cell Mol Biol 2012; 47:436-44. [PMID: 22592923 DOI: 10.1165/rcmb.2012-0104oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IL-1 has been associated with acute lung injury (ALI) in both humans and animal models, but further investigation of the precise mechanisms involved is needed, and may identify novel therapeutic targets. To discover the IL-1 mediators essential to the initiation and resolution phases of acute lung inflammation, knockout mice (with targeted deletions for either the IL-1 receptor-1, i.e., Il-1r1(-/-), or the IL-1 receptor antagonist, i.e., Il-1rn(-/-)) were exposed to aerosolized LPS, and indices of lung and systemic inflammation were examined over the subsequent 48 hours. The resultant cell counts, histology, protein, and RNA expression of key cytokines were measured. Il-1r1(-/-) mice exhibited decreased neutrophil influx, particularly at 4 and 48 hours after exposure to LPS, as well as reduced bronchoalveolar lavage (BAL) expression of chemokines and granulocyte colony-stimulating factor (G-CSF). On the contrary, Il-1rn(-/-) mice demonstrated increased BAL neutrophil counts, increased BAL total protein, and greater evidence of histologic injury, all most notably 2 days after LPS exposure. Il-1rn(-/-) mice also exhibited higher peripheral neutrophil counts and greater numbers of granulocyte receptor-1 cells in their bone marrow, potentially reflecting their elevated plasma G-CSF concentrations. Furthermore, IL-17A expression was increased in the BAL and lungs of Il-1rn(-/-) mice after exposure to LPS, likely because of increased numbers of γδ T cells in the Il-1rn(-/-) lungs. Blockade with IL-17A monoclonal antibody before LPS exposure decreased the resultant BAL neutrophil counts and lung G-CSF expression in Il-1rn(-/-) mice, 48 hours after exposure to LPS. In conclusion, Il-1rn(-/-) mice exhibit delayed resolution in acute lung inflammation after exposure to LPS, a process that appears to be mediated via the G-CSF/IL-17A axis.
Collapse
Affiliation(s)
- Kristin M Hudock
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Pearlman School of Medicine, University of Pennsylvania, Abramson Research Building, Rm. 414E, 3615 Civic Center Blvd., Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | |
Collapse
|
57
|
Peng Z, Zhu Y, Zhang Y, Wilhelmsen K, Jia C, Jin J, Xue Q, Feng X, Zhang F, Yu B. Effects of ghrelin on pulmonary NOD2 mRNA expression and NF-κB activation when protects against acute lung injury in rats challenged with cecal ligation and puncture. Int Immunopharmacol 2012; 13:440-5. [PMID: 22575870 DOI: 10.1016/j.intimp.2012.04.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Revised: 04/09/2012] [Accepted: 04/11/2012] [Indexed: 12/18/2022]
Abstract
BACKGROUND Many studies have shown that ghrelin can down-regulate inflammatory cytokine expression via the inhibition of NF-κB activity and therefore, its administration to septic patients is considered beneficial. However, our knowledge of ghrelin's effects on the upstream activators of the NF-κB pathway, such as NOD2, is still limited. This study aimed to investigate the possible involvement of the NOD2 signaling pathway in the anti-inflammatory effects of ghrelin. METHODS Twenty-four male SD rats received cecal ligation and puncture (CLP) or sham operation, followed by infusion of saline or ghrelin. The lungs were harvested 6h after CLP or sham operation and analyzed for lung histopathology, neutrophil infiltration, inflammatory cytokines (TNF-α, and IL-6), NOD2 mRNA expression, and activation of NF-κB. Furthermore, survival was recorded for ten days in additional groups of rats. RESULTS Compared with sham group, neutrophil infiltration, TNF-α and IL-6 levels, NOD2 mRNA expression, as well as NF-κB activation in lungs from rats undergoing CLP were significantly increased. After the administration of ghrelin, all inflammatory parameters analyzed were lower than those without ghrelin following CLP. In addition, ghrelin improved survival after CLP. CONCLUSION Our results indicate that in a CLP model of sepsis, the beneficial effects that ghrelin has on inflammatory outcomes are mediated at least in part through inhibition of NOD2 expression upstream of NF-κB.
Collapse
Affiliation(s)
- Zhiyou Peng
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Yang KY, Shih HC, How CK, Chen CY, Hsu HS, Yang CW, Lee YC, Perng RP, Peng CH, Li HY, Chang CM, Mou CY, Chiou SH. IV delivery of induced pluripotent stem cells attenuates endotoxin-induced acute lung injury in mice. Chest 2011; 140:1243-1253. [PMID: 21835903 DOI: 10.1378/chest.11-0539] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Induced pluripotent stem (iPS) cells are novel stem cell populations, but the role of iPS cells in acute lung injury (ALI) is not currently known. We investigated the effect of iPS cells in modifying the pathophysiology of endotoxin-induced ALI. METHODS Male C57BL/6 8- to 12-week-old mice were enrolled in this study. Mouse iPS cells were delivered through the tail veins of mice 4 h after intratracheal instillation of endotoxin. Lung histopathologic findings, the pulmonary levels of cytokines, and functional parameters were analyzed after either 24 h or 48 h. RESULTS More iPS cells integrated into the lungs of mice with ALI than those of the control mice, as demonstrated by in vivo radionuclide imaging and in vitro Hoechst-labeled fluorescent staining. iPS cells significantly diminished the histopathologic changes of ALI and the lung injury score. There was also a significant reduction in the activity of nuclear factor-κB (NF-κB) and neutrophil accumulation in the lung, confirmed by immunostaining, electrophoretic mobility shift assays, and the decrease of myeloperoxidase activity, in the iPS-cell-treated mice with ALI. These protective effects were not replicated by the control cell therapy with fibroblasts. iPS cells mediated a downregulation of the proinflammatory response to endotoxin (reducing tumor necrosis factor-α, IL-6, and macrophage inflammatory peptide-2). In addition, iPS cells rescued the hypoxemia and pulmonary function of ALI. Treatment with a conditioned medium of iPS cells showed effects similar to those of iPS cells, which may suggest the therapeutic benefits of iPS mediated by paracrine factors. CONCLUSIONS IV delivery of iPS cells provides a beneficial effect to attenuate the severity of endotoxin-induced ALI and improve physiologic impairment, which is partly mediated by a reduction in NF-κB activity and neutrophils accumulation. The conditioned medium of iPS cells demonstrated effects equal to those of iPS cells.
Collapse
Affiliation(s)
- Kuang-Yao Yang
- Department of Chest Medicine, Taipei Veterans General Hospital; Institute of Clinical Medicine, National Yang-Ming University
| | - Hsin-Chin Shih
- Department of Emergency Medicine, Taipei Veterans General Hospital; Institute of Clinical Medicine, National Yang-Ming University; Institute of Emergency and Critical Care Meicine, National Yang-Ming University
| | - Chorng-Kuang How
- Department of Emergency Medicine, Taipei Veterans General Hospital; Institute of Clinical Medicine, National Yang-Ming University; Institute of Emergency and Critical Care Meicine, National Yang-Ming University
| | - Cheng-Yu Chen
- Institute of Clinical Medicine, National Yang-Ming University; School of Medicine, National Yang-Ming University; Department of Medical Research and Education, National Yang-Ming University Hospital
| | - Han-Shui Hsu
- Department of Surgery, Taipei Veterans General Hospital; School of Medicine, National Yang-Ming University
| | - Ching-Wen Yang
- Department of Surgery, Taipei Veterans General Hospital; School of Medicine, National Yang-Ming University
| | - Yu-Chin Lee
- Department of Chest Medicine, Taipei Veterans General Hospital; School of Medicine, National Yang-Ming University
| | - Reury-Perng Perng
- Department of Chest Medicine, Taipei Veterans General Hospital; School of Medicine, National Yang-Ming University
| | - Chi-Hsien Peng
- Institute of Clinical Medicine, National Yang-Ming University; Shin Kong Wu Ho-Su Memorial Hospital and Fu-Jen Catholic University, National Taiwan University, Taipei, Taiwan, China
| | - Hsin-Yang Li
- Department of Medical Research and Education, Taipei Veterans General Hospital; Department of Obstetrics and Gynecology, Taipei Veterans General Hospital; Institute of Clinical Medicine, National Yang-Ming University; School of Medicine, National Yang-Ming University
| | - Chia-Ming Chang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital; Institute of Oral Biology, National Yang-Ming University; School of Medicine, National Yang-Ming University
| | - Chung-Yuan Mou
- Department of Chemistry, College of Science, National Taiwan University, Taipei, Taiwan, China
| | - Shih-Hwa Chiou
- Department of Surgery, Taipei Veterans General Hospital; Institute of Clinical Medicine, National Yang-Ming University; Institute of Pharmacology, National Yang-Ming University; School of Medicine, National Yang-Ming University.
| |
Collapse
|
59
|
Perl M, Lomas-Neira J, Venet F, Chung CS, Ayala A. Pathogenesis of indirect (secondary) acute lung injury. Expert Rev Respir Med 2011; 5:115-26. [PMID: 21348592 DOI: 10.1586/ers.10.92] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
At present, therapeutic interventions to treat acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) remain largely limited to lung-protective strategies, as no real molecular-pathophysiologic-driven therapeutic intervention has yet become available. This is in part the result of the heterogeneous nature of the etiological processes that contribute to the state of ALI/ARDS. This article sets out to understand the development of ALI resulting from indirect pulmonary insults, such as extrapulmonary sepsis and trauma, shock, burn injury or mass transfusion, as opposed to direct pulmonary challenges, such as pneumonia, aspiration or lung contusion. Here, we consider not only the experimental and clinical data concerning the roles of various immune (neutrophil, macrophage, lymphocyte and dendritic) as well as nonimmune (epithelial and endothelial) cells in orchestrating the development of ALI resulting from indirect pulmonary stimuli, but also how these cell populations might be targeted therapeutically.
