1
|
Ou D, Xu W, Feng Z, Yang Y, Xue W, Zhang Q, Li X, Zhu Y, Huang J, Fang Y. Vascular endothelial glycocalyx shedding in ventilator-induced lung injury in rats. Microvasc Res 2024; 153:104658. [PMID: 38266910 DOI: 10.1016/j.mvr.2024.104658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
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.
Collapse
Affiliation(s)
- Dingqin Ou
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wenxia Xu
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhaosen Feng
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yihan Yang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wenqiang Xue
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Qinyu Zhang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xuan Li
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yuyang Zhu
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jie Huang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Yu Fang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
2
|
Xu Y, Zhang Y, Zhang J, Liang W, Wang Y, Zeng Z, Liang Z, Ling Z, Chen Y, Deng X, Huang Y, Liu X, Zhang H, Li Y. High driving pressure ventilation induces pulmonary hypertension in a rabbit model of acute lung injury. J Intensive Care 2023; 11:42. [PMID: 37749622 PMCID: PMC10518953 DOI: 10.1186/s40560-023-00689-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/04/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Mechanical ventilation may cause pulmonary hypertension in patients with acute lung injury (ALI), but the underlying mechanism remains elucidated. METHODS ALI was induced in rabbits by a two-hit injury, i.e., hydrochloric acid aspiration followed by mechanical ventilation for 1 h. Rabbits were then ventilated with driving pressure of 10, 15, 20, or 25 cmH2O for 7 h. Clinicopathological parameters were measured at baseline and different timepoints of ventilation. RNA sequencing was conducted to identify the differentially expressed genes in high driving pressure ventilated lung tissue. RESULTS The two-hit injury induced ALI in rabbits was evidenced by dramatically decreased PaO2/FiO2 in the ALI group compared with that in the control group (144.5 ± 23.8 mmHg vs. 391.6 ± 26.6 mmHg, P < 0.001). High driving pressure ventilation (20 and 25 cmH2O) significantly elevated the parameters of acute pulmonary hypertension at different timepoints compared with low driving pressure (10 and 15 cmH2O), along with significant increases in lung wet/dry ratios, total protein contents in bronchoalveolar lavage fluid, and lung injury scores. The high driving pressure groups showed more pronounced histopathological abnormalities in the lung compared with the low driving pressure groups, accompanied by significant increases in the cross-sectional areas of myocytes, right ventricular weight/body weight value, and Fulton's index. Furthermore, the expression of the genes related to ferroptosis induction was generally upregulated in high driving pressure groups compared with those in low driving pressure groups. CONCLUSIONS A rabbit model of ventilation-induced pulmonary hypertension in ALI was successfully established. Our results open a new research direction investigating the exact role of ferroptosis in ventilation-induced pulmonary hypertension in ALI.
Collapse
Affiliation(s)
- Yonghao Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Yu Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Jie Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Weibo Liang
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Ya Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Zitao Zeng
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Zhenting Liang
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Zhaoyi Ling
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Yubiao Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Xiumei Deng
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Yongbo Huang
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Xiaoqing Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China
| | - Haibo Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China.
- The Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, ON, M5B1W8, Canada.
- Department of Anesthesia, University of Toronto, Toronto, ON, Canada.
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, ON, Canada.
| | - Yimin Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Institute of Respiratory and Health, Medical Center for Respiratory Medicine, State Key Laboratory of Respiratory Disease, Guangzhou, 510120, China.
| |
Collapse
|
3
|
Tsay TB, Chang WH, Hsu CM, Chen LW. Mechanical ventilation enhances Acinetobacter baumannii-induced lung injury through JNK pathways. Respir Res 2021; 22:159. [PMID: 34022899 PMCID: PMC8140754 DOI: 10.1186/s12931-021-01739-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patients in intensive care units (ICUs) often received broad-spectrum antibiotic treatment and Acinetobacter baumannii (A.b.) and Pseudomonas aeruginosa (P.a.) were the most common pathogens causing ventilator-associated pneumonia (VAP). This study aimed to examine the effects and mechanism of mechanical ventilation (MV) on A.b.-induced lung injury and the involvement of alveolar macrophages (AMs). METHODS C57BL/6 wild-type (WT) and c-Jun N-terminal kinase knockout (JNK1-/-) mice received MV for 3 h at 2 days after nasal instillation of A.b., P.a. (1 × 106 colony-forming unit, CFU), or normal saline. RESULTS Intranasal instillation of 106 CFU A.b. in C57BL/6 mice induced a significant increase in total cells and protein levels in the bronchoalveolar lavage fluid (BALF) and neutrophil infiltration in the lungs. MV after A.b. instillation increases neutrophil infiltration, interleukin (IL)-6 and vascular cell adhesion molecule (VCAM) mRNA expression in the lungs and total cells, IL-6 levels, and nitrite levels in the BALF. The killing activity of AMs against A.b. was lower than against P.a. The diminished killing activity was parallel with decreased tumor necrosis factor-α production by AMs compared with A.b. Inducible nitric oxide synthase inhibitor, S-methylisothiourea, decreased the total cell number in BALF on mice receiving A.b. instillation and ventilation. Moreover, MV decreased the A.b. and P.a. killing activity of AMs. MV after A.b. instillation induced less total cells in the BALF and nitrite production in the serum of JNK1-/- mice than those of WT mice. CONCLUSION A.b. is potent in inducing neutrophil infiltration in the lungs and total protein in the BALF. MV enhances A.b.-induced lung injury through an increase in the expression of VCAM and IL-6 levels in the BALF and a decrease in the bacteria-killing activity of AMs. A lower inflammation level in JNK1-/- mice indicates that A.b.-induced VAP causes lung injury through JNK signaling pathway in the lungs.
Collapse
MESH Headings
- Acinetobacter Infections/enzymology
- Acinetobacter Infections/microbiology
- Acinetobacter Infections/pathology
- Acinetobacter baumannii/pathogenicity
- Animals
- Cells, Cultured
- Disease Models, Animal
- Interleukin-6/genetics
- Interleukin-6/metabolism
- Lung/enzymology
- Lung/microbiology
- Lung/pathology
- Macrophages, Alveolar/enzymology
- Macrophages, Alveolar/microbiology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mitogen-Activated Protein Kinase 8/genetics
- Mitogen-Activated Protein Kinase 8/metabolism
- Neutrophil Infiltration
- Nitric Oxide Synthase Type II/metabolism
- Pneumonia, Ventilator-Associated/enzymology
- Pneumonia, Ventilator-Associated/microbiology
- Pneumonia, Ventilator-Associated/pathology
- Respiration, Artificial/adverse effects
- Signal Transduction
- Tumor Necrosis Factor-alpha/metabolism
- Vascular Cell Adhesion Molecule-1/genetics
- Vascular Cell Adhesion Molecule-1/metabolism
- Ventilator-Induced Lung Injury/enzymology
- Ventilator-Induced Lung Injury/microbiology
- Ventilator-Induced Lung Injury/pathology
- Mice
Collapse
Affiliation(s)
- Tzyy-Bin Tsay
- Department of Surgery, Kaohsiung Armed Forces General Hospital Zuoying Branch, Kaohsiung, Taiwan
| | - Wan-Hsuan Chang
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Ching-Mei Hsu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Lee-Wei Chen
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan.
- Department of Surgery, Kaohsiung Veterans General Hospital, 386, Ta-Chung 1st Road, Kaohsiung, Taiwan.
- Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
4
|
Protective effect of combination of anakinra and MCC950 against acute lung injury is achieved through suppression of the NF-κB-mediated-MAPK and NLRP3-caspase pathways. Int Immunopharmacol 2021; 97:107506. [PMID: 34022766 DOI: 10.1016/j.intimp.2021.107506] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/03/2021] [Accepted: 02/11/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND It has been uncovered that the interleukin-1 receptor antagonist anakinra and the NLRP3 inflammasome blocker MCC950 can alleviate acute lung injury (ALI). However, the specific mechanism underlying these effects remains unknown. Thus, we sought to investigate the effects of anakinra and MCC950 in ALI as well as the molecular mechanisms. METHODS We treated C57BL/6 mice with aerosols of anakinra and/or MCC950 along with lipopolysaccharide (LPS), followed by mechanical ventilation (MV) treatment after 1.5 h of inhalation of aforementioned compounds. Lung injury was assessed by determining the level of inflammatory factors in the alveolar lavage fluid and monitoring blood oxygen saturation. We confirmed our findings of regulation of the ALI model through the nuclear factor-κB (NF-κB)/mitogen-activated protein kinase (MAPK)/nucleotide binding domain and leucine-rich repeat (NLR) pyrin domain containing 3 (NLRP3)-caspase pathway in further studies with RelA-/- mice. RESULTS Combined treatment of anakinra and MCC950 presented the best therapeutic effect on LPS and MV-induced ALI than did treatment with anakinra or MCC950 alone. Combined therapy with anakinra and MCC950 suppressed MAPK and NLRP3-caspase via inhibition of the NF-κB pathway to improve ALI, but the therapeutic pathway was revoked by knockout of NF-κB. CONCLUSION Taken together, combined treatment of anakinra and MCC950 was effective in alleviating ALI in the mouse model, highlighting a new insight into ALI treatment.
Collapse
|
5
|
Monjezi M, Jamaati H, Noorbakhsh F. Attenuation of ventilator-induced lung injury through suppressing the pro-inflammatory signaling pathways: A review on preclinical studies. Mol Immunol 2021; 135:127-136. [PMID: 33895577 DOI: 10.1016/j.molimm.2021.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/28/2021] [Accepted: 04/06/2021] [Indexed: 10/21/2022]
Abstract
Mechanical ventilation (MV) is a relatively common medical intervention in ICU patients. The main side effect of MV is the so-called "ventilator-induced lung injury" (VILI). The pathogenesis of VILI is not completely understood; however, it has been reported that MV might be associated with up-regulation of various inflammatory mediators within the lung tissue and that these mediators might act as pathogenic factors in lung tissue injury. One potential mechanism for the generation of inflammatory mediators is through the release of endogenous molecules known as damage associated molecular patterns (DAMPs). These molecules are released from injured tissues and can bind to pattern recognition receptors (PRRs). PRR activation generally leads to the production and release of inflammation-related molecules including innate immune cytokines and chemokines. It has been suggested that blocking DAMP/PRR signaling pathways might diminish the progression of VILI. Herein, we review the latest findings with regard to the effects of DAMP/PRRs and their blockade, as well as the potential therapeutic targets and future research directions in VILI. Results of studies performed on human samples, animal models of disease, as well as relevant in vitro systems will be discussed.
Collapse
Affiliation(s)
- Mojdeh Monjezi
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Jamaati
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Transcriptome-Wide Gene Expression in a Murine Model of Ventilator-Induced Lung Injury. DISEASE MARKERS 2021; 2021:5535890. [PMID: 33927789 PMCID: PMC8049808 DOI: 10.1155/2021/5535890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 11/24/2022]
Abstract
Background Mechanical ventilation could lead to ventilator-induced lung injury (VILI), but its underlying pathogenesis remains largely unknown. In this study, we aimed to determine the genes which were highly correlated with VILI as well as their expressions and interactions by analyzing the differentially expressed genes (DEGs) between the VILI samples and controls. Methods GSE11434 was downloaded from the gene expression omnibus (GEO) database, and DEGs were identified with GEO2R. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were conducted using DAVID. Next, we used the STRING tool to construct protein-protein interaction (PPI) network of the DEGs. Then, the hub genes and related modules were identified with the Cytoscape plugins: cytoHubba and MCODE. qRT-PCR was further used to validate the results in the GSE11434 dataset. We also applied gene set enrichment analysis (GSEA) to discern the gene sets that had a significant difference between the VILI group and the control. Hub genes were also subjected to analyses by CyTargetLinker and NetworkAnalyst to predict associated miRNAs and transcription factors (TFs). Besides, we used CIBERSORT to detect the contributions of different types of immune cells in lung tissues of mice in the VILI group. By using DrugBank, small molecular compounds that could potentially interact with hub genes were identified. Results A total of 141 DEGs between the VILI group and the control were identified in GSE11434. Then, seven hub genes were identified and were validated by using qRT-PCR. Those seven hub genes were largely enriched in TLR and JAK-STAT signaling pathways. GSEA showed that VILI-associated genes were also enriched in NOD, antigen presentation, and chemokine pathways. We predicted the miRNAs and TFs associated with hub genes and constructed miRNA-TF-gene regulatory network. An analysis with CIBERSORT showed that the proportion of M0 macrophages and activated mast cells was higher in the VILI group than in the control. Small molecules, like nadroparin and siltuximab, could act as potential drugs for VILI. Conclusion In sum, a number of hub genes associated with VILI were identified and could provide novel insights into the pathogenesis of VILI and potential targets for its treatment.
Collapse
|
7
|
Nikitopoulou I, Ninou I, Manitsopoulos N, Dimopoulou I, Orfanos SE, Aidinis V, Kotanidou A. A role for bronchial epithelial autotaxin in ventilator-induced lung injury. Intensive Care Med Exp 2021; 9:12. [PMID: 33778909 PMCID: PMC8005331 DOI: 10.1186/s40635-021-00379-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/22/2021] [Indexed: 01/13/2023] Open
Abstract
Background The pathophysiology of acute respiratory distress syndrome (ARDS) may eventually result in heterogeneous lung collapse and edema-flooded airways, predisposing the lung to progressive tissue damage known as ventilator-induced lung injury (VILI). Autotaxin (ATX; ENPP2), the enzyme largely responsible for extracellular lysophosphatidic acid (LPA) production, has been suggested to play a pathogenic role in, among others, pulmonary inflammation and fibrosis. Methods C57BL/6 mice were subjected to low and high tidal volume mechanical ventilation using a small animal ventilator: respiratory mechanics were evaluated, and plasma and bronchoalveolar lavage fluid (BALF) samples were obtained. Total protein concentration was determined, and lung histopathology was further performed Results Injurious ventilation resulted in increased BALF levels of ATX. Genetic deletion of ATX from bronchial epithelial cells attenuated VILI-induced pulmonary edema. Conclusion ATX participates in VILI pathogenesis.
Collapse
Affiliation(s)
- Ioanna Nikitopoulou
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece
| | - Ioanna Ninou
- Institute of Immunology, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| | - Nikolaos Manitsopoulos
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece
| | - Ioanna Dimopoulou
- 1st Department of Critical Care Medicine & Pulmonary Services, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece
| | - Stylianos E Orfanos
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece.,2nd Department of Critical Care, National and Kapodistrian University of Athens Medical School, Attikon" Hospital, Athens, Greece
| | - Vassilis Aidinis
- Institute of Immunology, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| | - Anastasia Kotanidou
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece. .,1st Department of Critical Care Medicine & Pulmonary Services, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece.
| |
Collapse
|
8
|
Hong W, Zhi FX, Kun TH, Hua FJ, Huan Ling L, Fang F, Wen C, Jie W, Yang LC. 6-Gingerol attenuates ventilator-induced lung injury via anti-inflammation and antioxidative stress by modulating the PPARγ/NF-κBsignalling pathway in rats. Int Immunopharmacol 2021; 92:107367. [PMID: 33461160 DOI: 10.1016/j.intimp.2021.107367] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/08/2020] [Accepted: 01/04/2021] [Indexed: 02/05/2023]
Abstract
Although mechanical ventilation (MV) is indispensable to life-support therapy in critically ill patients, it may promote or aggravatelunginjury known asventilator-inducedlunginjury(VILI). 6-Gingerol is the principal ingredient of ginger with potential anti-inflammatory and antioxidant properties in various diseases. Nevertheless, the role and mechanism of 6-gingerol in the process of VILI has not been explicitly investigated. In the study, we found that pre-treatment with 6-gingerol significantly improved the histological changes and pulmonary oedema, inhibited neutrophil accumulation and the release of early pro-inflammatory cytokines and MPO, and reduced oxidative stress reactions after high MV. Moreover, 6-gingerol treatment also increased PPARγ expression and decreased NF-κB activation in rats subjected to high MV. Furthermore, GW9662, a specific PPARγ inhibitor, was demonstrated to activatethe NF-κB pathway and cancele the protective role of 6-gingerol in VILI. This indicates that 6-gingerol exerted anti-inflammatory and antioxidative stress effects in VILI by activating PPARγ and inhibiting the NF-κBsignalling pathway.
Collapse
Affiliation(s)
- Wei Hong
- Department of Anesthesiology, Huazhong University of Science and Technology Union ShenZhen Hospital, ShenZhen, China; Department of Anesthesiology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China
| | - Fang Xiang Zhi
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Anesthesiology, Subei People's Hospital, YangZhou 225001, China
| | - Tu Han Kun
- Department of Anesthesiology, Huazhong University of Science and Technology Union ShenZhen Hospital, ShenZhen, China; Department of Anesthesiology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China
| | - Feng Jie Hua
- Department of Anesthesiology, Huazhong University of Science and Technology Union ShenZhen Hospital, ShenZhen, China; Department of Anesthesiology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China
| | - Li Huan Ling
- Department of Anesthesiology, Huazhong University of Science and Technology Union ShenZhen Hospital, ShenZhen, China; Department of Anesthesiology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China
| | - Fang Fang
- Department of General Medicine, Huazhong University of Science and Technology Union ShenZhen Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China
| | - Chen Wen
- Department of Anesthesiology, Yuebei People's Hospital, Shantou University Medical College, Shaoguan, Guangdong Province, China
| | - Wang Jie
- Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Li Chao Yang
- Department of Anesthesiology, Huazhong University of Science and Technology Union ShenZhen Hospital, ShenZhen, China; Department of Anesthesiology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, ShenZhen, China.
| |
Collapse
|
9
|
Zhu CH, Yu J, Wang BQ, Nie Y, Wang L, Shan SQ. Dexmedetomidine reduces ventilator-induced lung injury via ERK1/2 pathway activation. Mol Med Rep 2020; 22:5378-5384. [PMID: 33173983 PMCID: PMC7647005 DOI: 10.3892/mmr.2020.11612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Mechanical ventilation (MV) can contribute to ventilator-induced lung injury (VILI); dexmedetomidine (Dex) treatment attenuates MV-related pulmonary inflammation, but the mechanisms remain unclear. Therefore, the present study aimed to explore the protective effect and the possible molecular mechanisms of Dex in a VILI rodent model. Adult male Sprague-Dawley rats were randomly assigned to one of seven groups (n=24 rats/group). Rats were euthanized after 4 h of continuous MV, and pathological changes, lung wet/dry (W/D) weight ratio, the levels of inflammatory cytokines (IL-1β, TNF-α and IL-6) in the bronchoalveolar lavage fluid (BALF), and the expression levels of Bcl-2 homologous antagonist/killer (Bak), Bcl-2, pro-caspase-3, cleaved caspase-3 and the phosphorylation of ERK1/2 in the lung tissues were measured. Propidium iodide uptake and TUNEL staining were used to detect epithelial cell death. The Dex pretreatment group exhibited fewer pathological changes, lower W/D ratios and lower expression levels of inflammatory cytokines in BALF compared with the VILI group. Dex significantly attenuated the ratio of Bak/Bcl-2, cleaved caspase-3 expression levels and epithelial cell death, and increased the expression of phosphorylated ERK1/2. The protective effects of Dex could be partially reversed by PD98059, which is a mitogen-activated protein kinase (upstream of ERK1/2) inhibitor. Overall, dexmedetomidine was found to reduce the inflammatory response and epithelial cell death caused by VILI, via the activation of the ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Chun-Hua Zhu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Jian Yu
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Ben-Qing Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Yu Nie
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Lei Wang
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| | - Shi-Qiang Shan
- Department of Anesthesiology, Cangzhou Central Hospital, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
10
|
Fazza TF, Pinheiro BV, da Fonseca LMC, Sergio LPDS, Botelho MP, Lopes GDM, de Paoli F, da Fonseca ADS, Lucinda LMF, Reboredo MM. Effect of low-level laser therapy on the inflammatory response in an experimental model of ventilator-induced lung injury. Photochem Photobiol Sci 2020; 19:1356-1363. [PMID: 32761018 PMCID: PMC8047601 DOI: 10.1039/d0pp00053a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The effect of low-level laser therapy (LLLT) on an experimental model of ventilator-induced lung injury (VILI) was evaluated in this study. 24 adult Wistar rats were randomized into four groups: protective mechanical ventilation (PMV), PMV + laser, VILI and VILI + laser. The animals of the PMV and VILI groups were ventilated with tidal volumes of 6 and 35 ml kg−1, respectively, for 90 minutes. After the first 60 minutes of ventilation, the animals in the laser groups were irradiated (808 nm, 100 mW power density, 20 J cm−2 energy density, continuous emission mode, and exposure time of 5 s) and after 30 minutes of irradiation, the animals were euthanized. Lung samples were removed for morphological analysis, bronchoalveolar lavage (BAL) and real time quantitative polynucleotide chain reaction (RT-qPCR). The VILI group showed a greater acute lung injury (ALI) score with an increase in neutrophil infiltration, higher neutrophil count in the BAL fluid and greater cytokine mRNA expression compared to the PMV groups (p < 0.05). The VILI ± laser group when compared to the VILI group showed a lower ALI score (0.35 ± 0.08 vs. 0.54 ± 0.13, p < 0.05), alveolar neutrophil infiltration (7.00 ± 5.73 vs. 21.50 ± 9.52, p < 0.05), total cell count (1.90 ± 0.71 vs. 4.09 ± 0.96 × 105, p < 0.05) and neutrophil count in the BAL fluid (0.60 ± 0.37 vs. 2.28 ± 0.48 × 105, p < 0.05). Moreover, LLLT induced a decrease in pro-inflammatory and an increase of anti-inflammatory mRNA levels compared to the VILI group (p < 0.05). In conclusion, LLLT was found to reduce the inflammatory response in an experimental model of VILI.
Collapse
Affiliation(s)
- Thaís Fernanda Fazza
- Pulmonary Research Laboratory, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil. and Center of Reproductive Biology, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Bruno Valle Pinheiro
- Pulmonary Research Laboratory, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil. and Center of Reproductive Biology, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Lídia Maria Carneiro da Fonseca
- Pulmonary Research Laboratory, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil. and Center of Reproductive Biology, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Luiz Philippe da Silva Sergio
- Department of Biophysics and Biometry, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mateus Pinto Botelho
- Pulmonary Research Laboratory, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | - Gabrielle de Moura Lopes
- Pulmonary Research Laboratory, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | - Flavia de Paoli
- Department of Morphology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Adenilson de Souza da Fonseca
- Department of Biophysics and Biometry, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leda Marília Fonseca Lucinda
- Pulmonary Research Laboratory, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil. and Center of Reproductive Biology, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil and Department of Morphology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Maycon Moura Reboredo
- Pulmonary Research Laboratory, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil. and Center of Reproductive Biology, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| |
Collapse
|
11
|
Dexmedetomidine alleviates non-ventilation associated lung injury via modulating immunology phenotypes of macrophages. Life Sci 2020; 259:118249. [PMID: 32798558 DOI: 10.1016/j.lfs.2020.118249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 12/15/2022]
Abstract
AIMS We aimed to evaluate the effect of Dexmedetomidine (Dex) on immunology function of macrophages and inflammatory reactions in non-ventilated lung tissues from both humans and rats. MAIN METHODS Patients scheduled for lung lobectomy were randomly assigned to traditional anesthesia group or Dex anesthesia group, 15 subjects in each group. CD68, CD86 and CD206 were used to mark activate and polarized macrophages using immunofluorescence staining in human lung tissues. Sprague-Dawley rats were used to set lung injury model and randomly divided into Control group, one-lung ventilation group (CLI group) and CLI + Dex group. Lung tissues and bronchoalveolar lavage fluid (BALF) from non-ventilated lungs were collected. The acquired lung tissues were subjected to hematoxylin-eosin (H&E) staining and the inflammatory cells in BALF were calculated. Levels of cytokines and chemokines were detected by enzyme-linked immunosorbent assays (ELISA). KEY FINDINGS Results from humans showed that anesthesia with Dex decreased the number of both CD68 positive cells and CD86 positive cells and down-regulated level of pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and monocyte chemotactic protein 1 (MCP-1) in human lung. Results from rats demonstrated that treatment with Dex reversed the increased inflammatory cells in lung and the increased levels of TNF-α, interleukin-1β (IL-β), MCP-1 and chemokine (C-X-C motif) ligand 1 (CXCL1) resulted from non-ventilation; Dex increased the anti-inflammatory cytokine interleukin-10 (IL-10) in BALF from non-ventilated lung. SIGNIFICANCE This study showed that Dex modulated the activation and immunological function of macrophages in non-ventilated lung and revealed a protective role in collapsed lung injury.
Collapse
|
12
|
da Silva ACL, de Matos NA, de Souza ABF, Castro TDF, Cândido LDS, Oliveira MADGS, Costa GDP, Talvani A, Cangussú SD, Bezerra FS. Sigh maneuver protects healthy lungs during mechanical ventilation in adult Wistar rats. Exp Biol Med (Maywood) 2020; 245:1404-1413. [PMID: 32640895 DOI: 10.1177/1535370220940995] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mechanical ventilation (MV) is a tool used for the treatment of patients with acute or chronic respiratory failure. However, MV is a non-physiological resource, and it can cause metabolic disorders such as release of pro-inflammatory cytokines and production of reactive oxygen species. In clinical setting, maneuvers such as sigh, are used to protect the lungs. Thus, this study aimed to evaluate the effects of sigh on oxidative stress and lung inflammation in healthy adult Wistar rats submitted to MV. Male Wistar rats were divided into four groups: control (CG), mechanical ventilation (MVG), MV set at 20 sighs/h (MVG20), and MV set at 40 sighs/h (MVG40). The MVG, MVG20, and MVG40 were submitted to MV for 1 h. After the protocol, all animals were euthanized and the blood, bronchoalveolar lavage fluid, and lungs were collected for subsequent analysis. In the arterial blood, MVG40 presented higher partial pressure of oxygen and lower partial pressure of carbon dioxide compared to control. The levels of bicarbonate in MVG20 were lower compared to CG. The neutrophil influx in bronchoalveolar lavage fluid was higher in the MVG compared to CG and MVG40. In the lung parenchyma, the lipid peroxidation was higher in MVG compared to CG, MVG20, and MVG40. Superoxide dismutase and catalase activity were higher in MVG compared to CG, MVG20, and MVG40. The levels of IL-1, IL-6, and TNF in the lung homogenate were higher in MVG compared to CG, MVG20, and MVG40. The use of sigh plays a protective role as it reduced redox imbalance and pulmonary inflammation caused by MV.
Collapse
Affiliation(s)
- Andréa Cristiane Lopes da Silva
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Natália Alves de Matos
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Ana Beatriz Farias de Souza
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Thalles de Freitas Castro
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Leandro da Silva Cândido
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Michel Angelo das Graças Silva Oliveira
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Guilherme de Paula Costa
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Sílvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences (DECBI), Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto (UFOP), Ouro Preto, MG 35400-000, Brazil
| |
Collapse
|
13
|
Abstract
BACKGROUND This study hypothesized that, in experimental mild acute respiratory distress syndrome, lung damage caused by high tidal volume (VT) could be attenuated if VT increased slowly enough to progressively reduce mechanical heterogeneity and to allow the epithelial and endothelial cells, as well as the extracellular matrix of the lung to adapt. For this purpose, different strategies of approaching maximal VT were tested. METHODS Sixty-four Wistar rats received Escherichia coli lipopolysaccharide intratracheally. After 24 h, animals were randomly assigned to receive mechanical ventilation with VT = 6 ml/kg for 2 h (control); VT = 6 ml/kg during hour 1 followed by an abrupt increase to VT = 22 ml/kg during hour 2 (no adaptation time); VT = 6 ml/kg during the first 30 min followed by a gradual VT increase up to 22 ml/kg for 30 min, then constant VT = 22 ml/kg during hour 2 (shorter adaptation time); and a more gradual VT increase, from 6 to 22 ml/kg during hour 1 followed by VT = 22 ml/kg during hour 2 (longer adaptation time). All animals were ventilated with positive end-expiratory pressure of 3 cm H2O. Nonventilated animals were used for molecular biology analysis. RESULTS At 2 h, diffuse alveolar damage score and heterogeneity index were greater in the longer adaptation time group than in the control and shorter adaptation time animals. Gene expression of interleukin-6 favored the shorter (median [interquartile range], 12.4 [9.1-17.8]) adaptation time compared with longer (76.7 [20.8 to 95.4]; P = 0.02) and no adaptation (65.5 [18.1 to 129.4]) time (P = 0.02) strategies. Amphiregulin, metalloproteinase-9, club cell secretory protein-16, and syndecan showed similar behavior. CONCLUSIONS In experimental mild acute respiratory distress syndrome, lung damage in the shorter adaptation time group compared with the no adaptation time group was attenuated in a time-dependent fashion by preemptive adaptation of the alveolar epithelial cells and extracellular matrix. Extending the adaptation period increased cumulative power and did not prevent lung damage, because it may have exposed animals to injurious strain earlier and for a longer time, thereby negating any adaptive benefit.
Collapse
|
14
|
Yin D, Wang W, