Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Front Immunol ; 12: 693874, 2021.
Article in English | MEDLINE | ID: mdl-34349759

ABSTRACT

Background: The mechanisms by which moderate tidal volume ventilation (MTV) exacerbates preexisting lung injury are unclear. We hypothesized that systemic endotoxemia via the gut-lung axis would lead to non-canonical and canonical inflammasome activation and pyroptosis in a two-hit model involving polyinosinic-polycytidylic acid (Poly(I:C)), a synthetic analog of dsRNA and MTV and that this would associate with acute lung injury (ALI). Methods: Anesthetized mice were administered Poly(I:C) intratracheally and then 6 h later, they were mechanically ventilated for 4 h with otherwise non-injurious MTV (10ml/kg). Changes in intestinal and alveolar capillary permeability were measured. Further documentation of ALI was assessed by evans blue albumin permeability, protein and IL-1 family concentration in bronchoalveolar lavage fluid (BALF) or plasma, and histopathology in cohorts of wildtype (WT), whole body genetically ablated caspase-11 (caspase-11-/-), caspase-1/caspase-11 double knockout (caspase-1/11-/-), gasdermin D (GSDMD)-/-, nucleotide-binding domain leucine-rich repeat-containing protein 3 (NLRP3)-/- and advanced glycosylation end product-specific receptor (RAGE) -/- mice. Results: Non-injurious MTV exacerbated the mild lung injury associated with Poly(I:C) administration. This included the disruption of alveolar-capillary barrier and increased levels of interleukin (IL)-6, high mobility group proteins 1 (HMGB-1), IL-1ß in BALF and IL-18 in plasma. Combined (Poly(I:C)-MTV) injury was associated with increase in gastrointestinal permeability and endotoxin in plasma and BALF. Poly(I:C)-MTV injury was sensitive to caspase-11 deletion with no further contribution of caspase-1 except for maturation and release of IL-18 (that itself was sensitive to deletion of NLRP3). Combined injury led to large increases in caspase-1 and caspase-11. Genetic ablation of GSDMD attenuated alveolar-capillary disruption and release of cytokines in combined injury model. Conclusions: The previously noted exacerbation of mild Poly(I:C)-induced ALI by otherwise non-injurious MTV is associated with an increase in gut permeability resulting in systemic endotoxemia. The gut-lung axis resulted in activation of pulmonary non-canonical (cytosolic mediated caspase-11 activation) and canonical (caspase-1) inflammasome (NLRP3) mediated ALI in this two-hit model resulting in GSDMD sensitive alveolar capillary barrier disruption, pyroptosis (alveolar macrophages) and cytokine maturation and release (IL-1ß; IL-18). Pharmacologic strategies aimed at disrupting communication between gut and lung, inhibition of inflammasomes or GSDMD in pyroptosis may be useful in ALI.


Subject(s)
Acute Lung Injury/chemically induced , Caspases, Initiator/metabolism , Gastrointestinal Microbiome , Intestines/microbiology , Lung/enzymology , Poly I-C , Respiration, Artificial , Ventilator-Induced Lung Injury/etiology , Acute Lung Injury/enzymology , Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Animals , Bacteria/metabolism , Caspases, Initiator/genetics , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/metabolism , Lung/pathology , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phosphate-Binding Proteins/genetics , Phosphate-Binding Proteins/metabolism , Pyroptosis , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/microbiology , Ventilator-Induced Lung Injury/pathology
2.
Respir Res ; 22(1): 159, 2021 May 22.
Article in English | MEDLINE | ID: mdl-34022899

ABSTRACT

BACKGROUND: Patients in intensive care units (ICUs) often received broad-spectrum antibiotic treatment and Acinetobacter baumannii (A.b.) and Pseudomonas aeruginosa (P.a.) were the most common pathogens causing ventilator-associated pneumonia (VAP). This study aimed to examine the effects and mechanism of mechanical ventilation (MV) on A.b.-induced lung injury and the involvement of alveolar macrophages (AMs). METHODS: C57BL/6 wild-type (WT) and c-Jun N-terminal kinase knockout (JNK1-/-) mice received MV for 3 h at 2 days after nasal instillation of A.b., P.a. (1 × 106 colony-forming unit, CFU), or normal saline. RESULTS: Intranasal instillation of 106 CFU A.b. in C57BL/6 mice induced a significant increase in total cells and protein levels in the bronchoalveolar lavage fluid (BALF) and neutrophil infiltration in the lungs. MV after A.b. instillation increases neutrophil infiltration, interleukin (IL)-6 and vascular cell adhesion molecule (VCAM) mRNA expression in the lungs and total cells, IL-6 levels, and nitrite levels in the BALF. The killing activity of AMs against A.b. was lower than against P.a. The diminished killing activity was parallel with decreased tumor necrosis factor-α production by AMs compared with A.b. Inducible nitric oxide synthase inhibitor, S-methylisothiourea, decreased the total cell number in BALF on mice receiving A.b. instillation and ventilation. Moreover, MV decreased the A.b. and P.a. killing activity of AMs. MV after A.b. instillation induced less total cells in the BALF and nitrite production in the serum of JNK1-/- mice than those of WT mice. CONCLUSION: A.b. is potent in inducing neutrophil infiltration in the lungs and total protein in the BALF. MV enhances A.b.-induced lung injury through an increase in the expression of VCAM and IL-6 levels in the BALF and a decrease in the bacteria-killing activity of AMs. A lower inflammation level in JNK1-/- mice indicates that A.b.-induced VAP causes lung injury through JNK signaling pathway in the lungs.


Subject(s)
Acinetobacter Infections/enzymology , Acinetobacter baumannii/pathogenicity , Lung/enzymology , Mitogen-Activated Protein Kinase 8/metabolism , Pneumonia, Ventilator-Associated/enzymology , Respiration, Artificial/adverse effects , Ventilator-Induced Lung Injury/enzymology , Acinetobacter Infections/microbiology , Acinetobacter Infections/pathology , Animals , Cells, Cultured , Disease Models, Animal , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/microbiology , Lung/pathology , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/microbiology , Male , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Neutrophil Infiltration , Nitric Oxide Synthase Type II/metabolism , Pneumonia, Ventilator-Associated/microbiology , Pneumonia, Ventilator-Associated/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Ventilator-Induced Lung Injury/microbiology , Ventilator-Induced Lung Injury/pathology
3.
Mol Med Rep ; 21(2): 607-614, 2020 02.
Article in English | MEDLINE | ID: mdl-31789412

ABSTRACT

Ventilator­induced lung injury (VILI) is a life­threatening condition caused by the inappropriate use of mechanical ventilation (MV). However, the precise molecular mechanism inducing the development of VILI remains to be elucidated. In the present study, it was revealed that the calcineurin/NFATc4 signaling pathway mediates the expression of adhesion molecules and proinflammatory cytokines essential for the development of VILI. The present results revealed that a high tidal volume ventilation (HV) caused lung inflammation and edema in the alveolar walls and the infiltration of inflammatory cells. The calcineurin activity and protein expression in the lungs were increased in animals with VILI, and NFATc4 translocated into the nucleus following calcineurin activation. Furthermore, the translocation of NFATc4 and lung injury were prevented by a calcineurin inhibitor (CsA). Thus, the present results highlighted the critical role of the calcineurin/NFATc4 signaling pathway in VILI and suggest that this pathway coincides with the release of ICAM­1, VCAM­1, TNF­α and IL­1ß.


Subject(s)
Calcineurin/metabolism , NFATC Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Ventilator-Induced Lung Injury/metabolism , Animals , Calcineurin/genetics , Calcineurin Inhibitors/pharmacology , Cell Nucleus/metabolism , Edema/complications , Edema/metabolism , Inflammation/complications , Inflammation/metabolism , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Peroxidase/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/genetics , Ventilator-Induced Lung Injury/pathology
4.
Thorax ; 73(4): 321-330, 2018 04.
Article in English | MEDLINE | ID: mdl-28947666

ABSTRACT

BACKGROUND: Neutrophils may cause tissue disruption during migration and by releasing cytotoxic molecules. However, the benefits of neutrophil depletion observed in experimental models of lung injury do not correspond with the poor outcome of neutropenic patients. METHODS: To clarify the role of neutrophils during repair, mice with ventilator induced lung injury (VILI) were rendered neutropenic after damage, and followed for 48 hours of spontaneous breathing. Lungs were harvested and inflammatory mediators and matrix metalloproteinases measured. Bronchoalveolar lavage fluid (BALF) from ventilated patients with acute respiratory distress syndrome, with or without neutropenia, was collected, the same mediators measured and their effects in an ex vivo model of alveolar repair studied. Finally, neutropenic mice were treated after VILI with exogenous matrix metalloproteinase-9 (MMP-9). RESULTS: Lungs from neutropenic animals showed delayed repair and displayed higher levels of tumour necrosis factor α, interferon γ and macrophage inflammatory protein 2, and absence of MMP-9. BALF from ventilated neutropenic patients with acute respiratory distress syndrome showed similar results. BALFs from neutropenic patients yielded a delayed closure rate of epithelial wounds ex vivo, which was improved by removal of collagen or addition of exogenous MMP-9. Lastly, treatment of neutropenic mice with exogenous MMP-9 after VILI reduced tissue damage without modifying cytokine concentrations. CONCLUSION: Release of MMP-9 from neutrophils is required for adequate matrix processing and lung repair.


Subject(s)
Matrix Metalloproteinase 9/biosynthesis , Neutropenia/metabolism , Neutrophils/metabolism , Respiratory Distress Syndrome/metabolism , Ventilator-Induced Lung Injury/metabolism , Animals , Biomarkers/blood , Bronchoalveolar Lavage Fluid/cytology , Chemokine CXCL2/metabolism , Disease Models, Animal , Humans , Interferon-gamma/metabolism , Mice , Neutropenia/pathology , Respiratory Distress Syndrome/enzymology , Respiratory Distress Syndrome/pathology , Tumor Necrosis Factor-alpha/metabolism , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/pathology , Ventilator-Induced Lung Injury/prevention & control
5.
Am J Respir Cell Mol Biol ; 58(5): 604-613, 2018 05.
Article in English | MEDLINE | ID: mdl-29077485

ABSTRACT

Profound lung vascular permeability is a cardinal feature of acute respiratory distress syndrome (ARDS) and ventilator-induced lung injury (VILI), two syndromes known to centrally involve the nonmuscle isoform of myosin light chain kinase (nmMLCK) in vascular barrier dysregulation. Two main splice variants, nmMLCK1 and nmMLCK2, are well represented in human lung endothelial cells and encoded by MYLK, and they differ only in the presence of exon 11 in nmMLCK1, which contains critical phosphorylation sites (Y464 and Y471) that influence nmMLCK enzymatic activity, cellular translocation, and localization in response to vascular agonists. We recently demonstrated the functional role of SNPs in altering MYLK splicing, and in the present study we sought to identify the role of splicing factors in the generation of nmMLCK1 and nmMLCK2 spliced variants. Using bioinformatic in silico approaches, we identified a putative binding site for heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1), a recognized splicing factor. We verified hnRNPA1 binding to MYLK by gel shift analyses and that hnRNPA1 gene and protein expression is upregulated in mouse lungs obtained from preclinical models of ARDS and VILI and in human endothelial cells exposed to 18% cyclic stretch, a model that reproduces the excessive mechanical stress observed in VILI. Using an MYLK minigene approach, we established a direct role of hnRNPA1 in MYLK splicing and in the context of 18% cyclic stretch. In summary, these data indicate an important regulatory role for hnRNPA1 in MYLK splicing, and they increase understanding of MYLK splicing in the regulation of lung vascular integrity during acute lung inflammation and excessive mechanical stress, such as that observed in ARDS and VILI.


Subject(s)
Alternative Splicing , Calcium-Binding Proteins/metabolism , Endothelial Cells/enzymology , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Lung/blood supply , Myosin-Light-Chain Kinase/metabolism , Respiratory Distress Syndrome/enzymology , Ventilator-Induced Lung Injury/enzymology , Animals , Binding Sites , Calcium-Binding Proteins/genetics , Capillary Permeability , Disease Models, Animal , Electric Impedance , Exons , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein A1/genetics , Humans , Introns , Mechanotransduction, Cellular , Mice , Myosin-Light-Chain Kinase/genetics , Protein Binding , Pulmonary Stretch Receptors/metabolism , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/physiopathology , Ventilator-Induced Lung Injury/genetics , Ventilator-Induced Lung Injury/physiopathology
6.
Am J Respir Cell Mol Biol ; 58(4): 519-529, 2018 04.
Article in English | MEDLINE | ID: mdl-29216437

ABSTRACT

The erythropoietin-producing hepatoma (Eph) receptor tyrosine kinase A2 (EphA2) and its ligand, ephrinA1, play a pivotal role in inflammation and tissue injury by modulating the epithelial and endothelial barrier integrity. Therefore, EphA2 receptor may be a potential therapeutic target for modulating ventilator-induced lung injury (VILI). To support this hypothesis, here, we analyzed EphA2/ephrinA1 signaling in the process of VILI and determined the role of EphA2/ephrinA1 signaling in the protective mechanism of prone positioning in a VILI model. Wild-type mice were ventilated with high (24 ml/kg; positive end-expiratory pressure, 0 cm; 5 h) tidal volume in a supine or prone position. Anti-EphA2 receptor antibody or IgG was administered to the supine position group. Injury was assessed by analyzing the BAL fluid, lung injury scoring, and transmission electron microscopy. Lung lysates were evaluated using cytokine/chemokine ELISA and Western blotting of EphA2, ephrinA1, PI3Kγ, Akt, NF-κB, and P70S6 kinase. EphA2/ephrinA1 expression was higher in the supine high tidal volume group than in the control group, but it did not increase upon prone positioning or anti-EphA2 receptor antibody treatment. EphA2 antagonism reduced the extent of VILI and downregulated the expression of PI3Kγ, Akt, NF-κB, and P70S6 kinase. These findings demonstrate that EphA2/ephrinA1 signaling is involved in the molecular mechanism of VILI and that modulation of EphA2/ehprinA1 signaling by prone position or EphA2 antagonism may be associated with the lung-protective effect. Our data provide evidence for EphA2/ehprinA1 as a promising therapeutic target for modulating VILI.


Subject(s)
Lung/enzymology , Prone Position , Receptor, EphA2/metabolism , Ventilator-Induced Lung Injury/prevention & control , Animals , Antibodies/pharmacology , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Disease Models, Animal , Ephrin-A1/metabolism , Lung/drug effects , Lung/ultrastructure , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, EphA2/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/pathology
7.
Mol Med Rep ; 16(4): 4119-4126, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28765901

ABSTRACT

Although it is essential in critical care medicine, mechanical ventilation often results in ventilator­induced lung injury (VILI). Treating mice with lipopolysaccharide has been reported to upregulate the expression of miR­127, which has been implicated in the modulation of immune responses. However, the putative roles of miR­127 during the development of VILI have yet to be elucidated. The present study demonstrated that challenging mice with mechanical ventilation for 6 h significantly upregulated the expression of miR­127 in bronchoalveolar lavage fluid, serum and lung tissue samples. Conversely, following the downregulation of miR­127 expression in vivo using an adenovirus delivery system, VILI­associated pathologies, including alterations in the pulmonary wet/dry ratio, pulmonary permeability, lung neutrophil infiltration and levels of pro­inflammatory cytokines, were significantly attenuated. In addition, miR­127 knockdown inhibited the ventilation­induced activation of nuclear factor (NF)­κB and p38 mitogen­activated protein kinase (MAPK). These findings suggested that the upregulation of miR­127 expression may contribute to the development of VILI, through the modulation of pulmonary permeability, the induction of histopathological alterations, and the potentiation of inflammatory responses involving NF­κB and p38 MAPK­associated signaling pathways.


Subject(s)
MicroRNAs/genetics , Ventilator-Induced Lung Injury/genetics , Animals , Female , Gene Silencing , Inflammation/genetics , Inflammation/pathology , Mice, Inbred C57BL , MicroRNAs/metabolism , Protein Kinases/metabolism , Pulmonary Alveoli/pathology , Signal Transduction , Up-Regulation/genetics , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/pathology
8.
Respir Res ; 16: 112, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26377087

ABSTRACT

BACKGROUND: Mechanical ventilation and concomitant administration of hyperoxia in patients with acute respiratory distress syndrome can damage the alveolar epithelial and capillary endothelial barrier by producing inflammatory cytokines and reactive oxygen species. The Src tyrosine kinase and Smad3 are crucial inflammatory regulators used for ventilator-induced lung injury (VILI). The mechanisms regulating interactions between high-tidal-volume mechanical ventilation, hyperoxia, and acute lung injury (ALI) are unclear. We hypothesized that high-tidal-volume mechanical stretches and hyperoxia augment lung inflammation through upregulation of the Src and Smad3 pathways. METHODS: Wild-type or Src-deficient C57BL/6 mice, aged between 6 and 8 weeks, were exposed to high-tidal-volume (30 mL/kg) ventilation with room air or hyperoxia for 1-4 h after 2-mg/kg Smad3 inhibitor (SIS3) administration. Nonventilated mice were used as control subjects. RESULTS: We observed that the addition of hyperoxia to high-tidal-volume mechanical ventilation further induced microvascular permeability, neutrophil infiltration, macrophage inflammatory protein-2 and matrix metalloproteinase-9 (MMP-9) production, malondialdehyde, nicotinamide adenine dinucleotide phosphate oxidase activity, MMP-9 mRNA expression, hypoxemia, and Src and Smad3 activation (P < 0.05). Hyperoxia-induced augmentation of VILI was attenuated in Src-deficient mice and mice with pharmacological inhibition of Smad3 activity by SIS3 (P < 0.05). Mechanical ventilation of Src-deficient mice with hyperoxia further reduced the activation of Smad3. CONCLUSIONS: Our data suggest that hyperoxia-increased high-tidal-volume ventilation-induced ALI partially depends on the Src and Smad3 pathways.


Subject(s)
Hyperoxia/complications , Lung/enzymology , Neutrophils/enzymology , Oxidative Stress , Pneumonia/etiology , Respiration, Artificial/adverse effects , Signal Transduction , Smad3 Protein/metabolism , Ventilator-Induced Lung Injury/etiology , src-Family Kinases/metabolism , Animals , Capillary Permeability , Chemokine CXCL2/metabolism , Disease Models, Animal , Genetic Predisposition to Disease , Isoquinolines/pharmacology , Lung/blood supply , Lung/drug effects , Lung/immunology , Lung/pathology , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/metabolism , Neutrophil Infiltration , Neutrophils/drug effects , Neutrophils/immunology , Oxidative Stress/drug effects , Phenotype , Pneumonia/enzymology , Pneumonia/genetics , Pneumonia/immunology , Pneumonia/pathology , Pneumonia/prevention & control , Pyridines/pharmacology , Pyrroles/pharmacology , Signal Transduction/drug effects , Smad3 Protein/antagonists & inhibitors , Tidal Volume , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/genetics , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/pathology , Ventilator-Induced Lung Injury/prevention & control , src-Family Kinases/deficiency , src-Family Kinases/genetics
9.
J Surg Res ; 199(2): 601-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26163326

ABSTRACT

BACKGROUND: Simvastatin reduces ventilator-induced lung injury and is regularly used in clinical practice. This study aimed to test the hypotheses that long-term use of simvastatin could affect the incidence and severity of ventilator-induced lung injury after mechanical ventilation, and the process may involve heme oxygenase-1 (HO-1). MATERIALS AND METHODS: Forty healthy adult Sprague-Dawley rats were randomly divided into four groups, namely control, ventilation, simvastatin, and simvastatin + ventilation groups. Saline (control and ventilation groups) or 10 mg kg(-1) d(-1) simvastatin (simvastatin and simvastatin + ventilation groups) was administered by gavage to the animals for 4 wk. Mechanical ventilation (tidal volume 50 mL/kg) was then applied for 4 h to the ventilation and simvastatin + ventilation groups. Lung tissues were harvested for hematoxylin-eosin staining and pathologic examination, and HO-1 contents were measured by immunoblotting and polymerase chain reaction. RESULTS: A severe pathologic damage was observed in rats that underwent mechanical ventilation. Interestingly, protein concentration, wet/dry weight ratio, myeloperoxidase activity, and malondialdehyde level were increased, and superoxide dismutase activity decreased, in lung tissues after mechanical ventilation. The pathologic damage was substantially alleviated in rats treated with simvastatin before mechanical ventilation: reduced protein concentration, wet/dry weight ratio, myeloperoxidase activity, and malondialdehyde level, and increased superoxide dismutase activity in lung tissues, compared with the ventilation group. Both mechanical ventilation and simvastatin administration induced HO-1 messenger RNA and protein expression in lung tissues. CONCLUSIONS: Long-term administration of simvastatin significantly reduces the inflammatory response and pulmonary injury induced by mechanical ventilation, potentially by upregulating HO-1 in lung tissues.


Subject(s)
Heme Oxygenase (Decyclizing)/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Respiration, Artificial/adverse effects , Simvastatin/administration & dosage , Ventilator-Induced Lung Injury/prevention & control , Animals , Disease Models, Animal , Lung/enzymology , Lung/pathology , Malondialdehyde/metabolism , Peroxidase/metabolism , Random Allocation , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Up-Regulation/drug effects , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/pathology
10.
Respir Res ; 16: 24, 2015 Feb 14.
Article in English | MEDLINE | ID: mdl-25848815

ABSTRACT

BACKGROUND: Mortality from severe acute respiratory distress syndrome exceeds 40% and there is no available pharmacologic treatment. Mechanical ventilation contributes to lung dysfunction and mortality by causing ventilator-induced lung injury. We explored the utility of simvastatin in a mouse model of severe ventilator-induced lung injury. METHODS: Male C57BL6 mice (n = 7/group) were pretreated with simvastatin or saline and received protective (8 mL/kg) or injurious (25 mL/kg) ventilation for four hours. Three doses of simvastatin (20 mg/kg) or saline were injected intraperitoneally on days -2, -1 and 0 of the experiment. Lung mechanics, (respiratory system elastance, tissue damping and airway resistance), were evaluated by forced oscillation technique, while respiratory system compliance was measured with quasi-static pressure-volume curves. A pathologist blinded to treatment allocation scored hematoxylin-eosin-stained lung sections for the presence of lung injury. Pulmonary endothelial dysfunction was ascertained by bronchoalveolar lavage protein content and lung tissue expression of endothelial junctional protein Vascular Endothelial cadherin by immunoblotting. To assess the inflammatory response in the lung, we determined bronchoalveolar lavage fluid total cell content and neutrophil fraction by microscopy and staining in addition to Matrix-Metalloprotease-9 by ELISA. For the systemic response, we obtained plasma levels of Tumor Necrosis Factor-α, Interleukin-6 and Matrix-Metalloprotease-9 by ELISA. Statistical hypothesis testing was undertaken using one-way analysis of variance and Tukey's post hoc tests. RESULTS: Ventilation with high tidal volume (HVt) resulted in significantly increased lung elastance by 3-fold and decreased lung compliance by 45% compared to low tidal volume (LVt) but simvastatin abrogated lung mechanical alterations of HVt. Histologic lung injury score increased four-fold by HVt but not in simvastatin-pretreated mice. Lavage pleocytosis and neutrophilia were induced by HVt but were significantly attenuated by simvastatin. Microvascular protein permeability increase 20-fold by injurious ventilation but only 4-fold with simvastatin. There was a 3-fold increase in plasma Tumor Necrosis Factor-α, a 7-fold increase in plasma Interleukin-6 and a 20-fold increase in lavage fluid Matrix-Metalloprotease-9 by HVt but simvastatin reduced these levels to control. Lung tissue vascular endothelial cadherin expression was significantly reduced by injurious ventilation but remained preserved by simvastatin. CONCLUSION: High-dose simvastatin prevents experimental hyperinflation lung injury by angioprotective and anti-inflammatory effects.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lung/drug effects , Simvastatin/pharmacology , Ventilator-Induced Lung Injury/prevention & control , Airway Resistance/drug effects , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Capillary Permeability/drug effects , Disease Models, Animal , Elasticity , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Inflammation Mediators/blood , Lung/enzymology , Lung/pathology , Lung/physiopathology , Lung Compliance/drug effects , Male , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Pneumonia/enzymology , Pneumonia/pathology , Pneumonia/physiopathology , Pneumonia/prevention & control , Pulmonary Edema/enzymology , Pulmonary Edema/pathology , Pulmonary Edema/physiopathology , Pulmonary Edema/prevention & control , Time Factors , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/pathology , Ventilator-Induced Lung Injury/physiopathology
11.
Respir Res ; 15: 158, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25471013

ABSTRACT

BACKGROUND: Ventilator-induced lung injury (VILI) is characterized by increased alveolar permeability, pulmonary edema. The tyrosine kinase, c-Src, is involved in VILI but its role has not been fully elucidated. This study examined the relationship between c-Src activation and occludin levels in VILI both in vitro and in vivo. METHODS: For the in vivo study, Wistar rats were randomly divided into five groups: control (group C); normal tidal volume (group M); normal tidal volume + c-Src inhibitor (PP2) (group M + P); high tidal volume (group H); and high tidal volume + c-Src inhibitor (PP2) (group H + P). Rats in all groups but group C underwent mechanical ventilation for 4 h. For the in vitro study, MLE-12 cells pretreated with PP2 and siRNA underwent cyclic stretching at 8% or 20% for 0, 1, 2 and 4 h. The expressions of occludin, c-Src, and p-c-Src were analyzed by western blotting, hematoxylin and eosin (HE) staining, and immunofluorescence. RESULTS: For the in vivo study, rats in group H showed decreased occludin expression and activated c-Src compared with group C. HE staining and lung injury score showed more severe lung injury and alveolar edema in group H compared with group M and group C. Group H + P had less pulmonary edema induced by the high tidal volume ventilation. For the in vitro study, occludin expression decreased and c-Src activation increased as indicated by the phosphorylation of c-Src over time. Consistently, PP2 could restore occludin levels. CONCLUSIONS: Mechanical ventilation can activate c-Src by phosphorylation and increase the degradation of occludin. c-Src inhibitor can ameliorate barrier function and lung injury by up-regulating occludin.


Subject(s)
Lung/enzymology , Occludin/metabolism , Ventilator-Induced Lung Injury/enzymology , src-Family Kinases/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Line , Disease Models, Animal , Enzyme Activation , Lung/drug effects , Lung/physiopathology , Mechanotransduction, Cellular , Mice , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proteolysis , Pulmonary Edema/enzymology , RNA Interference , Rats, Wistar , Time Factors , Transfection , Ventilator-Induced Lung Injury/physiopathology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
12.
Biomed Res Int ; 2014: 749097, 2014.
Article in English | MEDLINE | ID: mdl-25019086

ABSTRACT

BACKGROUND: Ventilator-induced lung injury-(VILI-) induced endothelial permeability is regulated through the Rho-dependent signaling pathway. Ibuprofen inhibits Rho activation in animal models of spinal-cord injury and Alzheimer's disease. The study aims to investigate ibuprofen effects on high tidal volume associated VILI. METHODS: Twenty-eight adult male Sprague-Dawley rats were randomized to receive a ventilation strategy with three different interventions for 2 h: (1) a high-volume zero-positive end-expiratory pressure (PEEP) (HVZP) group; (2) an HVZP + ibuprofen 15 mg/kg group; and (3) an HVZP + ibuprofen 30 mg/kg group. A fourth group without ventilation served as the control group. Rho-kinase activity was determined by ratio of phosphorylated ezrin, radixin, and moesin (p-ERM), substrates of Rho-kinase, to total ERM. VILI was characterized by increased pulmonary protein leak, wet-to-dry weight ratio, cytokines level, and Rho guanine nucleotide exchange factor (GEF-H1), RhoA activity, p-ERM/total ERM, and p-myosin light chain (MLC) protein expression. RESULTS: Ibuprofen pretreatment significantly reduced the HVZP ventilation-induced increase in pulmonary protein leak, wet-to-dry weight ratio, bronchoalveolar lavage fluid interleukin-6 and RANTES levels, and lung GEF-H1, RhoA activity, p-ERM/total ERM, and p-MLC protein expression. CONCLUSION: Ibuprofen attenuated high tidal volume induced pulmonary endothelial hyperpermeability. This protective effect was associated with a reduced Rho-kinase activity.


Subject(s)
Ibuprofen/administration & dosage , Lung/metabolism , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/prevention & control , rho-Associated Kinases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Down-Regulation/drug effects , Enzyme Activation , Lung/drug effects , Male , Rats , Rats, Sprague-Dawley , Treatment Outcome
13.
J Korean Med Sci ; 29(4): 556-63, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24753704

ABSTRACT

This study was performed to examine the role of transglutaminase 2 (TG2) in ventilator-induced lung injury (VILI). C57BL/6 mice were divided into six experimental groups: 1) control group; 2) lipopolysaccharide (LPS) group; 3) lung protective ventilation (LPV) group; 4) VILI group; 5) VILI with cystamine, a TG2 inhibitor, pretreatment (Cyst+VILI) group; and 6) LPV with cystamine pretreatment (Cyst+LPV) group. Acute lung injury (ALI) score, TG2 activity and gene expression, inflammatory cytokines, and nuclear factor-κB (NF-κB) activity were measured. TG2 activity and gene expression were significantly increased in the VILI group (P < 0.05). Cystamine pretreatment significantly decreased TG2 activity and gene expression in the Cyst+VILI group (P < 0.05). Inflammatory cytokines were higher in the VILI group than in the LPS and LPV groups (P < 0.05), and significantly lower in the Cyst+VILI group than the VILI group (P < 0.05). NF-κB activity was increased in the VILI group compared with the LPS and LPV groups (P < 0.05), and significantly decreased in the Cyst+VILI group compared to the VILI group (P = 0.029). The ALI score of the Cyst+VILI group was lower than the VILI group, but the difference was not statistically significant (P = 0.105). These results suggest potential roles of TG2 in the pathogenesis of VILI.


Subject(s)
GTP-Binding Proteins/antagonists & inhibitors , Transglutaminases/antagonists & inhibitors , Ventilator-Induced Lung Injury/enzymology , Acute Lung Injury/pathology , Animals , Cystamine/therapeutic use , Cytokines/analysis , Enzyme Inhibitors/therapeutic use , Enzyme-Linked Immunosorbent Assay , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Gene Expression , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Respiration, Artificial , Transglutaminases/genetics , Transglutaminases/metabolism , Ventilator-Induced Lung Injury/pathology , Ventilator-Induced Lung Injury/prevention & control
14.
Am J Respir Cell Mol Biol ; 51(2): 223-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24588101

ABSTRACT

We previously identified the intracellular nicotinamide phosphoribosyltransferase (iNAMPT, aka pre-B-cell colony enhancing factor) as a candidate gene promoting acute respiratory distress syndrome (ARDS) and ventilator-induced lung injury (VILI) with circulating nicotinamide phosphoribosyltransferase potently inducing NF-κB signaling in lung endothelium. iNAMPT also synthesizes intracellular nicotinamide adenine dinucleotide (iNAD) in response to extracellular oxidative stress, contributing to the inhibition of apoptosis via ill-defined mechanisms. We now further define the role of iNAMPT activity in the pathogenesis of ARDS/VILI using the selective iNAMPT inhibitor FK-866. C57/B6 mice were exposed to VILI (40 ml/kg, 4 h) or LPS (1.5 mg/kg, 18 h) after osmotic pump delivery of FK-866 (100 mg/kg/d, intraperitoneally). Assessment of total bronchoalveolar lavage (BAL) protein, polymorphonuclear neutrophil (PMN) levels, cytokine levels (TNF-α, IL-6, IL-1α), lung iNAD levels, and injury scores revealed that FK-866-mediated iNAMPT inhibition successfully reduced lung tissue iNAD levels, BAL injury indices, inflammatory cell infiltration, and lung injury scores in LPS- and VILI-exposed mice. FK-866 further increased lung PMN apoptosis, as reflected by caspase-3 activation in BAL PMNs. These findings support iNAMPT inhibition via FK-866 as a novel therapeutic agent for ARDS via enhanced apoptosis in inflammatory PMNs.


Subject(s)
Acrylamides/pharmacology , Anti-Inflammatory Agents/pharmacology , Cytokines/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Lung/drug effects , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Piperidines/pharmacology , Pneumonia/drug therapy , Respiratory Distress Syndrome/drug therapy , Ventilator-Induced Lung Injury/drug therapy , Animals , Apoptosis/drug effects , Bronchoalveolar Lavage Fluid/immunology , Caspase 3/metabolism , Cytokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Lung/enzymology , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , NAD/metabolism , Neutrophils/drug effects , Neutrophils/enzymology , Neutrophils/immunology , Nicotinamide Phosphoribosyltransferase/metabolism , Pneumonia/enzymology , Pneumonia/immunology , Pneumonia/pathology , Respiratory Distress Syndrome/enzymology , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/pathology
15.
Article in English | WPRIM (Western Pacific) | ID: wpr-216480

ABSTRACT

This study was performed to examine the role of transglutaminase 2 (TG2) in ventilator-induced lung injury (VILI). C57BL/6 mice were divided into six experimental groups: 1) control group; 2) lipopolysaccharide (LPS) group; 3) lung protective ventilation (LPV) group; 4) VILI group; 5) VILI with cystamine, a TG2 inhibitor, pretreatment (Cyst+VILI) group; and 6) LPV with cystamine pretreatment (Cyst+LPV) group. Acute lung injury (ALI) score, TG2 activity and gene expression, inflammatory cytokines, and nuclear factor-kappaB (NF-kappaB) activity were measured. TG2 activity and gene expression were significantly increased in the VILI group (P < 0.05). Cystamine pretreatment significantly decreased TG2 activity and gene expression in the Cyst+VILI group (P < 0.05). Inflammatory cytokines were higher in the VILI group than in the LPS and LPV groups (P < 0.05), and significantly lower in the Cyst+VILI group than the VILI group (P < 0.05). NF-kappaB activity was increased in the VILI group compared with the LPS and LPV groups (P < 0.05), and significantly decreased in the Cyst+VILI group compared to the VILI group (P = 0.029). The ALI score of the Cyst+VILI group was lower than the VILI group, but the difference was not statistically significant (P = 0.105). These results suggest potential roles of TG2 in the pathogenesis of VILI.


Subject(s)
Animals , Male , Mice , Acute Lung Injury/pathology , Cystamine/therapeutic use , Cytokines/analysis , Enzyme Inhibitors/therapeutic use , Enzyme-Linked Immunosorbent Assay , GTP-Binding Proteins/antagonists & inhibitors , Gene Expression , Lipopolysaccharides/toxicity , Mice, Inbred C57BL , NF-kappa B/metabolism , Respiration, Artificial , Transglutaminases/antagonists & inhibitors , Ventilator-Induced Lung Injury/enzymology
16.
Am J Physiol Lung Cell Mol Physiol ; 304(10): L689-700, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23525785

ABSTRACT

Ventilation at high tidal volume may cause lung inflammation and barrier dysfunction that culminates in ventilator-induced lung injury (VILI). However, the mechanisms by which mechanical stimulation triggers the inflammatory response have not been fully elucidated. This study tested the hypothesis that onset of VILI is triggered by activation of secretory group V phospholipase A(2) (gVPLA2) in pulmonary vascular endothelium exposed to excessive mechanical stretch. High-magnitude cyclic stretch (18% CS) increased expression and surface exposure of gVPLA2 in human pulmonary endothelial cells (EC). CS-induced gVPLA2 activation was required for activation of ICAM-1 expression and polymorphonuclear neutrophil (PMN) adhesion to CS-preconditioned EC. By contrast, physiological CS (5% CS) had no effect on gVPLA2 activation or EC-PMN adhesion. CS-induced ICAM-1 expression and EC-PMN adhesion were attenuated by the gVPLA2-blocking antibody (MCL-3G1), general inhibitor of soluble PLA2, LY311727, or siRNA-induced EC gVPLA2 knockdown. In vivo, ventilator-induced lung leukocyte recruitment, cell and protein accumulation in the alveolar space, and total lung myeloperoxidase activity were strongly suppressed in gVPLA2 mouse knockout model or upon administration of MCL-3G1. These results demonstrate a novel role for gVPLA2 as the downstream effector of pathological mechanical stretch leading to an inflammatory response associated with VILI.


Subject(s)
Acute Lung Injury/enzymology , Phospholipases A2/biosynthesis , Pneumonia/enzymology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Cells, Cultured , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Enzyme Induction , Humans , Intercellular Adhesion Molecule-1/metabolism , Leukocytes/enzymology , Leukocytes/metabolism , Leukocytes/pathology , Lung/enzymology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/enzymology , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia/metabolism , Pneumonia/pathology , Stress, Mechanical , Tidal Volume/physiology , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/pathology
17.
Biomaterials ; 34(1): 78-91, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23063297

ABSTRACT

Mechanical ventilation in patients may increase the risk of an acute lung injury (ALI), termed ventilator-induced lung injury (VILI). Induced pluripotent stem cells (iPSCs) have previously been shown to improve tissue repair in different disease models, including ALI. However, the therapeutic efficacy of iPSCs-derived conditioned medium (iPSC-CM) on ALI or VILI remains unknown. Here, we demonstrated that both iPSCs and iPSC-CM effectively decrease high-tidal-volume-induced VILI-related inflammatory processes and HMGB1 and PAI-1 production, predominantly through suppressing PI3K/Akt signaling. Notably, iPSC-CM suppressed production of macrophage inflammatory protein-2, malondialdehyde, and increased total glutathione content. Transmission electron microscopy revealed that iPSC-CM potentially restored the bronchial microstructure. This iPSC-CM efficacy could be mimicked by PI3K inhibitor LY294002 or Akt heterozygous knockout, and either treatment showed no further improvement on VILI in iPSC-CM recipients. Furthermore, iPSC-CM increased interferon gamma-induced protein 10 (IP-10) production in injured lungs. Administration of IP-10-neutralizing antibodies increased neutrophil infiltration, impaired lung oxygenation and deteriorated the protective effects mediated by iPSC-CM. Our data provide a preclinical indication regarding the therapeutic potential of iPSC-CM in VILI and suggest that inhibiting PI3K/Akt pathway or increasing IP-10 is a prospective diagnostic and therapeutic target for VILI patients.


Subject(s)
Chemokine CXCL9/metabolism , Culture Media, Conditioned/pharmacology , Induced Pluripotent Stem Cells/metabolism , Paracrine Communication/drug effects , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Ventilator-Induced Lung Injury/therapy , Animals , Heterozygote , Induced Pluripotent Stem Cells/drug effects , Inflammation/pathology , Inflammation/physiopathology , Lung/drug effects , Lung/pathology , Lung/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Stem Cell Transplantation , Tidal Volume/drug effects , Treatment Outcome , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/pathology , Ventilator-Induced Lung Injury/physiopathology
18.
Nat Med ; 18(8): 1217-23, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22820644

ABSTRACT

Sepsis, a systemic inflammatory response to infection, commonly progresses to acute lung injury (ALI), an inflammatory lung disease with high morbidity. We postulated that sepsis-associated ALI is initiated by degradation of the pulmonary endothelial glycocalyx, leading to neutrophil adherence and inflammation. Using intravital microscopy, we found that endotoxemia in mice rapidly induced pulmonary microvascular glycocalyx degradation via tumor necrosis factor-α (TNF-α)-dependent mechanisms. Glycocalyx degradation involved the specific loss of heparan sulfate and coincided with activation of endothelial heparanase, a TNF-α-responsive, heparan sulfate-specific glucuronidase. Glycocalyx degradation increased the availability of endothelial surface adhesion molecules to circulating microspheres and contributed to neutrophil adhesion. Heparanase inhibition prevented endotoxemia-associated glycocalyx loss and neutrophil adhesion and, accordingly, attenuated sepsis-induced ALI and mortality in mice. These findings are potentially relevant to human disease, as sepsis-associated respiratory failure in humans was associated with higher plasma heparan sulfate degradation activity; moreover, heparanase content was higher in human lung biopsies showing diffuse alveolar damage than in normal human lung tissue.


Subject(s)
Acute Lung Injury/physiopathology , Endotoxemia/complications , Glycocalyx/physiology , Lung/physiopathology , Neutrophils/physiology , Acute Lung Injury/etiology , Acute Lung Injury/prevention & control , Adoptive Transfer , Animals , Cell Adhesion/physiology , Disease Models, Animal , Endothelium/enzymology , Endothelium/physiology , Endotoxemia/physiopathology , Enzyme Activation , Gene Expression Regulation/drug effects , Glucuronidase/analysis , Glucuronidase/deficiency , Glucuronidase/physiology , Heparitin Sulfate/antagonists & inhibitors , Heparitin Sulfate/metabolism , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Intestinal Perforation/complications , Intestinal Perforation/microbiology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/pathology , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/physiology , Respiratory Insufficiency/enzymology , Respiratory Insufficiency/pathology , Tumor Necrosis Factor-alpha/physiology , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/pathology
19.
Shock ; 38(4): 375-80, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22777120

ABSTRACT

Mechanical ventilation (MV) has the potential to induce lung damage in healthy lungs or aggravate existing lung injury. Polymorphonuclear neutrophil (PMN) recruitment plays an important role in driving the inflammatory response in ventilator-induced lung injury (VILI). The cyclin-dependent kinase inhibitor r-roscovitine has been shown to induce apoptosis in PMNs. In this study, we investigated the potential of r-roscovitine treatment in reducing lung damage in a mouse model of VILI. Mice were tracheotomized and subjected to lung-protective MV with lower (∼7.5 mL/kg) or lung-injurious MV with higher (∼15 mL/kg) tidal volume (VT). R-roscovitine treatment enhanced apoptosis in PMNs in vitro. Ventilator-induced lung injury was associated with pulmonary PMN influx in low and high VT MV. During lung-injurious MV, r-roscovitine treatment reduced the number of PMNs and lowered levels of the lung damage markers RAGE (receptor for advanced glycation end products) and total immunoglobulin M in bronchoalveolar lavage fluid. R-roscovitine did not affect cytokine or chemokine levels in the bronchoalveolar space, neither during lung-protective nor lung-injurious MV. Thus, r-roscovitine treatment reduces lung damage in VILI, possibly dependent on increased apoptosis of PMNs.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Lung/enzymology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Ventilator-Induced Lung Injury/drug therapy , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cells, Cultured , Cyclin-Dependent Kinases/metabolism , Disease Models, Animal , Female , Humans , Lung/pathology , Male , Mice , Neutrophils/enzymology , Neutrophils/pathology , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Roscovitine , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/pathology , Ventilators, Mechanical/adverse effects
20.
Am J Respir Cell Mol Biol ; 46(4): 461-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22052879

ABSTRACT

Both hyperoxia and mechanical ventilation can independently cause lung injury. In combination, these insults produce accelerated and severe lung injury. We recently reported that pre-exposure to hyperoxia for 12 hours, followed by ventilation with large tidal volumes, induced significant lung injury and epithelial cell apoptosis compared with either stimulus alone. We also reported that such injury and apoptosis are inhibited by antioxidant treatment. In this study, we hypothesized that apoptosis signal-regulating kinase-1 (ASK-1), a redox-sensitive, mitogen-activated protein kinase kinase kinase, plays a role in lung injury and apoptosis in this model. To determine the role of ASK-1 in lung injury, the release of inflammatory mediators and apoptosis, attributable to 12 hours of hyperoxia, were followed by large tidal volume mechanical ventilation with hyperoxia. Wild-type and ASK-1 knockout mice were subjected to hyperoxia (Fi(O(2)) = 0.9) for 12 hours before 4 hours of large tidal mechanical ventilation (tidal volume = 25 µl/g) with hyperoxia, and were compared with nonventilated control mice. Lung injury, apoptosis, and cytokine release were measured. The deletion of ASK-1 significantly inhibited lung injury and apoptosis, but did not affect the release of inflammatory mediators, compared with the wild-type mice. ASK-1 is an important regulator of lung injury and apoptosis in this model. Further study is needed to determine the mechanism of lung injury and apoptosis by ASK-1 and its downstream mediators in the lung.


Subject(s)
MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , Ventilator-Induced Lung Injury/enzymology , Ventilator-Induced Lung Injury/prevention & control , Animals , Apoptosis/genetics , Cytokines/metabolism , Disease Models, Animal , Enzyme Activation , Epithelial Cells/pathology , Female , Hyperoxia/enzymology , Inflammation Mediators/metabolism , Male , Mice , Mice, Knockout , Pulmonary Alveoli/pathology , Ventilator-Induced Lung Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...