Collapse
Affiliation(s)
- Mario Perl
- Department of Traumatology, Hand and Reconstructive Surgery, University of Ulm Medical School, Ulm, Germany
| | | | | | | | | |
Collapse
|
60
|
Arndt PG, Strahan B, Wang Y, Long C, Horiuchi K, Walcheck B. Leukocyte ADAM17 regulates acute pulmonary inflammation. PLoS One 2011; 6:e19938. [PMID: 21603616 PMCID: PMC3095620 DOI: 10.1371/journal.pone.0019938] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Accepted: 04/15/2011] [Indexed: 11/18/2022] Open
Abstract
The transmembrane protease ADAM17 regulates the release and density of various leukocyte cell surface proteins that modulate inflammation, including L-selectin, TNF-α, and IL-6R. At this time, its in vivo substrates and role in pulmonary inflammation have not been directly examined. Using conditional ADAM17 knock-out mice, we investigated leukocyte ADAM17 in acute lung inflammation. Alveolar TNF-α levels were significantly reduced (>95%) in ADAM17-null mice following LPS administration, as was the shedding of L-selectin, a neutrophil-expressed adhesion molecule. Alveolar IL-6R levels, however, were reduced by only ≈25% in ADAM17-null mice, indicating that ADAM17 is not its primary sheddase in our model. Neutrophil infiltration into the alveolar compartment is a key event in the pathophysiology of acute airway inflammation. Following LPS inhalation, alveolar neutrophil levels and lung inflammation in ADAM17-null mice were overall reduced when compared to control mice. Interestingly, however, neutrophil recruitment to the alveolar compartment occurred earlier in ADAM17-null mice after exposure to LPS. This decrease in alveolar neutrophil recruitment in ADAM17-null mice was accompanied by significantly diminished alveolar levels of the neutrophil-tropic chemokines CXCL1 and CXCL5. Altogether, our study suggests that leukocyte ADAM17 promotes inflammation in the lung, and thus this sheddase may be a potential target in the design of pharmacologic therapies for acute lung injury.
Collapse
Affiliation(s)
- Patrick G. Arndt
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Brian Strahan
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Yue Wang
- Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Chunmei Long
- Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
| | - Keisuke Horiuchi
- Department of Orthopedic Surgery, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Bruce Walcheck
- Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
61
|
Fudala R, Allen TC, Krupa A, Cagle PT, Nash S, Gryczynski Z, Gryczynski I, Kurdowska AK. Increased Levels of Nuclear Factor κB and Fos-Related Antigen 1 in Lung Tissues From Patients With Acute Respiratory Distress Syndrome. Arch Pathol Lab Med 2011; 135:647-54. [DOI: 10.5858/2009-0660-oar1.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Abstract
Context.—Both nuclear factor κB and Fos-related antigen 1 have been implicated in the pathogenesis of inflammatory lung diseases, including acute lung injury/acute respiratory distress syndrome.
Objective.—To evaluate lung tissues from patients with acute respiratory distress syndrome for presence of nuclear factor κB and Fos-related antigen 1.
Design.—Lung tissue sections from 5 patients with acute respiratory distress syndrome and sections of normal lung tissues of 4 patients were stained with antibodies against epithelial cell marker (surfactant protein B) and nuclear factor κB or Fos-related antigen 1. Samples were analyzed using confocal laser microscopy.
Results.—We have detected significantly increased levels of activated nuclear factor κB and Fos-related antigen 1 in lung tissues from patients with acute respiratory distress syndrome compared with control tissues, suggesting that these transcription factors undergo activation in lungs of patients suffering from acute respiratory distress syndrome.
Conclusions.—Our data demonstrate that activated nuclear factor κB and Fos-related antigen 1 are elevated in epithelial cells in lung tissues of patients with acute respiratory distress syndrome.
Collapse
|
62
|
Martin EL, Ranieri VM. Phosphorylation mechanisms in intensive care medicine. Intensive Care Med 2011; 37:7-18. [PMID: 20820992 DOI: 10.1007/s00134-010-2023-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Accepted: 07/18/2010] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The phosphorylation states of proteins, lipids, carbohydrates, amino acids, and nucleotides control the mechanisms behind nearly all cellular functions. Therefore, not surprisingly, recent findings have shown that alterations in these phosphorylation pathways play a central role in the development and progression of many disease states. This review provides a brief summary of the function and activity of various phosphorylation mechanisms, outlines some of the major phosphorylation signaling cascades, and describes the role of these phosphorylation mechanisms in intensive care medicine. METHODS This article will comprise a comprehensive review of the literature in the context of intensive care medicine. Specifically, we will discuss the involvement of phosphorylation in the pathogenesis, diagnosis, and treatment of heart failure, myocardial infarction, stroke, respiratory failure, ventilation-induced lung injury, traumatic brain injury, acute organ failure, systemic sepsis, and shock. CONCLUSION Phosphorylation mechanisms clearly play an important role in many pathologies and treatment strategies of intensive care and therefore further understanding of these mechanisms may lead to the development of novel therapies and improved patient care.
Collapse
Affiliation(s)
- Erica L Martin
- Department of Anaesthesiology and Critical Care, University of Turin, Ospedale S. Giovanni Battista-Molinette, Corso A.M. Dogliotti 14, 10126 Turin, Italy
| | | |
Collapse
|
63
|
Yang Y, Li Q, Deng Z, Zhang Z, Xu J, Qian G, Wang G. Protection from lipopolysaccharide-induced pulmonary microvascular endothelial cell injury by activation of hedgehog signaling pathway. Mol Biol Rep 2010; 38:3615-22. [PMID: 21110116 DOI: 10.1007/s11033-010-0473-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2010] [Accepted: 11/09/2010] [Indexed: 02/06/2023]
Abstract
Pulmonary microvascular endothelial cells (PMVECs) are critically involved in the pathogenesis of acute lung injury. Hedgehog signaling pathway plays a fundamental role in embryonic development as well as adult morphogenesis and carcinogenesis. As the priming protein of hedgehog signaling pathway, sonic hedgehog (Shh) may recently be advantage for decreasing endothelial injury and promoting the repair of endothelial barrier function. To investigate the expression and role of hedgehog signal pathway in PMVECs injured by lipopolysaccharide (LPS), cells were divided into six groups: control group, LPS group, rhShh group, LPS+rhShh group, rhShh+cyclopamine group, and LPS+rhShh+cyclopamine group. Real time RT-PCR and Western blotting were used to detect the mRNA and protein expression of hedgehog signal molecules including Shh, Patched-1 (Ptc-1) and Gli1 in nucleus. The activity of PMVECs was examined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. In this study, we found that Shh, Ptch1, and Gli1 were expressed in rat PMVECs and their expression decreased when cells were treated by LPS. In the other hand, LPS inhibited the activity of rat PMVECs and caused the cells injury. Activation of Hedgehog signaling pathway by Shh could elevate the activity of PMVECs with pretreatment by LPS. Therefore, hedgehog signaling pathway should play a protective role on injury PMVECs by LPS.
Collapse
Affiliation(s)
- Yu Yang
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | | | | | | | | | | | | |
Collapse
|
64
|
Villar J, Cabrera N, Casula M, Flores C, Valladares F, Muros M, Blanch L, Slutsky AS, Kacmarek RM. Mechanical ventilation modulates Toll-like receptor signaling pathway in a sepsis-induced lung injury model. Intensive Care Med 2010; 36:1049-57. [PMID: 20397011 DOI: 10.1007/s00134-010-1799-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2009] [Accepted: 01/22/2010] [Indexed: 11/26/2022]
Abstract
BACKGROUND Experimental and clinical studies on sepsis have demonstrated activation of the innate immune response following the initial host-bacterial interaction. In addition, mechanical ventilation (MV) can induce a pulmonary inflammatory response. How these two responses interact when present simultaneously remains to be elucidated. We hypothesized that MV modulates innate host response during sepsis by influencing Toll-like receptor (TLR) signaling. DESIGN Prospective, randomized, controlled animal study. SUBJECTS Male, septic Sprague-Dawley rats. INTERVENTIONS Sepsis was induced by cecal ligation and perforation. At 18 h, surviving animals had the cecum removed and were randomized to spontaneous breathing or two strategies of MV for 4 h: high (20 ml/kg) tidal volume (V (T)) with no positive end-expiratory pressure (PEEP) versus low V (T) (6 ml/kg) plus 10 cmH(2)O PEEP. MEASUREMENTS AND MAIN RESULTS Histological evaluation, TLR-2, TLR-4, inhibitory kappaB alpha (IkappaBalpha), interleukin-1 receptor-associated kinase-3 (IRAK-3) gene expression, protein levels and immunohistochemical lung localization, inflammatory cytokines gene expression, and protein serum concentrations were analyzed. MV with low V (T) plus PEEP attenuated sepsis-associated TLR-4 activation, and produced a significant decrease of IRAK-3 gene expression and protein levels, a significant increase of IkappaBalpha, and a decrease in lung gene expression and serum levels of cytokines. High-V (T) MV caused a significant increase of TLR-4 and IRAK-3 protein levels, lung and systemic cytokines, and mortality, and a significant decrease of IkappaBalpha. CONCLUSIONS Our findings suggest a novel mechanism that could partially explain how MV modulates the innate immune response in the lung by interfering with cellular signaling pathways that are activated in response to pathogens.
Collapse
Affiliation(s)
- Jesús Villar
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Kasten KR, Goetzman HS, Reid MR, Rasper AM, Adediran SG, Robinson CT, Cave CM, Solomkin JS, Lentsch AB, Johannigman JA, Caldwell CC. Divergent adaptive and innate immunological responses are observed in humans following blunt trauma. BMC Immunol 2010; 11:4. [PMID: 20100328 PMCID: PMC2823662 DOI: 10.1186/1471-2172-11-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Accepted: 01/25/2010] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The immune response to trauma has traditionally been modeled to consist of the systemic inflammatory response syndrome (SIRS) followed by the compensatory anti-inflammatory response syndrome (CARS). We investigated these responses in a homogenous cohort of male, severe blunt trauma patients admitted to a University Hospital surgical intensive care unit (SICU). After obtaining consent, peripheral blood was drawn up to 96 hours following injury. The enumeration and functionality of both myeloid and lymphocyte cell populations were determined. RESULTS Neutrophil numbers were observed to be elevated in trauma patients as compared to healthy controls. Further, neutrophils isolated from trauma patients had increased raft formation and phospho-Akt. Consistent with this, the neutrophils had increased oxidative burst compared to healthy controls. In direct contrast, blood from trauma patients contained decreased naïve T cell numbers. Upon activation with a T cell specific mitogen, trauma patient T cells produced less IFN-gamma as compared to those from healthy controls. Consistent with these results, upon activation, trauma patient T cells were observed to have decreased T cell receptor mediated signaling. CONCLUSIONS These results suggest that following trauma, there are concurrent and divergent immunological responses. These consist of a hyper-inflammatory response by the innate arm of the immune system concurrent with a hypo-inflammatory response by the adaptive arm.
Collapse
Affiliation(s)
- Kevin R Kasten
- Division of Research, Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267-0558, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Chen CY, Yang KY, Chen MY, Chen HY, Lin MT, Lee YC, Perng RP, Hsieh SL, Yang PC, Chou TY. Decoy receptor 3 levels in peripheral blood predict outcomes of acute respiratory distress syndrome. Am J Respir Crit Care Med 2009; 180:751-60. [PMID: 19644047 DOI: 10.1164/rccm.200902-0222oc] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
RATIONALE Acute respiratory distress syndrome (ARDS), a serious inflammatory reaction to acute lung injury, is associated with high mortality rates. Decoy receptor (DcR) 3 is a soluble protein with immunomodulatory effects. Biomarkers that reliably predict outcomes in ARDS are not currently available. OBJECTIVES Comparing DcR3 with the Acute Physiology and Chronic Health Evaluation (APACHE) II scores and three other plasma markers to explore the association of DcR3 and the clinical outcome in ARDS. METHODS Eighty-eight patients with ARDS were studied. Baseline APACHE II scores and clinical data were recorded. Plasma levels of DcR3, soluble triggering receptor expressed on myeloid cells (sTREM)-1, tumor necrosis factor (TNF)-alpha, and IL-6 were measured on Day 1 and later time points, and correlated with the survival status on Day 28 after the onset of ARDS. For validation, 59 patients with ARDS from another medical center were studied. MEASUREMENTS AND MAIN RESULTS Among the biomarkers evaluated, only DcR3 discriminated the survivors and nonsurvivors at all time points in the first week of ARDS. DcR3 independently associated with and best predicted the 28-day mortality of patients with ARDS. Plasma DcR3 levels most correlated to multiple-organ dysfunction and ventilator dependence. Compared with survivors, the nonsurvivors had higher DcR3 levels regardless of the APACHE II scores. Kaplan-Meier survival analysis showed higher mortality in patients with ARDS with higher DcR3 levels. The outcome prediction of patients with ARDS by plasma DcR3 levels was recapitulated by the validation cohort. CONCLUSIONS High plasma DcR3 levels correlate with development of multiple-organ dysfunction and independently predict the 28-day mortality in patients with ARDS.
Collapse
Affiliation(s)
- Cheng-Yu Chen
- Institute of Clinical Medicine, National Yang-Ming University, Taipei
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Cleavage of SIgA by gram negative respiratory pathogens enhance neutrophil inflammatory potential. ACTA ACUST UNITED AC 2009; 66:1336-42; discussion 1342. [PMID: 19430236 DOI: 10.1097/ta.0b013e31819dc577] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Secretory immunoglobulin A (SIgA), the principle immune defense at respiratory and other mucosal sites in the body, is highly dependant on its molecular structure for effective antibody function. Previous studies have demonstrated that gram-negative but not gram-positive isolates from patients with nosocomial pneumonia have IgA protease activity that contributes to the development of infection. We postulate that SIgA cleavage by bacteria would also affect anti-inflammatory properties of IgA and studied this in vitro. METHODS Sterile filtrates obtained from Pseudomonas, Acinetobacter, and methicillin resistant Staphylococcus aureus (MRSA) held in culture with SIgA were used to challenge polymorphonuclear neutrophils (PMNs) obtained from healthy volunteers. In a second group of experiments, blood monocytes were incubated with lipopolysaccharide (LPS) or LPS + IgA, and the resulting culture supernatants was used to stimulate PMNs in vitro. RESULTS LPS-stimulated monocytes increased CD11b expression, O2-generation and elastase release by PMNs. Secretory IgA but not IgG abrogated this response. Cleavage of SIgA by the gram-negative respiratory isolates, Pseudomonas aeruginosa and Acinetobacter baumanii also led to the loss of cellular effector function noted with intact SIgA. Additionally, PMN cytotoxic potential was similar to that noted with PMNs treated with supernatant from LPS-stimulated monocytes. CONCLUSION IgA cleavage by gram-negative respiratory isolates may lead to the development of pneumonia and the subsequent severity of the infection as a result of uncontrolled inflammatory responses by the host.
Collapse
|
68
|
Bonville CA, Percopo CM, Dyer KD, Gao J, Prussin C, Foster B, Rosenberg HF, Domachowske JB. Interferon-gamma coordinates CCL3-mediated neutrophil recruitment in vivo. BMC Immunol 2009; 10:14. [PMID: 19298652 PMCID: PMC2662797 DOI: 10.1186/1471-2172-10-14] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2008] [Accepted: 03/19/2009] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND We have shown previously that acute infection with the respiratory pathogen, pneumonia virus of mice (PVM), results in local production of the proinflammatory chemokine, CCL3, and that neutrophil recruitment in response to PVM infection is reduced dramatically in CCL3 -/- mice. RESULTS In this work, we demonstrate that CCL3-mediated neutrophil recruitment is coordinated by interferon-gamma (IFNgamma). Neutrophil recruitment in response to PVM infection was diminished five-fold in IFNgamma receptor gene-deleted mice, although neutrophils from IFNgammaR -/- mice expressed transcripts for the CCL3 receptor, CCR1 and responded functionally to CCL3 ex vivo. Similarly, in the absence of PVM infection, CCL3 overexpression alone could not elicit neutrophil recruitment in the absence of IFNgamma. Interestingly, although supplemental IFNgamma restored neutrophil recruitment and resulted in a sustained weight loss among CCL3-overexpressing IFNgamma -/- mice, CCL3-mediated neutrophil recruitment alone did not result in the pulmonary edema or respiratory failure characteristic of severe viral infection, suggesting that CCL3 and IFN-gamma together are sufficient to promote neutrophil recruitment but not pathologic activation. CONCLUSION Our findings reveal a heretofore unrecognized hierarchical interaction between the IFNgamma and CCL3, which demonstrate that IFNgamma is crucial for CCL3-mediated neutrophil recruitment in vivo.
Collapse
MESH Headings
- Animals
- Cell Movement/immunology
- Chemokine CCL3/genetics
- Chemokine CCL3/immunology
- Chemokine CCL3/metabolism
- Gene Expression Profiling
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/pathology
- Lung/immunology
- Lung/metabolism
- Lung/pathology
- Lung/virology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Murine pneumonia virus/immunology
- Murine pneumonia virus/pathogenicity
- Neutrophils/immunology
- Neutrophils/metabolism
- Neutrophils/pathology
- Pneumovirus Infections/genetics
- Pneumovirus Infections/immunology
- Pneumovirus Infections/physiopathology
- Pulmonary Edema
- Receptors, CCR1/genetics
- Receptors, CCR1/immunology
- Receptors, CCR1/metabolism
- Receptors, Interferon/genetics
- Receptors, Interferon/immunology
- Receptors, Interferon/metabolism
- Respiratory Insufficiency
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- Interferon gamma Receptor
Collapse
Affiliation(s)
| | - Caroline M Percopo
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kimberly D Dyer
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jiliang Gao
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Calman Prussin
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Barbara Foster
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
- Proteus Technologies, 133 National Business Parkway, Suite 150, Annapolis Junction, Maryland, 20701, USA
| | - Helene F Rosenberg
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
69
|
Zmijewski JW, Lorne E, Banerjee S, Abraham E. Participation of mitochondrial respiratory complex III in neutrophil activation and lung injury. Am J Physiol Lung Cell Mol Physiol 2009; 296:L624-34. [PMID: 19168575 DOI: 10.1152/ajplung.90522.2008] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Reactive oxygen species (ROS) produced during mitochondrial activity participate in the regulation of intracellular signaling pathways. However, there is only limited information concerning the role that ROS derived from the mitochondrial respiratory chain play in modulating neutrophil activity and participation in acute inflammatory processes. Because mitochondrial complex III is a major site of ROS formation, we examined whether selective complex III inhibition, through exposure of neutrophils to myxothiazol or antimycin A, would affect LPS-induced activation. Culture of neutrophils with antimycin A or myxothiazol resulted in increased intracellular levels of ROS, including superoxide and hydrogen peroxide (H(2)O(2)). Inhibition of complex III activity reduced LPS-induced degradation of IkappaB-alpha, nuclear accumulation of NF-kappaB, and proinflammatory cytokine production. The effects of antimycin A or myxothiazol appeared to be dependent on generation of H(2)O(2) since addition of pegylated catalase to neutrophils restored LPS-mediated IkappaB-alpha degradation and production of proinflammatory cytokines. Administration of myxothiazol to mice resulted in diminished mitochondrial complex III activity in the lungs and decreased severity of LPS-induced lung injury. These results indicate that inhibition of mitochondrial complex III diminishes Toll-like receptor 4-induced neutrophil activation through a mechanism dependent on H(2)O(2) generation and also reduces the severity of lung injury due to LPS exposure, a pathophysiologic process in which neutrophils play a major role.
Collapse
Affiliation(s)
- Jaroslaw W Zmijewski
- Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294-0012, USA
| | | | | | | |
Collapse
|
70
|
Zmijewski JW, Lorne E, Zhao X, Tsuruta Y, Sha Y, Liu G, Abraham E. Antiinflammatory effects of hydrogen peroxide in neutrophil activation and acute lung injury. Am J Respir Crit Care Med 2009; 179:694-704. [PMID: 19151196 DOI: 10.1164/rccm.200806-851oc] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
RATIONALE Although reactive oxygen species (ROS) are generally considered to be proinflammatory and to contribute to cellular and organ dysfunction when present in excessive amounts, there is evidence that specific ROS, particularly hydrogen peroxide (H(2)O(2)), may have antiinflammatory properties. OBJECTIVES To address the role that increases in intracellular H(2)O(2) may play in acute inflammatory processes, we examined the effects of catalase inhibition or the absence of catalase on LPS-induced inflammatory responses. METHODS Neutrophils from control or acatalasemic mice, or control neutrophils incubated with the catalase inhibitor aminotriazole, were treated with LPS, and levels of reactive oxygen species, proteasomal activity, NF-kappaB activation, and proinflammatory cytokine expression were measured. Acute lung injury (ALI) was produced by intratracheal injection of LPS into control, acatalasemic-, or aminotriazole-treated mice. MEASUREMENTS AND MAIN RESULTS Intracellular levels of H(2)O(2) were increased in acatalasemic neutrophils and in neutrophils exposed to aminotriazole. Compared with LPS-stimulated neutrophils from control mice, neutrophils from acatalasemic mice or neutrophils treated with aminotriazole demonstrated reduced 20S and 26S proteasomal activity, IkappaB-alpha degradation, NF-kappaB nuclear accumulation, and production of the proinflammatory cytokines TNF-alpha and macrophage inhibitory protein (MIP)-2. The severity of LPS-induced ALI was less in acatalasemic mice and in mice treated with aminotriazole as compared with that found in control mice. CONCLUSIONS These results indicate that H(2)O(2) has antiinflammatory effects on neutrophil activation and inflammatory processes, such as ALI, in which activated neutrophils play a major role.
Collapse
Affiliation(s)
- Jaroslaw W Zmijewski
- Department of Medicine, University of Alabama at Birmingham School of Medicine, 1530 3rd Avenue S, Birmingham, AL 35294-0012, USA
| | | | | | | | | | | | | |
Collapse
|
71
|
Lorne E, Zhao X, Zmijewski JW, Liu G, Park YJ, Tsuruta Y, Abraham E. Participation of mammalian target of rapamycin complex 1 in Toll-like receptor 2- and 4-induced neutrophil activation and acute lung injury. Am J Respir Cell Mol Biol 2009; 41:237-45. [PMID: 19131641 DOI: 10.1165/rcmb.2008-0290oc] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
mTOR complex 1 (mTORC1) plays a central role in cell growth and cellular responses to metabolic stress. Although mTORC1 has been shown to be activated after Toll-like receptor (TLR)-4 engagement, there is little information concerning the role that mTORC1 may play in modulating neutrophil function and neutrophil-dependent inflammatory events, such as acute lung injury. To examine these issues, we determined the effects of rapamycin-induced inhibition of mTORC1 on TLR2- and TLR4-induced neutrophil activation. mTORC1 was dose- and time-dependently activated in murine bone marrow neutrophils cultured with the TLR4 ligand, LPS, or the TLR2 ligand, Pam(3) Cys-Ser-(Lys)(4) (PAM). Incubation of PAM- or LPS-stimulated neutrophils with rapamycin inhibited expression of TNF-alpha and IL-6, but not IkappaB-alpha degradation or nuclear translocation of NF-kappaB. Exposure of PAM or LPS-stimulated neutrophils to rapamycin inhibited phosphorylation of serine 276 in the NF-kappaB p65 subunit, a phosphorylation event required for optimal transcriptional activity of NF-kappaB. Rapamycin pretreatment inhibited PAM- or LPS-induced mTORC1 activation in the lungs. Administration of rapamycin also decreased the severity of lung injury after intratracheal LPS or PAM administration, as determined by diminished neutrophil accumulation in the lungs, reduced interstitial pulmonary edema, and diminished levels of TNF-alpha and IL-6 in bronchoalveolar lavage fluid. These results indicate that mTORC1 activation is essential in TLR2- and TLR4-induced neutrophil activation, as well as in the development and severity of acute lung injury.
Collapse
Affiliation(s)
- Emmanuel Lorne
- Department of Medicine, University of Alabama, Birmingham, AL, USA
| | | | | | | | | | | | | |
Collapse
|
72
|
Krupa A, Fudala R, Stankowska D, Loyd T, Allen TC, Matthay MA, Gryczynski Z, Gryczynski I, Mettikolla YV, Kurdowska AK. Anti-chemokine autoantibody:chemokine immune complexes activate endothelial cells via IgG receptors. Am J Respir Cell Mol Biol 2008; 41:155-69. [PMID: 19109244 DOI: 10.1165/rcmb.2008-0183oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Our previous studies revealed that the presence in lung fluids of anti-IL-8 autoantibody:IL-8 immune complexes is an important prognostic indicator for the development and outcome of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). Anti-IL-8:IL-8 complexes purified from lung edema fluids trigger chemotaxis of neutrophils, induce activation of these cells, and regulate their apoptosis, all via IgG receptor, FcgammaRIIa. Importantly, increased levels of FcgammaRIIa are present in lungs of patients with ARDS, where FcgammaRIIa is partially associated with anti-IL-8:IL-8 complexes. In the current study, we demonstrate the ability of anti-IL-8:IL-8 complexes to promote an inflammatory phenotype of human umbilical vein endothelial cells via interaction with FcgammaRIIa. Human umbilical vein endothelial cells cultured in the presence of the complexes become activated, as shown by increased phosphorylation of ERK, JNK, and Akt, and augmented nuclear translocation of NF-kappaB. Anti-IL-8:IL-8 complexes also up-regulate expression of intracellular adhesion molecule (ICAM)-1 on the cell surface. Furthermore, we detected increased levels of ICAM-1 on lung endothelial cells from mice in which lung injury was induced by generating immune complexes in alveolar spaces. On the other hand, ICAM-1 expression was unchanged in lungs of gamma chain-deficient mice, lacking receptors that interact with immune complexes. Moreover, in lung tissues from patients with ARDS, anti-IL-8:IL-8 complexes were associated with endothelial cells that expressed higher levels of ICAM-1. Our current findings implicate that anti-chemokine autoantibody:chemokine immune complexes, such as IL-8:IL-8 complexes, may contribute to pathogenesis of lung inflammation by inducing activation of endothelial cells through engagement of IgG receptors.
Collapse
Affiliation(s)
- Agnieszka Krupa
- Department of Biochemistry, University of Texas Health Center, 11,937 U.S. Highway 271, Tyler, Texas 75708-3154, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Perl M, Lomas-Neira J, Chung CS, Ayala A. Epithelial cell apoptosis and neutrophil recruitment in acute lung injury-a unifying hypothesis? What we have learned from small interfering RNAs. Mol Med 2008; 14:465-75. [PMID: 18368145 PMCID: PMC2274893 DOI: 10.2119/2008-00011.perl] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 03/17/2008] [Indexed: 01/11/2023] Open
Abstract
In spite of protective ventilatory strategies, Acute Lung Injury (ALI) remains associated with high morbidity and mortality. One reason for the lack of therapeutic options might be that ALI is a co-morbid event associated with a diverse family of diseases and, thus, may be the result of distinct pathological processes. Among them, activated neutrophil- (PMN-) induced tissue injury and epithelial cell apoptosis mediated lung damage represent two potentially important candidate pathomechanisms that have been put forward. Several approaches have been undertaken to test these hypotheses, with substantial success in the treatment of experimental forms of ALI. With this in mind, we will summarize these two current hypotheses of ALI briefly, emphasizing the role of apoptosis in regulating PMN and/or lung epithelial cell responses. In addition, the contribution that Fas-mediated inflammation may play as a potential biological link between lung cell apoptosis and PMN recruitment will be considered, as well as the in vivo application of small interfering RNA (siRNA) as a novel approach to the inhibition of ALI and its therapeutic implications.
Collapse
Affiliation(s)
- Mario Perl
- Department of Traumatology, Hand- and Reconstructive Surgery, University of Ulm Medical School, Ulm, Germany
| | | | | | | |
Collapse
|
74
|
Zhao X, Zmijewski JW, Lorne E, Liu G, Park YJ, Tsuruta Y, Abraham E. Activation of AMPK attenuates neutrophil proinflammatory activity and decreases the severity of acute lung injury. Am J Physiol Lung Cell Mol Physiol 2008; 295:L497-504. [PMID: 18586954 DOI: 10.1152/ajplung.90210.2008] [Citation(s) in RCA: 249] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is activated by increases in the intracellular AMP-to-ATP ratio and plays a central role in cellular responses to metabolic stress. Although activation of AMPK has been shown to have anti-inflammatory effects, there is little information concerning the role that AMPK may play in modulating neutrophil function and neutrophil-dependent inflammatory events, such as acute lung injury. To examine these issues, we determined the effects of pharmacological activators of AMPK, 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR) and barberine, on Toll-like receptor 4 (TLR4)-induced neutrophil activation. AICAR and barberine dose-dependently activated AMPK in murine bone marrow neutrophils. Exposure of LPS-stimulated neutrophils to AICAR or barberine inhibited release of TNF-alpha and IL-6, as well as degradation of IkappaBalpha and nuclear translocation of NF-kappaB, compared with findings in neutrophil cultures that contained LPS without AICAR or barberine. Administration of AICAR to mice resulted in activation of AMPK in the lungs and was associated with decreased severity of LPS-induced lung injury, as determined by diminished neutrophil accumulation in the lungs, reduced interstitial pulmonary edema, and diminished levels of TNF-alpha and IL-6 in bronchoalveolar lavage fluid. These results suggest that AMPK activation reduces TLR4-induced neutrophil activation and diminishes the severity of neutrophil-driven proinflammatory processes, including acute lung injury.
Collapse
Affiliation(s)
- Xia Zhao
- Dept. of Medicine, Univ. of Alabama at Birmingham School of Medicine, BDB 420, 1530 3rd Ave. S, Birmingham, AL 35294-0012, USA
| | | | | | | | | | | | | |
Collapse
|
75
|
Lorne E, Zmijewski JW, Zhao X, Liu G, Tsuruta Y, Park YJ, Dupont H, Abraham E. Role of extracellular superoxide in neutrophil activation: interactions between xanthine oxidase and TLR4 induce proinflammatory cytokine production. Am J Physiol Cell Physiol 2008; 294:C985-93. [DOI: 10.1152/ajpcell.00454.2007] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Reactive oxygen species (ROS) contribute to neutrophil activation and the development of acute inflammatory processes in which neutrophils play a central role. However, there is only limited information concerning the mechanisms through which extracellular ROS, and particularly cell membrane-impermeable species, such as superoxide, enhance the proinflammatory properties of neutrophils. To address this issue, neutrophils were exposed to superoxide generating combinations of xanthine oxidase and hypoxanthine or lumazine. Extracellular superoxide generation induced nuclear translocation of nuclear factor-κB (NF-κB) and increased neutrophil production of the NF-κB-dependent cytokines tumor necrosis factor-α (TNF-α) and macrophage inhibitory protein-2 (MIP-2). In contrast, there were no changes in TNF-α or MIP-2 expression when neutrophils lacking Toll-like receptor-4 (TLR4) were exposed to extracellular superoxide. Immunoprecipitation, confocal microscopy, and fluorescence resonance energy transfer (FRET) studies demonstrated association between TLR4 and xanthine oxidase. Exposure of neutrophils to heparin attenuated binding of xanthine oxidase to the cell surface as well as interactions with TLR4. Heparin also decreased xanthine oxidase-induced nuclear translocation of NF-κB as well as production of proinflammatory cytokines. These results demonstrate that extracellular superoxide has proinflammatory effects on neutrophils, predominantly acting through an TLR4-dependent mechanism that enhances nuclear translocation of NF-κB and increases expression of NF-κB-dependent cytokines.
Collapse
|
76
|
Dolinay T, Wu W, Kaminski N, Ifedigbo E, Kaynar AM, Szilasi M, Watkins SC, Ryter SW, Hoetzel A, Choi AMK. Mitogen-activated protein kinases regulate susceptibility to ventilator-induced lung injury. PLoS One 2008; 3:e1601. [PMID: 18270588 PMCID: PMC2223071 DOI: 10.1371/journal.pone.0001601] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Accepted: 01/17/2008] [Indexed: 01/09/2023] Open
Abstract
Background Mechanical ventilation causes ventilator-induced lung injury in animals and humans. Mitogen-activated protein kinases have been implicated in ventilator-induced lung injury though their functional significance remains incomplete. We characterize the role of p38 mitogen-activated protein kinase/mitogen activated protein kinase kinase-3 and c-Jun-NH2-terminal kinase-1 in ventilator-induced lung injury and investigate novel independent mechanisms contributing to lung injury during mechanical ventilation. Methodology and Principle Findings C57/BL6 wild-type mice and mice genetically deleted for mitogen-activated protein kinase kinase-3 (mkk-3−/−) or c-Jun-NH2-terminal kinase-1 (jnk1−/−) were ventilated, and lung injury parameters were assessed. We demonstrate that mkk3−/− or jnk1−/− mice displayed significantly reduced inflammatory lung injury and apoptosis relative to wild-type mice. Since jnk1−/− mice were highly resistant to ventilator-induced lung injury, we performed comprehensive gene expression profiling of ventilated wild-type or jnk1−/− mice to identify novel candidate genes which may play critical roles in the pathogenesis of ventilator-induced lung injury. Microarray analysis revealed many novel genes differentially expressed by ventilation including matrix metalloproteinase-8 (MMP8) and GADD45α. Functional characterization of MMP8 revealed that mmp8−/− mice were sensitized to ventilator-induced lung injury with increased lung vascular permeability. Conclusions We demonstrate that mitogen-activated protein kinase pathways mediate inflammatory lung injury during ventilator-induced lung injury. C-Jun-NH2-terminal kinase was also involved in alveolo-capillary leakage and edema formation, whereas MMP8 inhibited alveolo-capillary protein leakage.
Collapse
Affiliation(s)
- Tamás Dolinay
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Unites States of America
- Department of Pulmonology, University of Debrecen Medical and Health Science Center, Debrecen, Hungary
| | - Wei Wu
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Unites States of America
| | - Naftali Kaminski
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Unites States of America
| | - Emeka Ifedigbo
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Unites States of America
| | - A. Murat Kaynar
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mária Szilasi
- Department of Pulmonology, University of Debrecen Medical and Health Science Center, Debrecen, Hungary
| | - Simon C. Watkins
- Department of Cell Biology and Physiology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stefan W. Ryter
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Unites States of America
| | - Alexander Hoetzel
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Unites States of America
- Department of Anesthesiology and Critical Care Medicine, University of Freiburg, Freiburg, Germany
| | - Augustine M. K. Choi
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, Unites States of America
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
77
|
Anti-interleukin-8 autoantibody:interleukin-8 immune complexes in acute lung injury/acute respiratory distress syndrome. Clin Sci (Lond) 2008; 114:403-12. [DOI: 10.1042/cs20070272] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
ALI/ARDS (acute lung injury/acute respiratory distress syndrome) is a severe inflammatory lung disease associated with very high mortality. Importantly, no effective therapy has been developed to date for ALI/ARDS. Neutrophils have been implicated in the pathogenesis of ALI/ARDS, and IL-8 (interleukin-8) has been identified as the main chemotactic factor for neutrophils in lung fluids of patients with ALI/ARDS. Significantly, studies from our laboratory have revealed the presence of anti-IL-8 autoantibody:IL-8 immune complexes in lung fluids from patients with ALI/ARDS. Autoantibodies to several cytokines, including IL-8, have been found in human plasma and other tissues. The function of anticytokine autoantibodies is far from clear; however, in some instances, it has been suggested that such autoantibodies may contribute to the pathogenesis of variety of human diseases. In addition, many of these autoantibodies can form immune complexes with target cytokines. Furthermore, immune complexes consisting of anti-IL-8 autoantibodies and IL-8 are very stable due to the high affinity of autoantibodies against IL-8. These complexes are present in various human tissues, including the lung, as they have been detected in lung fluids from patients with ALI/ARDS. In this review, the significance of the latter findings are explored, and the possible involvement of anti-IL-8 autoantibody:IL-8 immune complexes in pathogenesis of ALI/ARDS is discussed.
Collapse
|
78
|
Lorenz E, Muhlebach MS, Tessier PA, Alexis NE, Duncan Hite R, Seeds MC, Peden DB, Meredith W. Different expression ratio of S100A8/A9 and S100A12 in acute and chronic lung diseases. Respir Med 2007; 102:567-73. [PMID: 18164192 DOI: 10.1016/j.rmed.2007.11.011] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2006] [Revised: 08/28/2007] [Accepted: 11/19/2007] [Indexed: 10/22/2022]
Abstract
Calgranulins are a family of powerful chemoattractants, which have been implicated as biomarkers in inflammatory diseases. To determine how different respiratory diseases affect the expression of calgranulins, we measured the expression of S100A8/A9 and S100A12 in bronchoalveolar lavage fluid (BALF) of acute respiratory distress syndrome (ARDS) patients and healthy volunteers by ELISA. Analysis of calgranulin expression revealed a high level of S100A12 in the lavages of patients suffering from ARDS compared to controls (p<0.001). Based on the hypothesis that the increased expression of S100A12 relative to the S100A8/A9 heterodimer was a characteristic of respiratory diseases with neutrophilic inflammation, we measured calgranulin expression in BALF of cystic fibrosis (CF) patients. Despite similarly elevated levels of S100A8/A9, S100A12 was significantly higher in ARDS compared to CF BALF (p<0.001). The differential expression of calgranulins was unique for inflammatory markers, as an array of cytokines did not differ between CF and ARDS patients. Since ARDS is an acute event and CF a chronic inflammation with acute exacerbations, we compared calgranulin expression in sputum obtained from CF and patients with chronic obstructive lung disease (COPD). Levels of S100A12 and S100A8/9 were elevated in CF sputum compared to COPD sputum, but the ratio of S100A12 to S100A8/A9 was similar in COPD and CF and reflected more closely than seen in healthy controls. The results indicate that the regulation of human calgranulin expression and the ratio of S100A8/A9 to S100A12 may provide important insights in the mechanism of respiratory inflammation.
Collapse
Affiliation(s)
- Eva Lorenz
- Department of Rheumatology, TARC University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7280, USA.
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Liu G, Tsuruta Y, Gao Z, Park YJ, Abraham E. Variant IL-1 receptor-associated kinase-1 mediates increased NF-kappa B activity. THE JOURNAL OF IMMUNOLOGY 2007; 179:4125-34. [PMID: 17785851 DOI: 10.4049/jimmunol.179.6.4125] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-1R-associated kinase (IRAK)-1 is a critical mediator of TLR/IL-1R-induced activation of the transcription factor NF-kappaB. We previously described that a commonly occurring IRAK-1 variant haplotype, containing amino acid changes from serine to phenylalanine at position 196 and from leucine to serine at position 532, is associated with increased activation of NF-kappaB in LPS-stimulated neutrophils from patients with sepsis-induced acute lung injury and also higher mortality and more severe clinical outcomes in such patients. To investigate the underlying molecular mechanisms, we examined the ability of wild-type and variant IRAK-1 to modulate NF-kappaB activation. We found increased NF-kappaB transcriptional activity and expression of NF-kappaB-dependent proinflammatory cytokines in IL-1beta-stimulated IRAK-1-deficient cells transfected with variant IRAK-1 as compared with IRAK-1 wild type. IkappaB-alpha degradation was faster and p65 phosphorylation more prolonged after IL-1beta stimulation in cells expressing the IRAK-1 variant. However, IL-1-induced activation of MAPKs and nuclear translocation of NF-kappaB are comparable in both IRAK-1 variant- and IRAK-1 wild-type-expressing cells. Autophosphorylation of the IRAK-1 variant is greater than that found with wild-type IRAK-1. Additionally, variant IRAK-1 has greater interaction with TNFR-associated factor 6 than does wild-type IRAK-1. The enhanced activity of variant IRAK-1 appeared to be due to the alteration at aa 532, with only minimal effects being associated with change at aa 196. These results demonstrate that variant IRAK-1 is associated with alterations in multiple intracellular events that are likely to contribute to increased NF-kappaB activation and inflammatory responses in individuals with this IRAK-1 haplotype.
Collapse
Affiliation(s)
- Gang Liu
- Department of Medicine, University of Alabama, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
80
|
Tsuruta Y, Park YJ, Siegal GP, Liu G, Abraham E. Involvement of Vitronectin in Lipopolysaccaride-Induced Acute Lung Injury. THE JOURNAL OF IMMUNOLOGY 2007; 179:7079-86. [DOI: 10.4049/jimmunol.179.10.7079] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
81
|
Abstract
BACKGROUND The past several years have seen remarkable advances in understanding the basic cellular and physiologic mechanisms underlying organ dysfunction and recovery relating to sepsis. Although several new therapeutic approaches have improved outcome in septic patients, the far-reaching potential of these new insights into sepsis-associated mechanisms is only beginning to be realized. AIM The Brussels Round Table Conference in 2006 convened >30 experts in the field of inflammation and sepsis to review recent advances involving sepsis and to discuss directions that the field is likely to take in the near future. FINDINGS Current understanding of the pathophysiology underlying sepsis-induced multiple organ dysfunction highlights the multiple cell populations and cell-signaling pathways involved in this complex condition. There is an increasing appreciation of interactions existing between different cells and organs affected by the septic process. The intricate cross-talk provided by temporal changes in mediators, hormones, metabolites, neural signaling, alterations in oxygen delivery and utilization, and by modifications in cell phenotypes underlines the adaptive and even coordinated processes beyond the dysregulated chaos in which sepsis was once perceived. Many pathologic processes previously considered to be detrimental are now viewed as potentially protective. Applying systems approaches to these complex processes will permit better appreciation of the effectiveness or harm of treatments, both present and future, and also will allow development not only of better directed, but also of more appropriately timed, strategies to improve outcomes from this still highly lethal condition.
Collapse
Affiliation(s)
- Edward Abraham
- Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | | |
Collapse
|
82
|
Miskolci V, Rollins J, Vu HY, Ghosh CC, Davidson D, Vancurova I. NFkappaB is persistently activated in continuously stimulated human neutrophils. Mol Med 2007. [PMID: 17592547 DOI: 10.2119/2006-00072.miskolci] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Increased activation of the transcription factor NFkappaB in the neutrophils has been associated with the pathogenesis of sepsis, acute lung injury (ALI), bronchopulmonary dysplasia (BPD), and other neutrophil-mediated inflammatory disorders. Despite recent progress in analyzing early NFkappaB activation in human neutrophils, activation of NFkappaB in persistently stimulated neutrophils has not been previously studied. Because it is the persistent NFkappaB activation that is thought to be involved in the host response to sepsis and the pathogenesis of ALI and BPD, we hypothesized that continuously stimulated human neutrophils may exhibit a late phase of NFkappaB activity. The goal of this study was to analyze the NFkappaB activation and expression of IkappaB and NFkappaB proteins during neutrophil stimulation with inflammatory signals for prolonged times. We demonstrate that neutrophil stimulation with lipopolysaccharide (LPS) and tumor necrosis factor-alpha (TNFalpha) induces, in addition to the early activation at 30-60 min, a previously unrecognized late phase of NFkappaB activation. In LPS-stimulated neutrophils, this NFkappaB activity typically had a biphasic character, whereas TNFalpha-stimulated neutrophils exhibited a continuous NFkappaB activity peaking around 9 h after stimulation. In contrast to the early NFkappaB activation that inversely correlates to the nuclear levels of IkappaBalpha, however, in continuously stimulated neutrophils, NFkappaB is persistently activated despite considerable levels of IkappaBalpha present in the nucleus. Our data suggest that NFkappaB is persistently activated in human neutrophils during neutrophil-mediated inflammatory disorders, and this persistent NFkappaB activity may represent one of the underlying mechanisms for the continuous production of proinflammatory mediators.
Collapse
Affiliation(s)
- Veronika Miskolci
- Department of Biological Sciences, St. John's University, New York, NY 11439, and Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, North Shore--Long Island Jewish Health System, New Hyde Park, NY, USA
| | | | | | | | | | | |
Collapse
|
83
|
Wu R, Dong W, Zhou M, Zhang F, Marini CP, Ravikumar TS, Wang P. Ghrelin attenuates sepsis-induced acute lung injury and mortality in rats. Am J Respir Crit Care Med 2007; 176:805-13. [PMID: 17626913 PMCID: PMC2020826 DOI: 10.1164/rccm.200604-511oc] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
RATIONALE Our study has shown that plasma levels of ghrelin, a stomach-derived peptide, are significantly reduced in sepsis, and that ghrelin administration improves organ blood flow via a nuclear factor (NF)-kappaB-dependent pathway. However, it remains unknown whether ghrelin has any protective effects on severe sepsis-induced acute lung injury (ALI) and, if so, whether inhibition of NF-kappaB plays any role in it. OBJECTIVES To test the hypothesis that ghrelin reduces severe sepsis-induced ALI and mortality through inhibition of NF-kappaB. METHODS Sepsis was induced in rats by cecal ligation and puncture (CLP). Five hours after CLP, a bolus intravenous injection of 2 nmol of ghrelin was followed by continuous infusion of 12 nmol of ghrelin via a minipump for 15 hours. Samples were harvested 20 hours post-CLP (i.e., severe sepsis). Pulmonary levels of ghrelin and proinflammatory cytokines were measured by ELISA. NF-kappaB p65 and IkappaBalpha expression and NF-kappaB activity were measured by Western blot analysis and ELISA, respectively. Pulmonary blood flow was measured with radioactive microspheres. In additional animals, the necrotic cecum was excised 20 hours post-CLP and 10-day survival was recorded. MEASUREMENTS AND MAIN RESULTS Pulmonary levels of ghrelin decreased significantly 20 hours post-CLP. Ghrelin administration restored pulmonary levels of ghrelin, reduced lung injury, increased pulmonary blood flow, down-regulated proinflammatory cytokines, inhibited NF-kappaB activation, and improved survival in sepsis. Administration of a specific ghrelin receptor antagonist worsened the survival rate after CLP and cecal excision. CONCLUSIONS Ghrelin can be developed as a novel treatment for severe sepsis-induced ALI. The protective effect of ghrelin is mediated through inhibition of NF-kappaB.
Collapse
Affiliation(s)
- Rongqian Wu
- Department of Surgery, North Shore University Hospital, Manhasset, New York, USA
| | | | | | | | | | | | | |
Collapse
|
84
|
Miskolci V, Rollins J, Vu HY, Ghosh CC, Davidson D, Vancurova I. NFkappaB is persistently activated in continuously stimulated human neutrophils. MOLECULAR MEDICINE (CAMBRIDGE, MASS.) 2007; 13:134-42. [PMID: 17592547 PMCID: PMC1892764 DOI: 10.2119/2006–00072.miskolci] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Accepted: 01/22/2007] [Indexed: 11/06/2022]
Abstract
Increased activation of the transcription factor NFkappaB in the neutrophils has been associated with the pathogenesis of sepsis, acute lung injury (ALI), bronchopulmonary dysplasia (BPD), and other neutrophil-mediated inflammatory disorders. Despite recent progress in analyzing early NFkappaB activation in human neutrophils, activation of NFkappaB in persistently stimulated neutrophils has not been previously studied. Because it is the persistent NFkappaB activation that is thought to be involved in the host response to sepsis and the pathogenesis of ALI and BPD, we hypothesized that continuously stimulated human neutrophils may exhibit a late phase of NFkappaB activity. The goal of this study was to analyze the NFkappaB activation and expression of IkappaB and NFkappaB proteins during neutrophil stimulation with inflammatory signals for prolonged times. We demonstrate that neutrophil stimulation with lipopolysaccharide (LPS) and tumor necrosis factor-alpha (TNFalpha) induces, in addition to the early activation at 30-60 min, a previously unrecognized late phase of NFkappaB activation. In LPS-stimulated neutrophils, this NFkappaB activity typically had a biphasic character, whereas TNFalpha-stimulated neutrophils exhibited a continuous NFkappaB activity peaking around 9 h after stimulation. In contrast to the early NFkappaB activation that inversely correlates to the nuclear levels of IkappaBalpha, however, in continuously stimulated neutrophils, NFkappaB is persistently activated despite considerable levels of IkappaBalpha present in the nucleus. Our data suggest that NFkappaB is persistently activated in human neutrophils during neutrophil-mediated inflammatory disorders, and this persistent NFkappaB activity may represent one of the underlying mechanisms for the continuous production of proinflammatory mediators.
Collapse
Affiliation(s)
- Veronika Miskolci
- Department of Biological Sciences, St. John’s University, New York
- Division of Neonatal-Perinatal Medicine, and Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, North Shore – Long Island Jewish Health System, New Hyde Park, NY, USA
| | - Janet Rollins
- Department of Biological Sciences, St. John’s University, New York
| | - Hai Yen Vu
- Department of Biological Sciences, St. John’s University, New York
| | - Chandra C Ghosh
- Department of Biological Sciences, St. John’s University, New York
| | - Dennis Davidson
- Division of Neonatal-Perinatal Medicine, and Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, North Shore – Long Island Jewish Health System, New Hyde Park, NY, USA
| | - Ivana Vancurova
- Department of Biological Sciences, St. John’s University, New York
- Division of Neonatal-Perinatal Medicine, and Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, North Shore – Long Island Jewish Health System, New Hyde Park, NY, USA
- Address correspondence and reprint requests to Ivana Vancurova, Department of Biological Sciences, St. John’s University, 8000 Utopia Parkway, New York, NY 11439. Phone: 718-990-6409; Fax: 718-990-5958; E-mail:
| |
Collapse
|
85
|
HMGB1 and LPS induce distinct patterns of gene expression and activation in neutrophils from patients with sepsis-induced acute lung injury. Intensive Care Med 2007. [PMID: 17581740 DOI: 10.1007/s00134-] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
OBJECTIVES Circulating levels of the proinflammatory mediator High Mobility Group Box Protein 1 (HMGB1) are increased in septic patients and may contribute to sepsis-induced organ dysfunction. Although HMGB1 has been shown to activate neutrophils from healthy volunteers, the responses of neutrophils from septic patients to HMGB1 have not been reported. In the present study we evaluated gene expression and activation of major intracellular signaling pathways in peripheral blood neutrophils obtained from patients with sepsis-induced acute lung injury after culture with HMGB1 or LPS. DESIGN Ex-vivo study performed in neutrophils from patients with sepsis-induced acute lung injury. SETTING Immunology and genetics laboratory at an academic medical center. PATIENTS AND PARTICIPANTS Twenty-two adult patients with sepsis-induced acute lung injury. MEASUREMENTS AND RESULTS Using gene arrays, distinct patterns of gene expression were found in neutrophils from septic patients after stimulation with HMGB1 or LPS. While more than three-quarters of the genes upregulated by HMGB1 in neutrophils from septic patients also demonstrated increased expression after culture with LPS, the majority of genes affected by LPS did not show altered expression in neutrophils stimulated with HMGB1. Culture of neutrophils with HMGB1 induced downregulation of its own expression, a finding not present after exposure to LPS. Although HMGB1 and LPS both increased nuclear translocation of NF-kappaB, the magnitude of this effect was greater in LPS stimulated neutrophils from patients with sepsis-induced acute lung injury. CONCLUSION These findings demonstrate that the patterns of gene expression differ between neutrophils from septic patients stimulated with HMGB1 or LPS, and also that neutrophils from septic patients are not anergic but instead demonstrate intact activation of NF-kappaB after exposure to LPS or HMGB1.
Collapse
|
86
|
Silva E, Arcaroli J, He Q, Svetkauskaite D, Coldren C, Nick JA, Poch K, Park JS, Banerjee A, Abraham E. HMGB1 and LPS induce distinct patterns of gene expression and activation in neutrophils from patients with sepsis-induced acute lung injury. Intensive Care Med 2007; 33:1829-39. [PMID: 17581740 DOI: 10.1007/s00134-007-0748-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2006] [Accepted: 05/21/2007] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Circulating levels of the proinflammatory mediator High Mobility Group Box Protein 1 (HMGB1) are increased in septic patients and may contribute to sepsis-induced organ dysfunction. Although HMGB1 has been shown to activate neutrophils from healthy volunteers, the responses of neutrophils from septic patients to HMGB1 have not been reported. In the present study we evaluated gene expression and activation of major intracellular signaling pathways in peripheral blood neutrophils obtained from patients with sepsis-induced acute lung injury after culture with HMGB1 or LPS. DESIGN Ex-vivo study performed in neutrophils from patients with sepsis-induced acute lung injury. SETTING Immunology and genetics laboratory at an academic medical center. PATIENTS AND PARTICIPANTS Twenty-two adult patients with sepsis-induced acute lung injury. MEASUREMENTS AND RESULTS Using gene arrays, distinct patterns of gene expression were found in neutrophils from septic patients after stimulation with HMGB1 or LPS. While more than three-quarters of the genes upregulated by HMGB1 in neutrophils from septic patients also demonstrated increased expression after culture with LPS, the majority of genes affected by LPS did not show altered expression in neutrophils stimulated with HMGB1. Culture of neutrophils with HMGB1 induced downregulation of its own expression, a finding not present after exposure to LPS. Although HMGB1 and LPS both increased nuclear translocation of NF-kappaB, the magnitude of this effect was greater in LPS stimulated neutrophils from patients with sepsis-induced acute lung injury. CONCLUSION These findings demonstrate that the patterns of gene expression differ between neutrophils from septic patients stimulated with HMGB1 or LPS, and also that neutrophils from septic patients are not anergic but instead demonstrate intact activation of NF-kappaB after exposure to LPS or HMGB1.
Collapse
Affiliation(s)
- Eliezer Silva
- Intensive Care Unit, Hospital Israelita Albert Einstein, Sao Paulo, SP, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Krupa A, Walencka MJ, Shrivastava V, Loyd T, Fudala R, Frevert CW, Martin TR, Kurdowska AK. Anti-KC autoantibody:KC complexes cause severe lung inflammation in mice via IgG receptors. Am J Respir Cell Mol Biol 2007; 37:532-43. [PMID: 17585113 PMCID: PMC2048682 DOI: 10.1165/rcmb.2006-0395oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
We have shown previously that high concentrations of IL-8 associated with anti-IL-8 autoantibodies (anti-IL-8:IL-8 complexes) are present in lung fluids from patients with the acute respiratory distress syndrome (ARDS), and correlate both with the development and outcome of ARDS. We also detected deposition of these complexes in lung tissues from patients with ARDS but not in control tissues. Moreover, we determined that IgG receptors (FcgammaRs) mediate activity of anti-IL-8:IL-8 complexes. In the current study, we generated anti-KC (KC = chemokine (CXC motif) ligand 1 (CXCL1)) autoantibody:KC immune complexes (KC-functional IL-8) in lungs of mice to develop a mouse model of autoimmune complex-induced lung inflammation. Both wild-type (WT) and gamma-chain-deficient mice that lack receptors for immune complexes (FcgammaRs) were studied. First, the mice were immunized with KC to induce anti-KC autoantibodies. Then, KC was administered intratracheally to generate anti-KC:KC complexes in the lung. Presence of anti-KC:KC complexes was associated with development of severe pulmonary inflammation that was, however, dramatically suppressed in gamma-chain-deficient mice. Second, because sepsis is considered the major risk factor for development of ARDS, we evaluated LPS-treated WT as well as gamma-chain-deficient mice for the presence of anti-KC:KC complexes and pulmonary inflammatory responses. We detected complexes between anti-KC autoantibodies and KC in lung lavages and tissues of mice treated with LPS. Moreover, gamma-chain-deficient mice that lack receptors for immune complexes were protected from LPS-induced pulmonary inflammation. Our results suggest that immune complexes containing autoantibodies contribute to development of lung inflammation in LPS-treated mice.
Collapse
Affiliation(s)
- Agnieszka Krupa
- Department of Biochemistry, University of Texas Health Center, Tyler, Texas 75708-3154, USA
| | | | | | | | | | | | | | | |
Collapse
|
88
|
Huh JW, Hong SB, Kim MJ, Lim CM, Koh Y. The Efficacy of α-lipoic Acid on the Endotoxin-induced Acute Lung Injury. Tuberc Respir Dis (Seoul) 2007. [DOI: 10.4046/trd.2007.62.2.105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Jin Won Huh
- Department of Internal Medicine, Ilsan Paik Hospital, Inje University, Goyang, Korea
| | - Sang Bum Hong
- Division of Pulmonary and Critical Care Medicine, Asan Medical Center, University of ulsan College of Medicine, Seoul, Korea
| | - Mi Jung Kim
- Asan Institute for Life Sciences, Seoul, Korea
| | - Chae-Man Lim
- Division of Pulmonary and Critical Care Medicine, Asan Medical Center, University of ulsan College of Medicine, Seoul, Korea
| | - Younsuck Koh
- Division of Pulmonary and Critical Care Medicine, Asan Medical Center, University of ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
89
|
Thirunavukkarasu C, Watkins SC, Gandhi CR. Mechanisms of endotoxin-induced NO, IL-6, and TNF-alpha production in activated rat hepatic stellate cells: role of p38 MAPK. Hepatology 2006; 44:389-98. [PMID: 16871588 DOI: 10.1002/hep.21254] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Compelling experimental evidence indicates that the interactions between endotoxin and hepatic stellate cells (HSCs) can play a significant role in the pathogenesis of liver disease. Endotoxin-induced release of a multifunctional mediator NO (via inducible NO synthase) and the proinflammatory cytokines tumor necrosis factor alpha (TNF-alpha) and interleukin (IL)-6 by HSCs could be an important mechanism of pathological changes in the liver. However, the signaling mechanisms of these effects are poorly understood. In this study, we found that endotoxin causes activation of mitogen-activated protein kinases (MAPKs) (extracellular signal-regulated protein kinase [ERK] 1 and 2, p38, and c-Jun NH2-terminal kinase [JNK]) and nuclear factor kappaB (NF-kappaB) and production of H(2)O(2) in culture-activated HSCs. However, only p38 and NF-kappaB were found to be responsible for the synthesis of NO, IL-6, and TNF-alpha. Exogenous H(2)O(2) caused modest stimulation of TNF-alpha synthesis, did not affect the synthesis of NO or IL-6, and did not activate NF-kappaB or MAPKs. Inhibition of p38 and NF-kappaB activation by SB203580 and pyrrolidine dithiocarbamate, respectively, blocked endotoxin-induced H(2)O(2), NO, TNF-alpha, and IL-6 synthesis. Inhibition of ERK1/2 and JNK phosphorylation did not alter these effects of endotoxin. Whereas SB203580 inhibited endotoxin-induced NF-kappaB activation, pyrrolidine dithiocarbamate did not affect p38 phosphorylation in endotoxin-stimulated cells. In conclusion, endotoxin-induced synthesis of NO, TNF-alpha, and IL-6 in HSCs is mediated by p38 and NF-kappaB, with involvement of H(2)O(2) in TNF-alpha production.
Collapse
Affiliation(s)
- Chinnasamy Thirunavukkarasu
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
90
|
Coldren CD, Nick JA, Poch KR, Woolum MD, Fouty BW, O'Brien JM, Gruber MP, Zamora MR, Svetkauskaite D, Richter DA, He Q, Park JS, Overdier KH, Abraham E, Geraci MW. Functional and genomic changes induced by alveolar transmigration in human neutrophils. Am J Physiol Lung Cell Mol Physiol 2006; 291:L1267-76. [PMID: 16861384 DOI: 10.1152/ajplung.00097.2006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Although the accumulation of neutrophils in the lungs and airways is common to many inflammatory lung diseases, including acute lung injury, the alterations that neutrophils undergo as they leave the peripheral circulation and migrate into the lungs have not been well characterized. Human volunteers were exposed to endotoxin by bronchoscopic instillation. The resulting air space neutrophil accumulation and peripheral blood neutrophils were isolated 16 h later, compared with circulating neutrophils isolated before or after to the pulmonary endotoxin exposure, and compared with circulating neutrophils exposed to endotoxin in vitro. Microarray analysis was performed on air space, circulatory, and in vitro endotoxin-stimulated neutrophils. Functional analysis included the determination of neutrophil apoptosis, chemotaxis, release of cytokines and growth factors, and superoxide anion release. Dramatic gene expression differences were apparent between air space and circulating neutrophils: approximately 15% of expressed genes have altered expression levels, including broad increases in inflammatory- and chemotaxis-related genes, as well as antiapoptotic and IKK-activating pathways. Functional analysis of air space compared with circulating neutrophils showed increased superoxide release, diminished apoptosis, decreased IL-8-induced chemotaxis, and a pattern of IL-8, macrophage inflammatory protein-1beta, monocyte chemoattractant protein-1, and tumor necrosis factor-alpha release different from either unstimulated or LPS-stimulated circulating neutrophils. Many of these changes are not elicited by in vitro treatment with endotoxin. Limited differences were detected between circulating neutrophils isolated before and 16 h after pulmonary endotoxin instillation. These results suggest that neutrophils sequestered in the lung become fundamentally different from those resident in the circulation, and this difference is distinct from in vitro activation with endotoxin.
Collapse
Affiliation(s)
- Christopher D Coldren
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Health Sciences Center, Denver, CO 80262, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Abraham E, Nick JA, Azam T, Kim SH, Mira JP, Svetkauskaite D, He Q, Zamora M, Murphy J, Park JS, Overdier K, Dinarello CA. Peripheral blood neutrophil activation patterns are associated with pulmonary inflammatory responses to lipopolysaccharide in humans. THE JOURNAL OF IMMUNOLOGY 2006; 176:7753-60. [PMID: 16751423 DOI: 10.4049/jimmunol.176.12.7753] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Increased nuclear accumulation of NF-kappaB in LPS-stimulated peripheral blood neutrophils has been shown to be associated with more severe clinical course in patients with infection associated acute lung injury. Such observations suggest that differences in neutrophil response may contribute to the pulmonary inflammation induced by bacterial infection. To examine this question, we sequentially measured LPS-induced DNA binding of NF-kappaB in neutrophils collected from healthy humans on at least three occasions, each separated by at least 2 wk, and then determined pulmonary inflammatory responses after instillation of LPS into the lungs. Consistent patterns of peripheral blood neutrophil responses, as determined by LPS-induced NF-kappaB DNA binding, were present in volunteers, with a >80-fold difference between individuals in the mean area under the curve for NF-kappaB activation. The number of neutrophils recovered from bronchoalveolar lavage after exposure to pulmonary LPS was significantly correlated with NF-kappaB activation in peripheral blood neutrophils obtained over the pre-LPS exposure period (r = 0.65, p = 0.009). DNA binding of NF-kappaB in pulmonary neutrophils also was associated with the mean NF-kappaB area under the curve for LPS-stimulated peripheral blood neutrophils (r = 0.63, p = 0.01). Bronchoalveolar lavage levels of IL-6 and TNFRII were significantly correlated with peripheral blood neutrophil activation patterns (r = 0.75, p = 0.001 for IL-6; and r = 0.48, p = 0.049 for TNFRII. These results demonstrate that stable patterns in the response of peripheral blood neutrophils to LPS exist in the human population and correlate with inflammatory response following direct exposure to LPS in the lung.
Collapse
Affiliation(s)
- Edward Abraham
- Division of Pulmonary Sciences and Critical Care Medicine and Division of Infectious Diseases, University of Colorado Health Sciences Center, Denver, 80262, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Brown KA, Brain SD, Pearson JD, Edgeworth JD, Lewis SM, Treacher DF. Neutrophils in development of multiple organ failure in sepsis. Lancet 2006; 368:157-69. [PMID: 16829300 DOI: 10.1016/s0140-6736(06)69005-3] [Citation(s) in RCA: 495] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Multiple organ failure is a major threat to the survival of patients with sepsis and systemic inflammation. In the UK and in the USA, mortality rates are currently comparable with and projected to exceed those from myocardial infarction. The immune system combats microbial infections but, in severe sepsis, its untoward activity seems to contribute to organ dysfunction. In this Review we propose that an inappropriate activation and positioning of neutrophils within the microvasculature contributes to the pathological manifestations of multiple organ failure. We further suggest that targeting neutrophils and their interactions with blood vessel walls could be a worthwhile therapeutic strategy for sepsis.
Collapse
Affiliation(s)
- K A Brown
- Division of Medical Education, King's College School of Medicine, London, UK.
| | | | | | | | | | | |
Collapse
|
93
|
|
94
|
Arcaroli J, Silva E, Maloney JP, He Q, Svetkauskaite D, Murphy JR, Abraham E. Variant IRAK-1 haplotype is associated with increased nuclear factor-kappaB activation and worse outcomes in sepsis. Am J Respir Crit Care Med 2006; 173:1335-41. [PMID: 16528020 PMCID: PMC2662973 DOI: 10.1164/rccm.200603-341oc] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
RATIONALE The IL-1 receptor-associated kinase (IRAK-1) plays a central role in TLR2- and TLR4-induced activation of nuclear factor (NF)-kappaB, a critical event in the transcriptional regulation of many sepsis-associated proinflammatory mediators. There are two haplotypes for the IRAK-1 gene in Caucasians, with the variant haplotype consisting of five intron single-nucleotide polymorphisms (SNPs) and three exon SNPs. OBJECTIVES To examine the functional significance of the IRAK-1 variant haplotype in modifying nuclear translocation of NF-kappaB and affecting outcomes from sepsis. MEASUREMENTS AND MAIN RESULTS One hundred fifty-five Caucasian patients with sepsis were included. Twenty-one (14%) were homozygous for the IRAK-1 variant haplotype as determined by a SNP in which T is replaced with C at nucleotide 1,595 within exon 12 of the IRAK-1 gene. The IRAK-1 variant haplotype was associated with increased nuclear levels of NF-kappaB in LPS-stimulated peripheral blood neutrophils from patients with sepsis compared with that found in patients with wild-type IRAK-1 haplotype (p=0.0009). There was an increased incidence of shock (p=0.047) (odds ratio [OR], 2.9; 95% confidence interval [CI], 1.1-7.7), greater requirement for more prolonged mechanical ventilator support (p=0.04) (OR, 2.7; 95% CI, 1.05-6.9), and higher 60-d mortality (p=0.05) (OR, 2.7; 95% CI, 1.0-6.8) in patients with the IRAK-1 variant haplotype compared with wild type. CONCLUSIONS These results indicate that the IRAK-1 variant haplotype is functionally significant in patients with sepsis, being associated with increased nuclear translocation of NF-kappaB, more severe organ dysfunction, and higher mortality.
Collapse
Affiliation(s)
- John Arcaroli
- Intensive Care unit, Albert Einstein Hospital and Division of Applied Physiology, Heart Institute, University of Sao Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
95
|
Haegens A, van der Vliet A, Butnor KJ, Heintz N, Taatjes D, Hemenway D, Vacek P, Freeman BA, Hazen SL, Brennan ML, Mossman BT. Asbestos-induced lung inflammation and epithelial cell proliferation are altered in myeloperoxidase-null mice. Cancer Res 2005; 65:9670-7. [PMID: 16266986 DOI: 10.1158/0008-5472.can-05-1751] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Asbestos fibers are carcinogens causing oxidative stress and inflammation, but the sources and ramifications of oxidant production by asbestos are poorly understood. Here, we show that inhaled chrysotile asbestos fibers cause increased myeloperoxidase activity in bronchoalveolar lavage fluids (BALF) and myeloperoxidase immunoreactivity in epithelial cells lining distal bronchioles and alveolar ducts, sites of initial lung deposition of asbestos fibers. In comparison with sham mice, asbestos-exposed myeloperoxidase-null (MPO-/-) and normal (MPO+/+) mice exhibited comparable increases in polymorphonuclear leukocytes, predominately neutrophils, in BALF after 9 days of asbestos inhalation. Differential cell counts on BALF revealed decreased proportions of macrophages and increased lymphocytes in all mice exposed to asbestos, but numbers were decreased overall in asbestos-exposed myeloperoxidase-null versus normal mice. Asbestos-associated lung inflammation in myeloperoxidase-null mice was reduced (P < or = 0.05) in comparison with normal asbestos-exposed mice at 9 days. Decreased lung inflammation in asbestos-exposed myeloperoxidase-null mice at 9 days was accompanied by increases (P < or = 0.05) in Ki-67- and cyclin D1-positive immunoreactive cells, markers of cell cycle reentry, in the distal bronchiolar epithelium. Asbestos-induced epithelial cell proliferation in myeloperoxidase-null mice at 30 days was comparable to that found at 9 days. In contrast, inflammation and epithelial cell proliferation in asbestos-exposed normal mice increased over time. These results support the hypothesis that myeloperoxidase status modulates early asbestos-induced oxidative stress, epithelial cell proliferation, and inflammation.
Collapse
Affiliation(s)
- Astrid Haegens
- Department of Pathology, College of Medicine, University of Vermont, Burlington, Vermont 05404, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Affiliation(s)
- Edward Abraham
- Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado at Denver and Health Sciences Center, Denver, CO, USA
| |
Collapse
|
97
|
Abstract
Multiple intracellular signaling pathways involving kinases, transcriptional factors, and the expression of immunoregulatory mediators are altered in sepsis. Recent data have shown stable patterns of activation among peripheral blood mononuclear cells and neutrophils in healthy human subjects. Although polymorphisms in Toll-like receptors play a contributory role in determining cellular activation, other factors are involved as well. Increased activation of the mitogen-activated protein kinase protein 38, Akt, and nuclear factor (NF)-kappa B in neutrophils and other cell populations obtained at early time points in the clinical course of sepsis-induced acute lung injury or after accidental trauma is associated with a more-severe clinical course, suggesting that a proinflammatory cellular phenotype contributes to organ system dysfunction in such settings. Identification of patients with cellular phenotypes characterized by increased activation of NF-kappa B, Akt, and protein 38, as well as discrete patterns of gene activation, may permit identification of patients with sepsis who are likely to have a worse clinical outcome, thereby permitting early institution of therapies that modulate deleterious signaling pathways before organ system dysfunction develops, reducing morbidity and improving survival.
Collapse
Affiliation(s)
- Edward Abraham
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Health Sciences Center, Denver, CO 80262, USA.
| |
Collapse
|
98
|
|
99
|
Strassheim D, Kim JY, Park JS, Mitra S, Abraham E. Involvement of SHIP in TLR2-induced neutrophil activation and acute lung injury. THE JOURNAL OF IMMUNOLOGY 2005; 174:8064-71. [PMID: 15944314 DOI: 10.4049/jimmunol.174.12.8064] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The SHIP converts phosphatidylinositol 3,4,5 triphosphate to phosphatidyl 3,4 biphosphate. SHIP has negative regulatory functions on PI3K-dependent signaling pathways, which occupy important roles in modulating neutrophil functions. We used neutrophils from transgenic SHIP(-/-) and SHIP(+/+) mice that were stimulated with peptidoglycan (PGN) to examine the role of SHIP in TLR2-induced neutrophil activation. SHIP(-/-) neutrophils demonstrated significantly increased activation of the PI3K-dependent kinase Akt after exposure to PGN. Release of cytokines and chemokines, including TNF-alpha, IL-1beta, IL-6, IL-10, and MIP-2, was also increased in SHIP(-/-) compared with SHIP(+/+) neutrophils. There was no difference in the nuclear translocation of the transcriptional factor NF-kappaB between PGN-stimulated SHIP(-/-) and SHIP(+/+) neutrophils. However, phosphorylation of the p65 subunit of NF-kappaB, an event essential for optimal transcriptional activity of NF-kappaB, was increased in TLR2-activated SHIP(-/-) neutrophils. SHIP(-/-) neutrophils demonstrated greater activation of ERK1/2 and p38 MAPKs than did SHIP(+/+) neutrophils after exposure to PGN. The severity of acute lung injury induced by PGN was greater in SHIP(-/-) as compared with SHIP(+/+) mice. These results demonstrate that SHIP has a negative regulatory role in TLR2-induced neutrophil activation and in the development of related in vivo neutrophil-dependent inflammatory processes, such as acute lung injury.
Collapse
Affiliation(s)
- Derek Strassheim
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Health Sciences Center, Denver, CO 80262, USA.
| | | | | | | | | |
Collapse
|
100
|
Zhu Y, Guo C, Cao L, Gong X, Wang C, Sun B. Different effects of surfactant and inhaled nitric oxide in modulation of inflammatory injury in ventilated piglet lungs. Pulm Pharmacol Ther 2005; 18:303-13. [PMID: 15777614 DOI: 10.1016/j.pupt.2005.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2004] [Revised: 12/23/2004] [Accepted: 01/12/2005] [Indexed: 11/17/2022]
Abstract
Septic acute lung injury (ALI) causes high morbidity and mortality in intensive care service as a result of biotrauma and dysfunction in the lungs and other organ systems. We hypothesized that surfactant and/or inhaled nitric oxide (iNO) may have different effects in modulation of inflammatory injury in septic ALI. Twenty-four healthy, 6-9 kg piglets were anesthetized, and intraperitoneally injected with Escherichia coli, followed by a low tidal volume ventilation until sepsis and ALI developed within 4-6 h. They were then randomly treated in groups (n=6 each) as: control (C), inhaled NO at 10 ppm (NO), surfactant at 100mg/kg (Surf), or both surfactant and iNO (SNO). A normal control group (N) was sham-injected and similarly ventilated. Over the 24 h of treatment period, both Surf, and SNO groups had significantly improved PaO2/FiO2, dynamic compliance and resistance of respiratory system. At 24h, the best alveolar aeration and least protein leakage, the lowest wet-to-dry lung weight ratio and lung injury score were found in SNO. Activity of nuclear factor kappa B (NF-kappaB) and myeloperoxidase, interleukin 8 mRNA expression and melondialdehyde were significantly increased, and IL-10 mRNA decreased, in lung tissue of the C group, but were significantly altered in the SNO group, and moderately altered in either NO or Surf group. We conclude that the effects of lung protection by surfactant and/or iNO in this model may be different in modulation of inflammatory cytokine mRNA expression and activity of NF-kappaB, and iNO did not have adverse effects.
Collapse
Affiliation(s)
- Yourong Zhu
- Laboratory of Pediatric Respiratory and Intensive Care Medicine, Children's Hospital, Fudan University, 183 Feng Lin Road, Shanghai 200032, People's Republic of China
| | | | | | | | | | | |
Collapse
|