Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters

Publication year range
1.
Mol Med ; 30(1): 68, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778274

ABSTRACT

BACKGROUND: Acute respiratory distress syndrome (ARDS) is characterized by alveolar edema that can progress to septal fibrosis. Mechanical ventilation can augment lung injury, termed ventilator-induced lung injury (VILI). Connective tissue growth factor (CTGF), a mediator of fibrosis, is increased in ARDS patients. Blocking CTGF inhibits fibrosis and possibly vascular leakage. This study investigated whether neutralizing CTGF reduces pulmonary edema in VILI. METHODS: Following LPS administration, rats were mechanically ventilated for 6 h with low (6 mL/kg; low VT) or moderate (10 mL/kg; mod VT) tidal volume and treated with a neutralizing CTGF antibody (FG-3154) or placebo lgG (vehicle). Control rats without LPS were ventilated for 6 h with low VT. Lung wet-to-dry weight ratio, FITC-labeled dextran permeability, histopathology, and soluble RAGE were determined. RESULTS: VILI was characterized by reduced PaO2/FiO2 ratio (low VT: 540 [381-661] vs. control: 693 [620-754], p < 0.05), increased wet-to-dry weight ratio (low VT: 4.8 [4.6-4.9] vs. control: 4.5 [4.4-4.6], p < 0.05), pneumonia (low VT: 30 [0-58] vs. control: 0 [0-0]%, p < 0.05) and interstitial inflammation (low VT: 2 [1-3] vs. control: 1 [0-1], p < 0.05). FG-3154 did not affect wet-to-dry weight ratio (mod VT + FG-3154: 4.8 [4.7-5.0] vs. mod VT + vehicle: 4.8 [4.8-5.0], p > 0.99), extravasated dextrans (mod VT + FG-3154: 0.06 [0.04-0.09] vs. mod VT + vehicle: 0.04 [0.03-0.09] µg/mg tissue, p > 0.99), sRAGE (mod VT + FG-3154: 1865 [1628-2252] vs. mod VT + vehicle: 1885 [1695-2159] pg/mL, p > 0.99) or histopathology. CONCLUSIONS: 'Double hit' VILI was characterized by inflammation, impaired oxygenation, pulmonary edema and histopathological lung injury. Blocking CTGF does not improve oxygenation nor reduce pulmonary edema in rats with VILI.


Subject(s)
Connective Tissue Growth Factor , Pulmonary Edema , Ventilator-Induced Lung Injury , Animals , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/pathology , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/antagonists & inhibitors , Rats , Male , Pulmonary Edema/etiology , Pulmonary Edema/metabolism , Antibodies, Neutralizing/pharmacology , Rats, Sprague-Dawley , Lung/pathology , Lung/metabolism , Disease Models, Animal , Receptor for Advanced Glycation End Products/metabolism , Receptor for Advanced Glycation End Products/antagonists & inhibitors
2.
Mol Med ; 30(1): 148, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266965

ABSTRACT

BACKGROUND: Ventilator-induced lung injury (VILI) is one of the severe complications in the clinic concerning mechanical ventilation (MV). Capsaicin (CAP) has anti-inflammatory and inhibitory effects on oxidative stress, which is a significant element causing cellular ferroptosis. Nevertheless, the specific role and potential mechanistic pathways through which CAP modulates ferroptosis in VILI remain elusive. METHODS: VILI was established in vivo, and the pulmonary epithelial cell injury model induced by circulation stretching (CS) was established in vitro. Both mice and cells were pretreated with CAP. Transmission electron microscopy, ELISA, Western blot, immunofluorescence, RT-PCR, fluorescent probes, and other experimental methods were used to clarify the relationship between iron death and VILI in alveolar epithelial cells, and whether capsaicin alleviates VILI by inhibiting iron death and its specific mechanism. RESULTS: Ferroptosis was involved in VILI by utilizing in vivo models. CAP inhibited ferroptosis and alleviated VILI's lung damage and inflammation, and this protective effect of CAP was dependent on maintaining mitochondrial redox system through SITR3 signaling. In the CS-caused lung epithelial cell injury models, CAP reduced pathological CS-caused ferroptosis and cell injury. Knockdown SIRT3 reversed the role of CAP on the maintaining mitochondria dysfunction under pathological CS and eliminated its subsequent advantageous impacts for ferroptosis against overstretching cells. CONCLUSION: The outcomes showed that CAP alleviated ferroptosis in VILI via improving the activity of SITR3 to suppressing mitochondrial oxidative damage and maintaining mitochondrial redox homeostasis, illustrating its possibility as a novel therapeutic goal for VILI.


Subject(s)
Capsaicin , Ferroptosis , Homeostasis , Mitochondria , Oxidation-Reduction , Sirtuin 3 , Ventilator-Induced Lung Injury , Ferroptosis/drug effects , Animals , Mitochondria/metabolism , Mitochondria/drug effects , Mice , Sirtuin 3/metabolism , Sirtuin 3/genetics , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/drug therapy , Oxidation-Reduction/drug effects , Capsaicin/pharmacology , Male , Disease Models, Animal , Humans , Mice, Inbred C57BL , Oxidative Stress/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/drug effects , Signal Transduction/drug effects
3.
Microvasc Res ; 153: 104658, 2024 05.
Article in English | MEDLINE | ID: mdl-38266910

ABSTRACT

Endothelial permeability deterioration is involved in ventilator-induced lung injury (VILI). The integrality of vascular endothelial glycocalyx (EG) is closely associated with endothelial permeability. The hypothesis was that vascular EG shedding participates in VILI through promoting endothelial permeability. In the present study, male Sprague-Dawley (SD) rats were ventilated with high tidal volume (VT =40 ml/kg) or low tidal volume (VT =8 ml/kg) to investigate the effects of different tidal volume and ventilation durations on EG in vivo. We report disruption of EG during the period of high tidal volume ventilation characterized by increased glycocalyx structural components (such as syndecan-1, heparan sulfate, hyaluronan) in the plasma and decreased the expression of syndecan-1 in the lung tissues. Mechanistically, the disruption of EG was associated with increased proinflammatory cytokines and matrix metalloproteinase in the lung tissues. Collectively, these results demonstrate that the degradation of EG is involved in the occurrence and development of VILI in rats, and the inflammatory mechanism mediated by activation of the NF-κB signaling pathway may be partly responsible for the degradation of EG in VILI in rats. This study enhances our understanding of the pathophysiological processes underlying VILI, shedding light on potential therapeutic targets to mitigate VILI.


Subject(s)
Syndecan-1 , Ventilator-Induced Lung Injury , Rats , Male , Animals , Rats, Sprague-Dawley , Glycocalyx/metabolism , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/metabolism , Lung/metabolism
4.
Respir Res ; 25(1): 330, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227918

ABSTRACT

BACKGROUND: Vascular endothelial damage is involved in the development and exacerbation of ventilator-induced lung injury (VILI). Pulmonary endothelial glycocalyx and neutrophil extracellular traps (NETs) are endothelial protective and damaging factors, respectively; however, their dynamics in VILI and the effects of recombinant thrombomodulin and antithrombin on these dynamics remain unclear. We hypothesized that glycocalyx degradation and NETs are induced by VILI and suppressed by recombinant thrombomodulin, recombinant antithrombin, or their combination. METHODS: VILI was induced in male C57BL/6J mice by intraperitoneal lipopolysaccharide injection (20 mg/kg) and high tidal volume ventilation (20 mL/kg). In the intervention groups, recombinant thrombomodulin, recombinant antithrombin, or their combination was administered at the start of mechanical ventilation. Glycocalyx degradation was quantified by measuring serum syndecan-1, fluorescence-labeled lectin intensity, and glycocalyx-occupied area in the pulmonary vascular lumen. Double-stranded DNA in the bronchoalveolar fluid and fluorescent areas of citrullinated histone H3 and myeloperoxidase were quantified as NET formation. RESULTS: Serum syndecan-1 increased, and lectin fluorescence intensity decreased in VILI. Electron microscopy revealed decreases in glycocalyx-occupied areas within pulmonary microvessels in VILI. Double-stranded DNA levels in the bronchoalveolar lavage fluid and the fluorescent area of citrullinated histone H3 and myeloperoxidase in lung tissues increased in VILI. Recombinant thrombomodulin, recombinant antithrombin, and their combination reduced glycocalyx injury and NET marker levels. There was little difference in glycocalyx injury and NET makers between the intervention groups. CONCLUSION: VILI induced glycocalyx degradation and NET formation. Recombinant thrombomodulin and recombinant antithrombin attenuated glycocalyx degradation and NETs in our VILI model. The effect of their combination did not differ from that of either drug alone. Recombinant thrombomodulin and antithrombin have the potential to be therapeutic agents for biotrauma in VILI.


Subject(s)
Antithrombins , Endotoxemia , Extracellular Traps , Glycocalyx , Mice, Inbred C57BL , Recombinant Proteins , Thrombomodulin , Ventilator-Induced Lung Injury , Animals , Glycocalyx/metabolism , Glycocalyx/drug effects , Glycocalyx/pathology , Thrombomodulin/metabolism , Thrombomodulin/administration & dosage , Extracellular Traps/metabolism , Extracellular Traps/drug effects , Male , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Mice , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/pathology , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/prevention & control , Endotoxemia/metabolism , Endotoxemia/pathology , Endotoxemia/drug therapy , Endotoxemia/chemically induced , Antithrombins/pharmacology , Lung/metabolism , Lung/drug effects , Lung/pathology , Disease Models, Animal , Syndecan-1/metabolism
5.
Arch Biochem Biophys ; 752: 109853, 2024 02.
Article in English | MEDLINE | ID: mdl-38086523

ABSTRACT

Ventilator-induced lung injury (VILI) disturbs the disordered immune system and causes persistent inflammatory damage. 4-octyl itaconate (OI) is a synthetic cell-permeable itaconate derivative with antioxidant and anti-inflammatory effects. In this study, we assessed whether OI protects against VILI. OI was intraperitoneally injected for three days before mechanical ventilation (MV; 20 ml/kg at 70 breaths/min) for 2 h. Mouse lung vascular endothelial cells (MLVECs) were pretreated with OI (62.5, 125, and 250 µM) prior to cyclic stretch for 4 h. We found that OI attenuated VILI and inflammatory response. OI also increased superoxide dismutase, nuclear factor E2-related factor 2, and heme oxygenase-1 levels, and decreased reactive oxygen species and malondialdehyde levels. Furthermore, OI inhibited the expression of NLR family pyrin domain-containing 3 (NLRP3), caspase-1 p20, apoptosis-associated speck-like protein containing a CARD, and N-terminal fragment of gasdermin D. Therefore, OI attenuates VILI, potentially by suppressing oxidative stress and NLRP3 activation.


Subject(s)
NLR Family, Pyrin Domain-Containing 3 Protein , Succinates , Ventilator-Induced Lung Injury , Mice , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Endothelial Cells/metabolism , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/metabolism , NF-E2-Related Factor 2/metabolism , Lung/metabolism
6.
Phytother Res ; 38(7): 3401-3416, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38666397

ABSTRACT

Our previous research confirmed that rutin reduced ventilator-induced lung injury (VILI) in mice. Ferroptosis has been reported to participate in the pathogenic process of VILI. We will explore whether rutin inhibits ferroptosis to alleviate VILI. A mouse model of VILI was constructed with or without rutin pretreatment to perform a multiomics analysis. Hematoxylin-eosin (HE) staining and transmission electron microscopy were used to evaluate lung injury in VILI mice. Dihydroethidium (DHE) staining and the malondialdehyde (MDA) and superoxide dismutase (SOD) levels were detected. Molecular docking was performed to determine the binding affinity between rutin and ferroptosis-related proteins. Western blot analysis, real-time PCR (RT-PCR) and immunohistochemical (IHC) staining were conducted to detect the expression levels of GPX4, XCT, ACSL4, FTH1, AKT and p-AKT in lung tissues. Microscale thermophoresis (MST) was used to evaluate the binding between rutin and AKT1. Transcriptomic and proteomic analyses showed that ferroptosis may play a key role in VILI mice. Metabolomic analysis demonstrated that rutin may affect ferroptosis via the AKT pathway. Molecular docking analysis indicated that rutin may regulate the expression of ferroptosis-related proteins. Moreover, rutin upregulated GPX4 expression and downregulated the expression of XCT, ACSL4 and FTH1 in the lung tissues. Rutin also increased the ratio of p-AKT/AKT and p-AKT expression. MST analysis showed that rutin binds to AKT1. Rutin binds to AKT to activate the AKT signaling pathway, contributing to inhibit ferroptosis, thus preventing VILI in mice. Our study elucidated a possible novel strategy of involving the use of rutin for preventing VILI.


Subject(s)
Ferroptosis , Molecular Docking Simulation , Proto-Oncogene Proteins c-akt , Rutin , Ventilator-Induced Lung Injury , Animals , Ferroptosis/drug effects , Rutin/pharmacology , Mice , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/prevention & control , Proto-Oncogene Proteins c-akt/metabolism , Male , Disease Models, Animal , Mice, Inbred C57BL , Malondialdehyde/metabolism , Lung/drug effects , Lung/pathology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Superoxide Dismutase/metabolism , Signal Transduction/drug effects , Amino Acid Transport System y+/metabolism
7.
Inflamm Res ; 72(5): 971-988, 2023 May.
Article in English | MEDLINE | ID: mdl-37004548

ABSTRACT

BACKGROUND: The ability of p38 to phosphorylate substrates in the nucleus and the role of nuclear p38 in the regulation of inflammation have focused attention on the subcellular localization of the kinase. Although it is clear that p38 shuttles to the nucleus upon stimulation, the mechanisms that regulate p38 nuclear input in response to mechanical stretch remain to be determined. METHODS: Cyclic stretch (CS)-induced nuclear translocation of p38 was determined by Western blotting and immunofluorescence. The p38 interacting protein was identified using endogenous pull-down and protein binding assays. The potential role of importin-7 (Imp7) in CS-induced nuclear translocation of p38 and p38-dependent gene expression was confirmed using a series of in vitro and in vivo experiments. Furthermore, we tested the therapeutic potential of intratracheal administration of Imp7 siRNA-loaded nanoparticles in the ventilator-induced lung injury (VILI) mouse model. RESULTS: We show that CS induced phosphorylation-dependent nuclear translocation of p38, which required the involvement of microtubules and dynein. Endogenous pull-down assay revealed Imp7 to be a potential p38-interacting protein, and the direct interaction between p38 and Imp7 was confirmed by in vitro and in vivo binding assays. Furthermore, silencing Imp7 inhibited CS-induced nuclear translocation of p38 and subsequent cytokine production. Notably, intratracheal administration of Imp7 siRNA nanoparticles attenuated lung inflammation and histological damage in the VILI mouse model. CONCLUSIONS: Our findings uncover a key role for Imp7 in the process of p38 nuclear import after CS stimulation and highlight the potential of preventing p38 nuclear translocation in treatment of VILI.


Subject(s)
Cell Nucleus , Ventilator-Induced Lung Injury , Mice , Animals , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , RNA, Small Interfering/metabolism , Karyopherins/metabolism , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/metabolism
8.
Crit Care ; 27(1): 196, 2023 05 22.
Article in English | MEDLINE | ID: mdl-37218012

ABSTRACT

BACKGROUND: Ventilator-induced lung injury (VILI) is caused by overdistension of the alveoli by the repetitive recruitment and derecruitment of alveolar units. This study aims to investigate the potential role and mechanism of fibroblast growth factor 21 (FGF21), a metabolic regulator secreted by the liver, in VILI development. METHODS: Serum FGF21 concentrations were determined in patients undergoing mechanical ventilation during general anesthesia and in a mouse VILI model. Lung injury was compared between FGF21-knockout (KO) mice and wild-type (WT) mice. Recombinant FGF21 was administrated in vivo and in vitro to determine its therapeutic effect. RESULTS: Serum FGF21 levels in patients and mice with VILI were significantly higher than in those without VILI. Additionally, the increment of serum FGF21 in anesthesia patients was positively correlated with the duration of ventilation. VILI was aggravated in FGF21-KO mice compared with WT mice. Conversely, the administration of FGF21 alleviated VILI in both mouse and cell models. FGF21 reduced Caspase-1 activity, suppressed the mRNA levels of Nlrp3, Asc, Il-1ß, Il-18, Hmgb1 and Nf-κb, and decreased the protein levels of NLRP3, ASC, IL-1ß, IL-18, HMGB1 and the cleaved form of GSDMD. CONCLUSIONS: Our findings reveal that endogenous FGF21 signaling is triggered in response to VILI, which protects against VILI by inhibiting the NLRP3/Caspase-1/GSDMD pyroptosis pathway. These results suggest that boosting endogenous FGF21 or the administration of recombinant FGF21 could be promising therapeutic strategies for the treatment of VILI during anesthesia or critical care.


Subject(s)
HMGB1 Protein , Ventilator-Induced Lung Injury , Animals , Mice , Caspase 1/metabolism , Disease Models, Animal , Inflammasomes , Interleukin-18 , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/prevention & control , Humans
9.
Nanomedicine ; 50: 102679, 2023 06.
Article in English | MEDLINE | ID: mdl-37116556

ABSTRACT

Acute respiratory distress syndrome (ARDS) has high mortality (~40 %) and requires the lifesaving intervention of mechanical ventilation. A variety of systemic inflammatory insults can progress to ARDS, and the inflamed and injured lung is susceptible to ventilator-induced lung injury (VILI). Strategies to mitigate the inflammatory response while restoring pulmonary function are limited, thus we sought to determine if treatment with CNP-miR146a, a conjugate of novel free radical scavenging cerium oxide nanoparticles (CNP) to the anti-inflammatory microRNA (miR)-146a, would protect murine lungs from acute lung injury (ALI) induced with intratracheal endotoxin and subsequent VILI. Lung injury severity and treatment efficacy were evaluated via lung mechanical function, relative gene expression of inflammatory biomarkers, and lung morphometry (stereology). CNP-miR146a reduced the severity of ALI and slowed the progression of VILI, evidenced by improvements in inflammatory biomarkers, atelectasis, gas volumes in the parenchymal airspaces, and the stiffness of the pulmonary system.


Subject(s)
Acute Lung Injury , Respiratory Distress Syndrome , Ventilator-Induced Lung Injury , Humans , Mice , Animals , Lung/metabolism , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/genetics , Ventilator-Induced Lung Injury/metabolism , Respiratory Distress Syndrome/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/genetics
10.
Cytokine ; 157: 155950, 2022 09.
Article in English | MEDLINE | ID: mdl-35780712

ABSTRACT

BACKGROUND: Ventilator-induced lung injury (VILI) is a complex pathophysiological process leading to acute respiratory distress syndrome (ARDS) and poor outcomes in affected patients. As a form of programmed cell death, pyroptosis is proposed to play an important role in the development of ARDS. Here we investigated whether treating mice with the specific RIPK1 inhibitor Necrostatin-1 (Nec-1) before mechanical ventilation could inhibit pyroptosis and alleviate lung injury in a mouse model. METHODOLOGYS: Anesthetized C57BL/6J mice received a transtracheal injection of Nec-1 (5 mg/kg) or vehicle (DMSO) 30 min before the experiment which was ventilated for up to 4 h. Lung damage was assessed macroscopically and histologically with oedema measured as the wet/dry ratio of lung tissues. The release of inflammatory mediators into bronchoalveolar lavage fluid (BALF) was assessed by ELISA measurements of TNF-α,interleukin-1ß (IL-1ß), and IL-6. The expression of RIPK1, ZBP1, caspase-1, and activated (cleaved) caspase-1 were analyzed using western blot and immunohistochemistry, and the levels of gasdermin-D (GSDMD) and IL-1ß were analyzed by immunofluorescence staining. RESULTS: High tidal ventilation produced time-dependent inflammation and lung injury in mice which could be significantly reduced by pretreatment with Nec-1. Notably, Nec-1 reduced the expression of key pyroptosis mediator proteins in lung tissues exposed to mechanical ventilation, including caspase-1, cleaved caspase-1, and GSDMD together with inhibiting the release of inflammatory cytokines. CONCLUSION: Nec-1 pretreatment alleviates pulmonary inflammatory responses and protects the lung from mechanical ventilation damage. The beneficial effects were mediated at least in part by inhibiting caspase-1-dependent pyroptosis through the RIPK1/ZBP1 pathway.


Subject(s)
Respiratory Distress Syndrome , Ventilator-Induced Lung Injury , Animals , Caspase 1 , Imidazoles , Indoles , Lung/pathology , Mice , Mice, Inbred C57BL , Pyroptosis , RNA-Binding Proteins , Receptor-Interacting Protein Serine-Threonine Kinases , Ventilator-Induced Lung Injury/drug therapy
11.
BMC Pulm Med ; 22(1): 9, 2022 Jan 06.
Article in English | MEDLINE | ID: mdl-34986829

ABSTRACT

BACKGROUND: As one of the basic treatments performed in the intensive care unit, mechanical ventilation can cause ventilator-induced acute lung injury (VILI). The typical features of VILI are an uncontrolled inflammatory response and impaired lung barrier function; however, its pathogenesis is not fully understood, and c-Fos protein is activated under mechanical stress. c-Fos/activating protein-1 (AP-1) plays a role by binding to AP-1 within the promoter region, which promotes inflammation and apoptosis. T-5224 is a specific inhibitor of c-Fos/AP-1, that controls the gene expression of many proinflammatory cytokines. This study investigated whether T-5224 attenuates VILI in rats by inhibiting inflammation and apoptosis. METHODS: The SD rats were divided into six groups: a control group, low tidal volume group, high tidal volume group, DMSO group, T-5224 group (low concentration), and T-5224 group (high concentration). After 3 h, the pathological damage, c-Fos protein expression, inflammatory reaction and apoptosis degree of lung tissue in each group were detected. RESULTS: c-Fos protein expression was increased within the lung tissue of VILI rats, and the pathological damage degree, inflammatory reaction and apoptosis in the lung tissue of VILI rats were significantly increased; T-5224 inhibited c-Fos protein expression in lung tissues, and T-5224 inhibit the inflammatory reaction and apoptosis of lung tissue by regulating the Fas/Fasl pathway. CONCLUSIONS: c-Fos is a regulatory factor during ventilator-induced acute lung injury, and the inhibition of its expression has a protective effect. Which is associated with the antiinflammatory and antiapoptotic effects of T-5224.


Subject(s)
Benzophenones/pharmacology , Isoxazoles/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/pharmacology , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/physiopathology , Animals , Apoptosis/drug effects , Inflammation/pathology , Male , Rats , Rats, Sprague-Dawley
12.
Am J Respir Cell Mol Biol ; 65(4): 403-412, 2021 10.
Article in English | MEDLINE | ID: mdl-34014798

ABSTRACT

Mechanical ventilation is a known risk factor for delirium, a cognitive impairment characterized by dysfunction of the frontal cortex and hippocampus. Although IL-6 is upregulated in mechanical ventilation-induced lung injury (VILI) and may contribute to delirium, it is not known whether the inhibition of systemic IL-6 mitigates delirium-relevant neuropathology. To histologically define neuropathological effects of IL-6 inhibition in an experimental VILI model, VILI was simulated in anesthetized adult mice using a 35 cc/kg tidal volume mechanical ventilation model. There were two control groups, as follow: 1) spontaneously breathing or 2) anesthetized and mechanically ventilated with 10 cc/kg tidal volume to distinguish effects of anesthesia from VILI. Two hours before inducing VILI, mice were treated with either anti-IL-6 antibody, anti-IL-6 receptor antibody, or saline. Neuronal injury, stress, and inflammation were assessed using immunohistochemistry. CC3 (cleaved caspase-3), a neuronal apoptosis marker, was significantly increased in the frontal (P < 0.001) and hippocampal (P < 0.0001) brain regions and accompanied by significant increases in c-Fos and heat shock protein-90 in the frontal cortices of VILI mice compared with control mice (P < 0.001). These findings were not related to cerebral hypoxia, and there was no evidence of irreversible neuronal death. Frontal and hippocampal neuronal CC3 were significantly reduced with anti-IL-6 antibody (P < 0.01 and P < 0.0001, respectively) and anti-IL-6 receptor antibody (P < 0.05 and P < 0.0001, respectively) compared with saline VILI mice. In summary, VILI induces potentially reversible neuronal injury and inflammation in the frontal cortex and hippocampus, which is mitigated with systemic IL-6 inhibition. These data suggest a potentially novel neuroprotective role of systemic IL-6 inhibition that justifies further investigation.


Subject(s)
Antibodies/pharmacology , Apoptosis/drug effects , Delirium/metabolism , Interleukin-6/antagonists & inhibitors , Neurons/metabolism , Ventilator-Induced Lung Injury/metabolism , Animals , Delirium/drug therapy , Delirium/pathology , Disease Models, Animal , Female , Frontal Lobe/injuries , Frontal Lobe/metabolism , Frontal Lobe/pathology , HSP90 Heat-Shock Proteins/metabolism , Hippocampus/injuries , Hippocampus/metabolism , Hippocampus/pathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/metabolism , Mice , Neurons/pathology , Proto-Oncogene Proteins c-fos/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/pathology
13.
Am J Physiol Lung Cell Mol Physiol ; 320(1): L17-L28, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33026237

ABSTRACT

Oxidative stress caused by mechanical ventilation contributes to the pathophysiology of ventilator-induced lung injury (VILI). A key mechanism maintaining redox balance is the upregulation of nuclear factor-erythroid-2-related factor 2 (Nrf2)-dependent antioxidant gene expression. We tested whether pretreatment with an Nrf2-antioxidant response element (ARE) pathway activator tert-butylhydroquinone (tBHQ) protects against VILI. Male C57BL/6J mice were pretreated with an intraperitoneal injection of tBHQ (n = 10), an equivalent volume of 3% ethanol (EtOH3%, vehicle, n = 13), or phosphate-buffered saline (controls, n = 10) and were then subjected to high tidal volume (HVT) ventilation for a maximum of 4 h. HVT ventilation severely impaired arterial oxygenation ([Formula: see text] = 49 ± 7 mmHg, means ± SD) and respiratory system compliance, resulting in a 100% mortality among controls. Compared with controls, tBHQ improved arterial oxygenation ([Formula: see text] = 90 ± 41 mmHg) and respiratory system compliance after HVT ventilation. In addition, tBHQ attenuated the HVT ventilation-induced development of lung edema and proinflammatory response, evidenced by lower concentrations of protein and proinflammatory cytokines (IL-1ß and TNF-α) in the bronchoalveolar lavage fluid, respectively. Moreover, tBHQ enhanced the pulmonary redox capacity, indicated by enhanced Nrf2-depentent gene expression at baseline and by the highest total glutathione concentration after HVT ventilation among all groups. Overall, tBHQ pretreatment resulted in 60% survival (P < 0.001 vs. controls). Interestingly, compared with controls, EtOH3% reduced the proinflammatory response to HVT ventilation in the lung, resulting in 38.5% survival (P = 0.0054 vs. controls). In this murine model of VILI, tBHQ increases the pulmonary redox capacity by activating the Nrf2-ARE pathway and protects against VILI. These findings support the efficacy of pharmacological Nrf2-ARE pathway activation to increase resilience against oxidative stress during injurious mechanical ventilation.


Subject(s)
Gene Expression Regulation , Hydroquinones/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Protective Agents/pharmacology , Pulmonary Edema/prevention & control , Ventilator-Induced Lung Injury/mortality , Animals , Antioxidant Response Elements , Antioxidants/pharmacology , Bronchoalveolar Lavage Fluid , Cytokines/metabolism , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , Pulmonary Edema/etiology , Respiration, Artificial/adverse effects , Survival Rate , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/etiology , Ventilator-Induced Lung Injury/pathology
14.
Int J Mol Sci ; 22(4)2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33567713

ABSTRACT

Mechanical ventilation (MV) is required to maintain life for patients with sepsis-related acute lung injury but can cause diaphragmatic myotrauma with muscle damage and weakness, known as ventilator-induced diaphragm dysfunction (VIDD). Hypoxia-inducible factor 1α (HIF-1α) plays a crucial role in inducing inflammation and apoptosis. Low-molecular-weight heparin (LMWH) was proven to have anti-inflammatory properties. However, HIF-1α and LMWH affect sepsis-related diaphragm injury has not been investigated. We hypothesized that LMWH would reduce endotoxin-augmented VIDD through HIF-1α. C57BL/6 mice, either wild-type or HIF-1α-deficient, were exposed to MV with or without endotoxemia for 8 h. Enoxaparin (4 mg/kg) was administered subcutaneously 30 min before MV. MV with endotoxemia aggravated VIDD, as demonstrated by increased interleukin-6 and macrophage inflammatory protein-2 levels, oxidative loads, and the expression of HIF-1α, calpain, caspase-3, atrogin-1, muscle ring finger-1, and microtubule-associated protein light chain 3-II. Disorganized myofibrils, disrupted mitochondria, increased numbers of autophagic and apoptotic mediators, substantial apoptosis of diaphragm muscle fibers, and decreased diaphragm function were also observed (p < 0.05). Endotoxin-exacerbated VIDD and myonuclear apoptosis were attenuated by pharmacologic inhibition by LMWH and in HIF-1α-deficient mice (p < 0.05). Our data indicate that enoxaparin reduces endotoxin-augmented MV-induced diaphragmatic injury, partially through HIF-1α pathway inhibition.


Subject(s)
Diaphragm/drug effects , Disease Models, Animal , Endotoxemia/complications , Heparin, Low-Molecular-Weight/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Oxidative Stress/drug effects , Ventilator-Induced Lung Injury/drug therapy , Animals , Endotoxemia/chemically induced , Endotoxemia/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Ventilator-Induced Lung Injury/etiology , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/pathology
15.
Mol Med ; 26(1): 98, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33126860

ABSTRACT

BACKGROUND: Mechanical ventilation, in combination with supraphysiological concentrations of oxygen (i.e., hyperoxia), is routinely used to treat patients with respiratory distress, such as COVID-19. However, prolonged exposure to hyperoxia compromises the clearance of invading pathogens by impairing macrophage phagocytosis. Previously, we have shown that the exposure of mice to hyperoxia induces the release of the nuclear protein high mobility group box-1 (HMGB1) into the pulmonary airways. Furthermore, extracellular HMGB1 impairs macrophage phagocytosis and increases the mortality of mice infected with Pseudomonas aeruginosa (PA). The aim of this study was to determine whether GTS-21 (3-(2,4-dimethoxybenzylidene) anabaseine), an α7 nicotinic acetylcholine receptor (α7nAChR) agonist, could (1) inhibit hyperoxia-induced HMGB1 release into the airways; (2) enhance macrophage phagocytosis and (3) increase bacterial clearance from the lungs in a mouse model of ventilator-associated pneumonia. METHOD: GTS-21 (0.04, 0.4, and 4 mg/kg) or saline were administered by intraperitoneal injection to mice that were exposed to hyperoxia (≥ 99% O2) and subsequently challenged with PA. RESULTS: The systemic administration of 4 mg/kg i.p. of GTS-21 significantly increased bacterial clearance, decreased acute lung injury and decreased accumulation of airway HMGB1 compared to the saline control. To determine the mechanism of action of GTS-21, RAW 264.7 cells, a macrophage-like cell line, were incubated with different concentrations of GTS-21 in the presence of 95% O2. The phagocytic activity of macrophages was significantly increased by GTS-21 in a dose-dependent manner. In addition, GTS-21 significantly inhibited the cytoplasmic translocation and release of HMGB1 from RAW 264.7 cells and attenuated hyperoxia-induced NF-κB activation in macrophages and mouse lungs exposed to hyperoxia and infected with PA. CONCLUSIONS: Our results indicate that GTS-21 is efficacious in improving bacterial clearance and reducing acute lung injury via enhancing macrophage function by inhibiting the release of nuclear HMGB1. Therefore, the α7nAChR represents a possible pharmacological target to improve the clinical outcome of patients on ventilators by augmenting host defense against bacterial infections.


Subject(s)
Benzylidene Compounds/pharmacology , Hyperoxia/immunology , Macrophages, Alveolar/drug effects , Pseudomonas Infections/drug therapy , Pyridines/pharmacology , Ventilator-Induced Lung Injury/drug therapy , alpha7 Nicotinic Acetylcholine Receptor/antagonists & inhibitors , Animals , Disease Models, Animal , HMGB1 Protein/metabolism , Hyperoxia/diet therapy , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL , Phagocytosis/drug effects , Pseudomonas aeruginosa , RAW 264.7 Cells
16.
Exp Mol Pathol ; 114: 104427, 2020 06.
Article in English | MEDLINE | ID: mdl-32199914

ABSTRACT

Ventilator-induced lung injury (VILI) causes problems during acute lung injury treatment, and propofol is a well-known drug to prevent VILI. Herein, we discussed how propofol protects against VILI-induced inflammation with the interaction of nuclear factor E2-related factor 2 (Nrf2)/NOD-like receptor protein 3 (NLRP3). We established VILI mouse models for collecting lung tissues, and these mice were later treated with propofol and Nrf2/NLRP3 activator or inhibitor to observe their effects on VILI with inflammatory factors, 8-hydroxy-2 deoxyguanosine, malondialchehyche level, mitochondrial reactive oxygen species production rate, lung wet/dry weight ratio, lung permeability index measured. Propofol treatment improved VILI, alleviated pulmonary inflammation induced by mechanical ventilation. Propofol up-regulated Nrf2 and down-regulated NLRP3 in VILI model. Activating Nrf2 or inhibiting NLRP3 downregulated pro-inflammatory factors in lung tissues in VILI mice. Above all, we can conclude that propofol exerts it protective function against VILI and the subsequent inflammatory responses through activating Nrf2 and inhibiting NLRP3 expression. Therefore, Nrf2 activator and NLRP3 inhibitor might be latent targets in the VILI prevention.


Subject(s)
NF-E2-Related Factor 2/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Propofol/pharmacology , Ventilator-Induced Lung Injury/drug therapy , Animals , Disease Models, Animal , Humans , Inflammation/drug therapy , Inflammation/genetics , Inflammation/pathology , Lung/drug effects , Lung/pathology , Mice , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Ventilator-Induced Lung Injury/genetics , Ventilator-Induced Lung Injury/pathology
17.
Mediators Inflamm ; 2020: 6705985, 2020.
Article in English | MEDLINE | ID: mdl-33299377

ABSTRACT

Ventilator-induced lung injury (VILI) is a severe and inevitable complication in patients who require mechanical ventilation (MV) for respiratory support. Lipoxin A4 is an endogenous anti-inflammatory and antioxidant mediator. The present study determined the effects of lipoxin A4 on VILI. Twenty-four rats were randomized to the sham, VILI, and lipoxin A4 (LX4) groups. The rats in the VILI and LX4 groups received large-volume MV for 4 hours to simulate VILI. Capillary permeability was evaluated using the PaO2/FiO2 ratio, lung wet/dry weight ratio, and protein level in the lung. VILI-induced inflammation was assessed by measuring cytokines in serum and lung tissue, the expression and activity of NF-κB, and phosphorylated myosin light chain. The oxidative stress response, lung tissue injury, and apoptosis in lung tissue were also estimated, and the expression of apoptotic proteins was examined. MV worsened all of the indices compared to the sham group. Compared to the VILI group, the LX4 group showed significantly improved alveolar-capillary permeability (increased PaO2/FiO2 and decreased wet/dry weight ratios and protein levels), ameliorated histological injury, and reduced local and systemic inflammation (downregulated proinflammatory factors and NF-κB expression and activity). Lipoxin A4 notably inhibited the oxidative stress response and apoptosis and balanced apoptotic protein levels in lung tissue. Lipoxin A4 protects against VILI via anti-inflammatory, antioxidant, and antiapoptotic effects.


Subject(s)
Lipoxins/pharmacology , Ventilator-Induced Lung Injury/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis , Bronchoalveolar Lavage Fluid , Capillaries , Capillary Permeability/drug effects , Cytokines/metabolism , Inflammation , Lung/pathology , Oxidative Stress , Permeability , Phosphorylation , Pulmonary Alveoli/drug effects , Rats , Rats, Sprague-Dawley , Respiration, Artificial , Ventilator-Induced Lung Injury/metabolism
18.
Int J Mol Sci ; 21(9)2020 Apr 28.
Article in English | MEDLINE | ID: mdl-32353952

ABSTRACT

Patients with sepsis frequently require mechanical ventilation (MV) to survive. However, MV has been shown to induce the production of proinflammatory cytokines, causing ventilator-induced lung injury (VILI). It has been demonstrated that hypoxia-inducible factor (HIF)-1α plays a crucial role in inducing both apoptotic and inflammatory processes. Low-molecular-weight heparin (LMWH) has been shown to have anti-inflammatory activities. However, the effects of HIF-1α and LMWH on sepsis-related acute lung injury (ALI) have not been fully delineated. We hypothesized that LMWH would reduce lung injury, production of free radicals and epithelial apoptosis through the HIF-1α pathway. Male C57BL/6 mice were exposed to 6-mL/kg or 30-mL/kg MV for 5 h. Enoxaparin, 4 mg/kg, was administered subcutaneously 30 min before MV. We observed that MV with endotoxemia induced microvascular permeability; interleukin-6, tumor necrosis factor-α, macrophage inflammatory protein-2 and vascular endothelial growth factor protein production; neutrophil infiltration; oxidative loads; HIF-1α mRNA activation; HIF-1α expression; bronchial epithelial apoptosis; and decreased respiratory function in mice (p < 0.05). Endotoxin-induced augmentation of VILI and epithelial apoptosis were reduced in the HIF-1α-deficient mice and in the wild-type mice following enoxaparin administration (p < 0.05). Our data suggest that enoxaparin reduces endotoxin-augmented MV-induced ALI, partially by inhibiting the HIF-1α pathway.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Endotoxemia/rehabilitation , Enoxaparin/administration & dosage , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lipopolysaccharides/adverse effects , Salmonella typhi/metabolism , Ventilator-Induced Lung Injury/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Chemokine CXCL2/metabolism , Disease Models, Animal , Endotoxemia/chemically induced , Endotoxemia/genetics , Endotoxemia/metabolism , Enoxaparin/pharmacology , Gene Expression Regulation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Injections, Subcutaneous , Interleukin-6/metabolism , Male , Mice , Oxidative Stress/drug effects , Respiration, Artificial/adverse effects , Salmonella typhi/pathogenicity , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/metabolism , Ventilator-Induced Lung Injury/etiology , Ventilator-Induced Lung Injury/genetics , Ventilator-Induced Lung Injury/metabolism
19.
Respir Res ; 20(1): 274, 2019 Dec 04.
Article in English | MEDLINE | ID: mdl-31801520

ABSTRACT

By airway surface liquid, we mean a thin fluid continuum consisting of the airway lining layer and the alveolar lining layer, which not only serves as a protective barrier against foreign particles but also contributes to maintaining normal respiratory mechanics. In recent years, measurements of the rheological properties of airway surface liquid have attracted considerable clinical attention due to new advances in microrheology instruments and methods. This article reviews the clinical relevance of measurements of airway surface liquid viscoelasticity and surface tension from four main aspects: maintaining the stability of the airways and alveoli, preventing ventilator-induced lung injury, optimizing surfactant replacement therapy for respiratory syndrome distress, and characterizing the barrier properties of airway mucus to improve drug and gene delivery. Primary measuring techniques and methods suitable for determining the viscoelasticity and surface tension of airway surface liquid are then introduced with respect to principles, advantages and limitations. Cone and plate viscometers and particle tracking microrheometers are the most commonly used instruments for measuring the bulk viscosity and microviscosity of airway surface liquid, respectively, and pendant drop methods are particularly suitable for the measurement of airway surface liquid surface tension in vitro. Currently, in vivo and in situ measurements of the viscoelasticity and surface tension of the airway surface liquid in humans still presents many challenges.


Subject(s)
Bronchitis/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Surfactants/administration & dosage , Ventilator-Induced Lung Injury/pathology , Animals , Bronchitis/drug therapy , Chronic Disease , Humans , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Pulmonary Disease, Chronic Obstructive/drug therapy , Respiratory Mechanics , Rheology , Sensitivity and Specificity , Surface Tension/drug effects , Ventilator-Induced Lung Injury/drug therapy , Viscosity
20.
Pulm Pharmacol Ther ; 58: 101833, 2019 10.
Article in English | MEDLINE | ID: mdl-31376462

ABSTRACT

While effective treatments for acute respiratory distress syndrome (ARDS) are lacking, mechanical lung ventilation can sustain adequate gas exchange in critically ill patients with respiratory failure due to ARDS. However, as a result of the phenomenon of ventilator-induced lung injury (VILI), there is an increasing need to seek beneficial pharmacological therapies for ARDS. Recent studies have suggested the renin-angiotensin system (RAS), which consists of the ACE/Ang-II/AT1R axis and ACE2/Ang-(1-7)/MasR axis, plays a dual role in the pathogenesis of ARDS and VILI. This review highlights the deleterious action of ACE/Ang-II/AT1R axis and the beneficial role of ACE2/Ang-(1-7)/MasR axis, as well as AT2R, in VILI and ARDS, and also discusses the possibility of targeting RAS components with pharmacological interventions to improve outcomes in ARDS.


Subject(s)
Renin-Angiotensin System/drug effects , Respiration, Artificial/adverse effects , Respiratory Distress Syndrome/therapy , Ventilator-Induced Lung Injury/drug therapy , Animals , Humans , Proto-Oncogene Mas , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/prevention & control , Ventilator-Induced Lung Injury/pathology , Ventilator-Induced Lung Injury/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL