Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Shock ; 57(2): 298-308, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34107528

ABSTRACT

ABSTRACT: Ventilator-induced lung injury (VILI) can be life-threatening and it is important to prevent the development of VILI. It remains unclear whether the prone position affects neutrophilic inflammation in the lung regions in vivo, which plays a crucial role in the pathogenesis of VILI. This study aimed to assess the relationship between the use of the prone position and the development of VILI-associated regional neutrophilic lung inflammation. Regional neutrophilic lung inflammation and lung aeration during low tidal volume mechanical ventilation were assessed using in vivo 2-deoxy-2-[(18)F] fluoro-D-glucose (18F-FDG) positron emission tomography and computed tomography in acutely experimentally injured rabbit lungs (lung injury induced by lung lavage and excessive ventilation). Direct comparisons were made among three groups: control, supine, and prone positions. After approximately 7 h, tissue-normalized 18F-FDG uptake differed significantly between the supine and prone positions (SUP: 0.038 ±â€Š0.014 vs. PP: 0.029 ±â€Š0.008, P = 0.038), especially in the ventral region (SUP: 0.052 ±â€Š0.013 vs. PP: 0.026 ±â€Š0.007, P = 0.003). The use of the prone position reduced lung inhomogeneities, which was demonstrated by the correction of the disproportionate rate of voxel gas over the given lung region. The progression of neutrophilic inflammation was affected by the interaction between the total strain (for aeration) and the inhomogeneity. The prone position is effective in slowing down the progression of VILI-associated neutrophilic inflammation. Under low-tidal-volume ventilation, the main drivers of its effect may be homogenization of lung tissue and that of mechanical forces.


Subject(s)
Fluorodeoxyglucose F18 , Neutrophils , Pneumonia/diagnostic imaging , Pneumonia/immunology , Positron-Emission Tomography , Prone Position , Radiopharmaceuticals , Ventilator-Induced Lung Injury/diagnostic imaging , Ventilator-Induced Lung Injury/immunology , Animals , Disease Models, Animal , Male , Rabbits
2.
Am J Respir Crit Care Med ; 204(4): 421-430, 2021 08 15.
Article in English | MEDLINE | ID: mdl-33848447

ABSTRACT

Rationale: Mechanical ventilation is a mainstay of intensive care but contributes to the mortality of patients through ventilator-induced lung injury. eCypA (extracellular CypA [cyclophilin A]) is an emerging inflammatory mediator and metalloproteinase inducer, and the gene responsible for its expression has recently been linked to coronavirus disease (COVID-19). Objectives: To explore the involvement of eCypA in the pathophysiology of ventilator-induced lung injury. Methods: Mice were ventilated with a low or high Vt for up to 3 hours, with or without blockade of eCypA signaling, and lung injury and inflammation were evaluated. Human primary alveolar epithelial cells were exposed to in vitro stretching to explore the cellular source of eCypA, and CypA concentrations were measured in BAL fluid from patients with acute respiratory distress syndrome to evaluate the clinical relevance. Measurements and Main Results: High-Vt ventilation in mice provoked a rapid increase in soluble CypA concentration in the alveolar space but not in plasma. In vivo ventilation and in vitro stretching experiments indicated the alveolar epithelium as the likely major source. In vivo blockade of eCypA signaling substantially attenuated physiological dysfunction, macrophage activation, and MMPs (matrix metalloproteinases). Finally, we found that patients with acute respiratory distress syndrome showed markedly elevated concentrations of eCypA within BAL fluid. Conclusions: CypA is upregulated within the lungs of injuriously ventilated mice (and critically ill patients), where it plays a significant role in lung injury. eCypA represents an exciting novel target for pharmacological intervention.


Subject(s)
Anti-Inflammatory Agents/immunology , Cyclophilin A/immunology , Inflammation/immunology , Respiration, Artificial/adverse effects , Respiratory Distress Syndrome/immunology , Respiratory Mucosa/immunology , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/physiopathology , Animals , COVID-19/genetics , COVID-19/physiopathology , Cells, Cultured/drug effects , Cyclophilin A/pharmacology , Humans , Inflammation/physiopathology , Male , Mice , Models, Animal , Respiratory Distress Syndrome/physiopathology , SARS-CoV-2 , Ventilator-Induced Lung Injury/genetics
3.
Front Immunol ; 11: 207, 2020.
Article in English | MEDLINE | ID: mdl-32117318

ABSTRACT

Targeting inflammasome activation to modulate interleukin (IL)-1ß is a promising treatment strategy against acute respiratory distress syndrome and ventilator-induced lung injury (VILI). Autophagy is a key regulator of inflammasome activation in macrophages. Here, we investigated the role of autophagy in the development of acute lung injury (ALI) induced by lipopolysaccharide (LPS) and mechanical ventilation (MV). Two hours before starting MV, 0.2 mg/kg LPS was administered to mice intratracheally. Mice were then placed on high-volume MV (30 ml/kg with 3 cmH2O positive end-expiratory pressure for 2.5 h without additional oxygen application). Mice with myeloid-specific deletion of the autophagic protein ATG16L1 (Atg16l1fl/flLysMCre) suffered severe hypoxemia (adjusted p < 0.05) and increased lung permeability (p < 0.05, albumin level in bronchoalveolar lavage fluid) with significantly higher IL-1ß release into alveolar space (p < 0.05). Induction of autophagy by fasting-induced starvation led to improved arterial oxygenation (adjusted p < 0.0001) and lung permeability (p < 0.05), as well as significantly suppressed IL-1ß production (p < 0.01). Intratracheal treatment with anti-mouse IL-1ß monoclonal antibody (mAb; 2.5 mg/kg) significantly improved arterial oxygenation (adjusted p < 0.01) as well as lung permeability (p < 0.05). On the other hand, deletion of IL-1α gene or use of anti-mouse IL-1α mAb (2.5 mg/kg) provided no significant protection, suggesting that the LPS and MV-induced ALI is primarily dependent on IL-1ß, but independent of IL-1α. These observations suggest that autophagy has a protective role in controlling inflammasome activation and production of IL-1ß, which plays a critical role in developing hypoxemia and increased lung permeability in LPS plus MV-induced acute lung injury.


Subject(s)
Autophagy/physiology , Hypoxia/prevention & control , Inflammasomes/physiology , Interleukin-1beta/physiology , Lipopolysaccharides/toxicity , Lung/metabolism , Ventilator-Induced Lung Injury/etiology , Animals , Down-Regulation , Interleukin-18/physiology , Male , Mice , Mice, Inbred C57BL , Permeability , TOR Serine-Threonine Kinases/physiology , Trehalose/therapeutic use , Ventilator-Induced Lung Injury/immunology
4.
Int Immunopharmacol ; 78: 106069, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31841755

ABSTRACT

Inflammation plays a criticalrole in the development of ventilator-induced lung injury (VILI). Endoplasmic reticulum (ER) stress is associated with a variety of diseases through the modulation of inflammatory responses. However, little is known about how ER stress is implicated in VILI. In this study, murine mechanical ventilation models were constructed. Total protein and inflammatory cytokines were measured in bronchoalveolar lavage fluid (BALF),and lung tissue injurywasassessedby histology. Our data revealed that mice subjected to high tidal ventilation (TV) for 4 h showed more severe pulmonary edema and inflammation than those of mice with spontaneous breathing and low TV-treatment. In addition, the high TV-treated animals upregulated the ER stress markers GRP78, CHOP, p-IRE1α, TRAF2, and p-NF-κB expression at both the mRNA and protein levels in lung tissue. Administration of thapsigargin exacerbated the histological changes, inflammation and expression of GRP78 and CHOP after high TV, but treatment with ER stress and IRE1α kinase inhibitors attenuated the pathological damage and downregulated the high expression of GRP78, CHOP, p-IRE1α, TRAF2, and p-NF-κB, suggesting that ER stress is involved in VILI though the IRE1α/TRAF2/NF-κB signaling pathway in mice.


Subject(s)
Endoplasmic Reticulum Stress/immunology , Inflammation/immunology , Signal Transduction/immunology , Ventilator-Induced Lung Injury/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/metabolism , Humans , Inflammation/diagnosis , Inflammation/drug therapy , Inflammation/pathology , Lung/immunology , Lung/pathology , Male , Mice , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Severity of Illness Index , Signal Transduction/drug effects , TNF Receptor-Associated Factor 2/metabolism , Up-Regulation/immunology , Ventilator-Induced Lung Injury/diagnosis , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/pathology
5.
Int Immunopharmacol ; 78: 106015, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31780369

ABSTRACT

Lymphocyte antigen 6Chigh (Ly-6Chigh) inflammatory monocytes, as novel mononuclear cells in the innate immune system, participate in infectious diseases. In this study, we investigated the potential role of these monocytes in ventilator-induced lung injury (VILI) and the possible mechanism involved in their migration to lung tissue. Our results showed that mechanical ventilation with high tidal volume (HTV) increased the accumulation of Ly-6Chigh inflammatory monocytes in lung tissues and that blocking C­C chemokine receptor 2 (CCR2) could significantly reduce Ly-6Chigh inflammatory-monocyte migration and attenuate the degree of inflammation of lung tissues. In addition, inhibition of p38 mitogen-activated protein kinase (p38 MAPK) activity could decrease the secretion of monocyte chemoattractant protein 1 (MCP-1), which in turn decreased the migration of Ly-6Chigh inflammatory monocytes into lung tissue. We also demonstrated that high ventilation caused Ly-6Chigh inflammatory monocytes in the bone marrow to migrate into and aggregate in the lungs, creating inflammation, and that the mechanism was quite different from that of infectious diseases. Ly-6Chigh inflammatory monocytes might play a pro-inflammatory role in VILI, and blocking their infiltration into lung tissue might become a new target for the treatment of this injury.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chemokine CCL2/metabolism , Monocytes/immunology , Ventilator-Induced Lung Injury/immunology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antigens, Ly/metabolism , Benzoxazines/pharmacology , Benzoxazines/therapeutic use , Bone Marrow/immunology , Bone Marrow/pathology , Cell Movement/drug effects , Cell Movement/immunology , Disease Models, Animal , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Lung/cytology , Lung/immunology , Lung/pathology , Mice , Monocytes/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/metabolism , Spiro Compounds/pharmacology , Spiro Compounds/therapeutic use , Tidal Volume , Ventilator-Induced Lung Injury/diagnosis , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/pathology , Ventilators, Mechanical/adverse effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
6.
Int Immunopharmacol ; 75: 105795, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31421547

ABSTRACT

Mechanical ventilation (MV) is an essential life-support technique, but it can induce ventilator-induced lung injury (VILI) and subsequent pulmonary fibrosis. The mechanisms underlying this fibrosis are largely unknown. Because excessive polarization of M2 macrophages has increasingly been cited as possible inciting factor for tissue remodeling and organ fibrosis, we here hypothesize it might be involved in the development of pulmonary fibrosis after high tidal volume (VT) MV. In our prospective, randomized, controlled animal study, C57BL/6 mice were randomly placed in either a VILI group or sham group. After ventilation, surviving mice were allowed to recover for 0, 1, 3, 5, 7, or 14 days. 200 mice were involved in our in vivo experiment, and the results calculated here refer only to the surviving mice. The results clearly showed that high-VT MV caused early inflammation and a subsequent fibroproliferative response in mice without pre-existing lung disease. High-VT MV was also found to lead to a dramatic increase in the number of M2 macrophages in mouse bronchoalveolar lavage fluid (BALF) cell and lung tissues. Consistent with the progression of fibrosis, there were far more M2 macrophages at the 5th day after ventilation and remained dominant for 2 weeks. High-VT MV induced epithelial-mesenchymal transition (EMT) on day 7, accompanied by the increased expression of TGF-ß1 and p-Smad2/3. In vitro experiments, the co-culture of M2 macrophage and MLE-12 cells resulted in a significant EMT and upregulation of TGF-ß1 and p-Smad2/3 in MLE-12 cells. To summarize, our findings suggested the persistent tilt polarization toward M2 macrophages was associated with EMT during the course of ventilator-induced pulmonary fibrosis, which may play its roles through activation of epithelial TGF-ß1/Smad2/3 signaling.


Subject(s)
Epithelial-Mesenchymal Transition , Macrophages/immunology , Pulmonary Fibrosis/immunology , Respiration, Artificial/adverse effects , Ventilator-Induced Lung Injury/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Coculture Techniques , Lung/immunology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/pathology , Tidal Volume , Ventilator-Induced Lung Injury/pathology
7.
Int Immunopharmacol ; 75: 105825, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31437789

ABSTRACT

Mechanical ventilation (MV) is a major support for patients with severe clinical disease, surgery and anesthesia. However, complications of mechanical ventilation especially ventilator-induced lung injury(VILI) can make the course and prognosis worse. Resolvin D1(RvD1) is a class of endogenous polyunsaturated fatty acid derivative, which has protective effects on various pulmonary inflammatory diseases. However, the mechanism of RvD1 in the process of VILI has not been fully elucidated. Our study found that RvD1 does have a protective effect on VILI including inhibiting inflammatory responses, reducing tissue damage and improving pulmonary function. The protective effect of RvD1 is positively related to its dose. Our research suggested RvD1 plays a role that increases the expression of heme oxygenase­1 (HO-1) and decreases the expression of the high mobility group chromosomal protein B1 (HMGB-1) in VILI. HO-1 can exert the protective effect of organism through multiple mechanisms such as anti-inflammatory, anti-oxidation, anti-apoptosis, etc. HMGB1 is a potent inflammatory response factor in the body, which can aggravate the inflammatory response of the body. Our study demonstrated that RvD1 can ameliorate lung inflammation and reduce pathological changes in lung tissue in a model of lung injury induced by mechanical ventilation. The protective role of RvD1 is closely linked to the increased expression of HO-1 and the decreased expression of HMGB1. Moreover, we found that RvD1 can increase the expression of Nrf2 and inhibit the expression of NF-κB. We found the specific inhibitor of HO-1, ZnPP, can significantly inhibit the protective role of RvD1 in VILI. When HO-1 is inhibited, pathological damage and inflammatory reaction in the lungs are considerably aggravated, and pulmonary function is significantly weakened. In addition, the expression of HMGB1 is drastically increased. This indicates that the HO-1-HMGB1 pathway plays an important role in the protective effect of RvD1 on mechanical ventilation lung injury.


Subject(s)
Docosahexaenoic Acids/pharmacology , HMGB1 Protein/immunology , Heme Oxygenase-1/immunology , Membrane Proteins/immunology , Protective Agents/pharmacology , Ventilator-Induced Lung Injury/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Docosahexaenoic Acids/therapeutic use , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Mice, Inbred C57BL , Protective Agents/therapeutic use , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/pathology , Ventilators, Mechanical/adverse effects
8.
Eur Rev Med Pharmacol Sci ; 23(11): 5004-5011, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31210340

ABSTRACT

OBJECTIVE: Ventilator-induced lung injury (VILI) remains a challenge. This study was designed to investigate the effects of ambroxol on VILI and the underlying mechanisms in a rodent model. MATERIALS AND METHODS: Male Wistar rats weighing 310-380 g were divided into four groups (n=8 per group): 1) saline only, 2) ventilation plus saline, 3) ventilation plus ambroxol (2 mg/kg), and 4) ventilation plus ambroxol (50 mg/kg). Rats in groups 1 and 2 were treated (i.p.) with 2.5 ml of saline once a day for six days and last injected 1 h prior to tracheotomy. Rats in groups 3 and 4 received ambroxol on the same schedule. Rats were ventilated for 90 minutes at a tidal volume (VT) of 30 ml/kg. The expression levels of c-Jun, a component of activator protein-1 (AP-1), and gamma-glutamylcysteine synthetase (γ-GCS), the rate-limiting enzyme in the synthesis of glutathione (gamma-glutamyl-cysteinyl-glycine, GSH), an endogenous antioxidant, were measured with immunohistochemical staining and in situ hybridization. Both AP-1 and GSH are involved in VILI. RESULTS: Ambroxol at 50 mg/kg inhibited ventilation-induced lung inflammation, significantly elevated the ventilation-induced down-regulation of γ-GCS mRNA and protein, and significantly decreased the ventilation-induced up-regulation of c-Jun mRNA and protein. It has been reported that reactive oxygen species (ROS) can activate AP-1, leading to the production of pro-inflammatory cytokines and lung inflammation. CONCLUSIONS: Ambroxol increases γ-GCS to promote GSH production, which in turn, inhibits ROS-dependent AP-1 activation and inflammation.


Subject(s)
Ambroxol/pharmacology , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Transcription Factor AP-1/metabolism , Ventilator-Induced Lung Injury/drug therapy , Ambroxol/therapeutic use , Animals , Disease Models, Animal , Down-Regulation/drug effects , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Proto-Oncogene Proteins c-jun/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Up-Regulation/drug effects , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/pathology
9.
Int Immunopharmacol ; 72: 74-81, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30959374

ABSTRACT

Microvesicles (MVs) have been extensively identified in various biological fluids including bronchoalveolar lavage fluid (BALF), peripheral blood and ascitic fluids. Our previous study showed that MVs are responsible for acute lung injury, but the exact mechanism underlying MVs formation remains poorly understood. In the present study, we investigate the potential role of RhoA/Rock signaling in MVs generation and the biological activity of MVs in ventilator-induced lung injury (VILI). Our results revealed that high tide ventilation induced super MVs releasing into the lung and subsequently caused lung inflammation. Strikingly, intratracheal instillation of MVs that isolated from highly ventilated mice triggered significant lung inflammation in naive mice. The MVs production is strongly correlated with lung inflammation and the upregulation of RhoA, Rock and phospho-Limk (phosphorylation of Limk is the activated form). RhoA inhibitor decreased the expression of Rock and the phosphorylation of Limk, decreased MVs production and alleviated lung inflammation. Rock inhibitor also decreased the phosphorylation of Limk, decreased MVs production and alleviated lung inflammation. Our data demonstrated that the production of MVs requires RhoA/Rock signaling, and VILI might be potentially prevented by targeting RhoA/Rock signaling pathway.


Subject(s)
ADP Ribose Transferases/therapeutic use , Botulinum Toxins/therapeutic use , Cell-Derived Microparticles/drug effects , Ventilator-Induced Lung Injury/drug therapy , rhoA GTP-Binding Protein/antagonists & inhibitors , Amides/therapeutic use , Animals , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Mice, Inbred C57BL , Pyridines/therapeutic use , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/pathology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/immunology , rhoA GTP-Binding Protein/immunology
10.
Am J Respir Cell Mol Biol ; 60(5): 569-577, 2019 05.
Article in English | MEDLINE | ID: mdl-30428271

ABSTRACT

The aim of this study was to assess the association between regional tidal volume (Vt), regional functional residual capacity (FRC), and the expression of genes linked with ventilator-induced lung injury. Two groups of BALB/c mice (n = 8 per group) were ventilated for 2 hours using a protective or injurious ventilation strategy, with free-breathing mice used as control animals. Regional Vt and FRC of the ventilated mice was determined by analysis of high-resolution four-dimensional computed tomographic images taken at baseline and after 2 hours of ventilation and corrected for the volume of the region (i.e., specific [s]Vt and specific [s]FRC). RNA concentrations of 21 genes in 10 different lung regions were quantified using a quantitative PCR array. sFRC at baseline varied regionally, independent of ventilation strategy, whereas sVt varied regionally depending on ventilation strategy. The expression of IL-6 (P = 0.04), Ccl2 (P < 0.01), and Ang-2 (P < 0.05) was associated with sVt but not sFRC. The expression of seven other genes varied regionally (IL-1ß and RAGE [receptor for advanced glycation end products]) or depended on ventilation strategy (Nfe2l2 [nuclear factor erythroid-derived 2 factor 2], c-fos, and Wnt1) or both (TNF-α and Cxcl2), but it was not associated with regional sFRC or sVt. These observations suggest that regional inflammatory responses to mechanical ventilation are driven primarily by tidal stretch.


Subject(s)
Biomechanical Phenomena/immunology , Gene Expression Regulation/immunology , Lung/immunology , Respiration, Artificial/methods , Ventilator-Induced Lung Injury/genetics , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CXCL2/genetics , Chemokine CXCL2/immunology , Four-Dimensional Computed Tomography , Image Interpretation, Computer-Assisted , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Lung/diagnostic imaging , Lung/physiopathology , Male , Mice , Mice, Inbred BALB C , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/immunology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/immunology , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/immunology , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/immunology , Signal Transduction , Tidal Volume/genetics , Tidal Volume/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Ventilator-Induced Lung Injury/diagnostic imaging , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/physiopathology , Wnt1 Protein/genetics , Wnt1 Protein/immunology
11.
Int Immunopharmacol ; 63: 74-83, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30075431

ABSTRACT

Microvesicles shed from pulmonary cells are capable of transferring inflammatory cargo to recipient cells nearby or in distant to enhance inflammation. Some authors believe that cofilin controls actin dynamics and regulates vesicle mobilization. We therefore investigated the potential role and mechanism of microvesicles in ventilator-induced lung injury (VILI). Fifty male C57BL/6 mice were orotracheally intubated and either allowed to breathe spontaneously or they were mechanically ventilated with different tidal volumes (Vt) and ventilation times. Lung tissue injury was assessed in terms of lung histopathologic examination, wet/dry weight ratios, and levels of total proteins and of cytokines. Microvesicle characteristics, sizes, contents and levels as well as cofilin were also measured. We found that lung inflammation increased significantly after ventilation with high Vt for 4 h; these conditions led to secretion of larger and more microvesicles into the alveoli than animals with/without ventilation at low Vt. Intratracheal instillation of microvesicles obtained from animals ventilated with low or high Vt triggered significant lung inflammation in naive mice, and these high-Vt microvesicles not only carried more IL-1ß and TNF-α but also induced more severe lung inflammation compared to low-Vt microvesicles; And high-Vt microvesicles at 2 h carried more molecular cargo than that at 1 h or 4 h, which may involve the shift and amplification of inflammation. Furthermore, blocking the phosphorylation of cofilin can not only inhibit microvesicle formation in the lung, but also reduce lung injury. Collectively, our data suggest that microvesicles packaging IL-1ß and TNF-α enhance lung inflammation in VILI.


Subject(s)
Actin Depolymerizing Factors/immunology , Cell-Derived Microparticles/immunology , Interleukin-1beta/immunology , Tumor Necrosis Factor-alpha/immunology , Ventilator-Induced Lung Injury/immunology , Animals , Male , Mice, Inbred C57BL , Signal Transduction , Ventilator-Induced Lung Injury/pathology
12.
J Immunol Res ; 2018: 6963754, 2018.
Article in English | MEDLINE | ID: mdl-29850636

ABSTRACT

Ventilator-induced lung injury is well recognized, and appropriate arterial saturation target is unknown, so gentle modes of ventilation and minimizing oxidative stress have been well studied. Our objective was to analyze any association between the oxygen levels at blood sampling and plasma levels of the interleukins IL-6, IL-1ß, IL-10, and IL-8 and TNF-α in preterm newborns under mechanical ventilation (MV) in their first two days. Methods. Prospective cohort including neonates with severe respiratory distress. Blood samples were collected right before and 2 hours after invasive MV. For analysis purposes, newborns were separated according to oxygen requirement: low oxygen (≤30%) and high oxygen (>30%) groups. Interleukins were measured using a commercially available kit. Results. 20 neonates (gestational age 32.2 ± 3 weeks) were evaluated. Median O2 saturation levels pre-MV were not different in both oxygen groups. In the high oxygen group, IL-6, IL-8, and TNF-α plasma levels increased significantly after two hours under MV. Conclusions. Despite the small sample studied, data showed that there is a relationship between VILI, proinflammatory cytokines, and oxygen-induced lung injury, but a study considering oxidative marker measurements is needed. It seems that less oxygen may keep safer saturation targets playing a less harmful role.


Subject(s)
Oxidative Stress , Oxygen/physiology , Respiratory Distress Syndrome, Newborn/immunology , Ventilator-Induced Lung Injury/immunology , Cohort Studies , Cytokines/blood , Cytokines/metabolism , Female , Humans , Infant, Newborn , Inflammation Mediators/metabolism , Lung/metabolism , Lung/pathology , Male , Prospective Studies , Respiration, Artificial
13.
Chin Med J (Engl) ; 131(7): 804-812, 2018 Apr 05.
Article in English | MEDLINE | ID: mdl-29578124

ABSTRACT

BACKGROUND: Ventilator-induced lung injury (VILI) is commonly associated with barrier dysfunction and inflammation reaction. Glutamine could ameliorate VILI, but its role has not been fully elucidated. This study examined the relationship between inflammatory cytokines (interleukin [IL]-6, tumor necrosis factor [TNF]-α, and IL-10) and adherens junctions (E-cadherin, p120-catenin), which were ameliorated by glutamine in VILI, both in vitro and in vivo. METHODS: For the in vivo study, 30 healthy C57BL/6 mice weighing 25-30 g were randomly divided into five groups with random number table (n = 6 in each group): control (Group C); low tidal volume (Group L); low tidal volume + glutamine (Group L + G); high tidal volume (Group H); and high tidal volume + glutamine (Group H + G). Mice in all groups, except Group C, underwent mechanical ventilation for 4 h. For the in vitro study, mouse lung epithelial 12 (MLE-12) cells pretreated with glutamine underwent cyclic stretching at 20% for 4 h. Cell lysate and lung tissue were obtained to detect the junction proteins, inflammatory cytokines, and lung pathological changes by the Western blotting, cytokine assay, hematoxylin and eosin staining, and immunofluorescence. RESULTS: In vivo, compared with Group C, total cell counts (t = -28.182, P < 0.01), the percentage of neutrophils (t = -28.095, P < 0.01), IL-6 (t = -28.296, P < 0.01), and TNF-α (t = -19.812, P < 0.01) in bronchoalveolar lavage (BAL) fluid, lung injury scores (t = -6.708, P < 0.01), and the wet-to-dry ratio (t = -15.595, P < 0.01) were increased in Group H; IL-10 in BAL fluid (t = 9.093, P < 0.01) and the expression of E-cadherin (t = 10.044, P < 0.01) and p120-catenin (t = 13.218, P < 0.01) were decreased in Group H. Compared with Group H, total cell counts (t = 14.844, P < 0.01), the percentage of neutrophils (t = 18.077, P < 0.01), IL-6 (t = 18.007, P < 0.01), and TNF-α (t = 10.171, P < 0.01) in BAL fluid were decreased in Group H + G; IL-10 in BAL fluid (t = -7.531, P < 0.01) and the expression of E-cadherin (t = -14.814, P < 0.01) and p120-catenin (t = -9.114, P < 0.01) were increased in Group H + G. In vitro, compared with the nonstretching group, the levels of IL-6 (t = -21.111, P < 0.01) and TNF-α (t = -15.270, P < 0.01) were increased in the 20% cyclic stretching group; the levels of IL-10 (t = 5.450, P < 0.01) and the expression of E-cadherin (t = 17.736, P < 0.01) and p120-catenin (t = 16.136, P < 0.01) were decreased in the 20% cyclic stretching group. Compared with the stretching group, the levels of IL-6 (t = 11.818, P < 0.01) and TNF-α (t = 8.631, P < 0.01) decreased in the glutamine group; the levels of IL-10 (t = -3.203, P < 0.05) and the expression of E-cadherin (t = -13.567, P < 0.01) and p120-catenin (t = -10.013, P < 0.01) were increased in the glutamine group. CONCLUSIONS: High tidal volume mechanical ventilation and 20% cyclic stretching could cause VILI. Glutamine regulates VILI by improving cytokines and increasing the adherens junctions, protein E-cadherin and p120-catenin, to enhance the epithelial barrier function.


Subject(s)
Cadherins/metabolism , Glutamine/metabolism , Lung/metabolism , Ventilator-Induced Lung Injury/metabolism , Animals , Catenins/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Ventilator-Induced Lung Injury/immunology , Delta Catenin
14.
Sci Rep ; 7(1): 14704, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29089616

ABSTRACT

Mechanical ventilation of preterm neonates causes lung inflammation and injury, with potential life-long consequences. Inert 50-nm polystyrene nanoparticles (PS50G) reduce allergic inflammation in the lungs of adult mice. We aimed to confirm the anti-inflammatory effects of PS50G in a sheep asthma model, and investigate the effects of prophylactic administration of PS50G on ventilation-induced lung injury (VILI) in preterm lambs. We assessed lung inflammatory cell infiltration, with and without PS50G, after airway allergen challenge in ewes sensitised to house dust mite. Preterm lambs (0.83 gestation) were delivered by caesarean section for immediate tissue collection (n = 5) or ventilation either with (n = 6) or without (n = 5) prophylactic intra-tracheal administration of PS50G nanoparticles (3% in 2 ml). Ventilation was continued for a total of 2 h before tissue collection for histological and biomolecular assessment of lung injury and inflammation. In ewes with experimental asthma, PS50G decreased eosinophilic infiltration of the lungs. Ventilated preterm lambs showed molecular and histological signs of lung injury and inflammation, which were exacerbated in lambs that received PSG50G. PS50G treatment decreased established inflammation in the lungs of asthmatic sheep. However, prophylactic administration of PSG50 exacerbated ventilation-induced lung injury and lung inflammation in preterm lambs.


Subject(s)
Asthma/therapy , Eosinophils/immunology , Lung/immunology , Nanoparticles/adverse effects , Pneumonia/immunology , Polystyrenes/adverse effects , Ventilator-Induced Lung Injury/immunology , Animals , Animals, Newborn , Antigens, Dermatophagoides/immunology , Cattle , Disease Models, Animal , Disease Progression , Humans , Immunization , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Polystyrenes/administration & dosage , Polystyrenes/chemistry , Pulmonary Ventilation , Pyroglyphidae/immunology , Sheep
15.
Biomed Res Int ; 2017: 8272504, 2017.
Article in English | MEDLINE | ID: mdl-29387725

ABSTRACT

The pathogenesis of ventilator-induced lung injury (VILI) is associated with neutrophils. Neutrophils release neutrophil extracellular traps (NETs), which are composed of DNA and granular proteins. However, the role of NETs in VILI remains incompletely understood. Normal saline and deoxyribonuclease (DNase) were used to study the role of NETs in VILI. To further determine the role of Toll-like receptor 4 (TLR4) in NETosis, we evaluated the lung injury and NET formation in TLR4 knockout mice and wild-type mice that were mechanically ventilated. Some measures of lung injury and the NETs markers were significantly increased in the VILI group. DNase treatment markedly reduced NETs markers and lung injury. After high-tidal mechanical ventilation, the NETs markers in the TLR4 KO mice were significantly lower than in the WT mice. These data suggest that NETs are generated in VILI and pathogenic in a mouse model of VILI, and their formation is partially dependent on TLR4.


Subject(s)
Extracellular Traps/immunology , Toll-Like Receptor 4/immunology , Ventilator-Induced Lung Injury/immunology , Animals , Deoxyribonucleases/pharmacology , Extracellular Traps/genetics , Mice , Mice, Knockout , Toll-Like Receptor 4/genetics , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/genetics , Ventilator-Induced Lung Injury/pathology
16.
Sci Rep ; 6: 30943, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27476670

ABSTRACT

Bacterial pneumonia is a major cause of acute lung injury and acute respiratory distress syndrome, characterized by alveolar barrier disruption. NLRP3 is best known for its ability to form inflammasomes and to regulate IL-1ß and IL-18 production in myeloid cells. Here we show that NLRP3 protects the integrity of the alveolar barrier in a mouse model of Streptococcus pneumoniae-induced pneumonia, and ex vivo upon treatment of isolated perfused and ventilated lungs with the purified bacterial toxin, pneumolysin. We reveal that the preserving effect of NLRP3 on the lung barrier is independent of inflammasomes, IL-1ß and IL-18. NLRP3 improves the integrity of alveolar epithelial cell monolayers by enhancing cellular adherence. Collectively, our study uncovers a novel function of NLRP3 by demonstrating that it protects epithelial barrier function independently of inflammasomes.


Subject(s)
Epithelial Cells/immunology , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Pneumonia, Pneumococcal/prevention & control , Pulmonary Alveoli/immunology , Ventilator-Induced Lung Injury/prevention & control , Animals , Bacterial Proteins/metabolism , Cell Adhesion , Cells, Cultured , Disease Models, Animal , Epithelial Cells/microbiology , Epithelial Cells/pathology , Female , Interleukin-18/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/microbiology , Pulmonary Alveoli/microbiology , Pulmonary Alveoli/pathology , Signal Transduction , Streptococcus pneumoniae/pathogenicity , Streptolysins/metabolism , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/microbiology
17.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L208-18, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27288491

ABSTRACT

Animal models play a critical role in the study of acute respiratory distress syndrome (ARDS) and ventilator-induced lung injury (VILI). One limitation has been the lack of a suitable method for serial assessment of acute lung injury (ALI) in vivo. In this study, we demonstrate the sensitivity of magnetic resonance imaging (MRI) to assess ALI in real time in rat models of VILI. Sprague-Dawley rats were untreated or treated with intratracheal lipopolysaccharide or PBS. After 48 h, animals were mechanically ventilated for up to 15 h to induce VILI. Free induction decay (FID)-projection images were made hourly. Image data were collected continuously for 30 min and divided into 13 phases of the ventilatory cycle to make cinematic images. Interleaved measurements of respiratory mechanics were performed using a flexiVent ventilator. The degree of lung infiltration was quantified in serial images throughout the progression or resolution of VILI. MRI detected VILI significantly earlier (3.8 ± 1.6 h) than it was detected by altered lung mechanics (9.5 ± 3.9 h, P = 0.0156). Animals with VILI had a significant increase in the Index of Infiltration (P = 0.0027), and early regional lung infiltrates detected by MRI correlated with edema and inflammatory lung injury on histopathology. We were also able to visualize and quantify regression of VILI in real time upon institution of protective mechanical ventilation. Magnetic resonance lung imaging can be utilized to investigate mechanisms underlying the development and propagation of ALI, and to test the therapeutic effects of new treatments and ventilator strategies on the resolution of ALI.


Subject(s)
Ventilator-Induced Lung Injury/diagnostic imaging , Airway Resistance , Animals , Lipopolysaccharides/pharmacology , Lung/diagnostic imaging , Lung/immunology , Lung/physiopathology , Magnetic Resonance Imaging , Male , Neutrophil Infiltration , Rats, Sprague-Dawley , Respiration, Artificial , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/physiopathology
18.
Am J Physiol Lung Cell Mol Physiol ; 310(11): L1062-70, 2016 06 01.
Article in English | MEDLINE | ID: mdl-26993523

ABSTRACT

Ventilator-induced lung injury (VILI) is associated with activated inflammatory signaling, such as cytokine production by endothelial and epithelial cells and macrophages, although the precise mechanisms of inflammatory activation induced by VILI-relevant cyclic stretch (CS) amplitude remain poorly understood. We show that exposure of human pulmonary endothelial cells (EC) to chronic CS at 18% linear distension (18% CS), but not at physiologically relevant 5% CS, induces "EC-activated phenotype," which is characterized by time-dependent increase in ICAM1 and VCAM1 expression. A preconditioning of 18% CS also increased in a time-dependent fashion the release of soluble ICAM1 (sICAM1) and IL-8. Investigation of potential signaling mechanisms of CS-induced EC inflammatory activation showed that 18% CS, but not 5% CS, induced time-dependent upregulation of VEGF receptor 2 (VEGFR2), as monitored by increased protein expression and VEGFR2 tyrosine phosphorylation. Both CS-induced VEGFR2 expression and tyrosine phosphorylation were abrogated by cotreatment with reactive oxygen species inhibitor, N-acetyl cysteine. Molecular inhibition of VEGFR2 expression by gene-specific siRNA or treatment with VEGFR2 pharmacological inhibitor SU-1498 attenuated CS-induced activation of ICAM1 and VCAM1 expression and sICAM1 release. Chronic EC preconditioning at 18% CS augmented EC inflammation and barrier-disruptive response induced by proinflammatory cytokine TNF-α. This effect of chronic 18% CS preconditioning was attenuated by siRNA-induced VEGFR2 knockdown. This study demonstrates for the first time a VEGFR2-dependent mechanism of EC inflammatory activation induced by pathological CS. We conclude that, despite the recognized role of VEGF as a prosurvival and angiogenic factor, excessive activation of VEGFR2 signaling by high-tidal-volume lung mechanical ventilation may contribute to ventilator-induced (biotrauma) lung inflammation and barrier dysfunction by augmenting cell response to VILI-associated inflammatory mediators.


Subject(s)
Endothelial Cells/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Ventilator-Induced Lung Injury/immunology , Cells, Cultured , Humans , Intercellular Adhesion Molecule-1/metabolism , Interleukin-8/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Ventilator-Induced Lung Injury/metabolism
19.
J Physiol Pharmacol ; 67(6): 911-918, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28195072

ABSTRACT

The complement system contributes to ventilator induced lung injury (VILI). We hypothesized that pretreatment with the C1 esterase inhibitor (C1INH) Berinert® constrains complement activation consecutively inducing improvements in arterial oxygenation and histological pulmonary damage. At baseline, male Sprague-Dawley rats underwent mechanical ventilation in a conventional mode (PIP 13 cm H2O, PEEP 3 cm H2O). In the Control group, the ventilator setting was maintained (Control, n = 15). The other animals randomly received intravenous pretreatment with either 100 units/kg of the C1-INH Berinert® (VILI-C1INH group, n = 15) or 1 ml saline solution (VILI-C group, n = 15). VILI was induced by invasive ventilation (PIP 35 cm H2O, PEEP 0 cm H2O). After two hours of mechanical ventilation, the complement component C3a remained low in the Control group (258 ± 82 ng/ml) but increased in both VILI groups (VILI-C: 1017 ± 283 ng/ml; VILIC1INH: 817 ± 293 ng/ml; P < 0.05 for both VILI groups versus Control). VILI caused a profound deterioration of arterial oxygen tension (VILI-C: 193 ± 167 mmHg; VILI/C1-INH: 154 ± 115 mmHg), whereas arterial oxygen tension remained unaltered in the Control group (569 ± 26 mmHg; P < 0.05 versus both VILI groups). Histological investigation revealed prominent overdistension and interstitial edema in both VILI groups compared to the Control group. C3a plasma level in the VILI group were inversely correlated with arterial oxygen tension (R = -0.734; P < 0.001). We conclude that in our animal model of VILI the complement system was activated in parallel with the impairment in arterial oxygenation and that pretreatment with 100 units/kg Berinert® did neither prevent systemic complement activation nor lung injury.


Subject(s)
Complement Activation/immunology , Lung/immunology , Ventilator-Induced Lung Injury/immunology , Animals , Arteries/drug effects , Arteries/immunology , Complement Activation/drug effects , Complement C1 Inhibitor Protein/pharmacology , Complement C3a/immunology , Disease Models, Animal , Male , Oxygen/immunology , Rats , Rats, Sprague-Dawley , Respiration, Artificial/methods
20.
Eur J Pharmacol ; 769: 333-41, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26610718

ABSTRACT

Hesperetin, a major bioflavonoid in sweet oranges and lemons, has been reported to have anti-inflammatory properties. However, the effect of hesperetin on ventilator-induced acute lung injury has not been studied. In present study, we investigated the protective effect of hesperetin on ventilator-induced acute lung injury in rats. Rats were orally administered hesperetin (10, 20, or 40mg/kg) two hour before acute lung injury was induced by mechanical ventilation. Rats were then randomly divided into six groups: the lung protective ventilation group (n=20, LV group), injurious ventilation group (n=20, HV group), vehicle-treated injurious ventilation group (n=20, LV+vehicle group), hesperetin (10mg/kg)-treated acute lung injury group (n=20, HV+Hsp (10mg)), hesperetin (20mg/kg)-treated acute lung injury group (n=20, HV+Hsp (20mg)), and hesperetin (40mg/kg)-treated acute lung injury group (n=20, HV+Hsp (40mg)). The lung tissues and bronchoalveolar lavage fluid were isolated for subsequent measurements. Treatment with hesperetin dramatically improved the histology of lung tissue, and reduced the wet/dry ratio, myeloperoxidase activity, protein concentration, and production of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1ß, and MIP-2 in the bronchoalveolar lavage fluid of rats with ventilator-induced acute lung injury. Additionally, our study indicated that this protective effect of hesperetin results from its ability to increase the expression of peroxisome proliferator-activated receptor (PPAR)-γ and inhibit the activation of the nuclear factor (NF)-κB pathway. These results suggest that hesperetin may be a potential novel therapeutic candidate for protection against ventilator-induced acute lung injury.


Subject(s)
Hesperidin/pharmacology , Transcription Factor RelA/metabolism , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/metabolism , Animals , Bronchoalveolar Lavage Fluid , Cell Survival/drug effects , Cytokines/biosynthesis , Cytokines/metabolism , Gene Expression Regulation/drug effects , Hesperidin/therapeutic use , Inflammation/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages/drug effects , Male , PPAR gamma/metabolism , Peroxidase/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Ventilator-Induced Lung Injury/immunology , Ventilator-Induced Lung Injury/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...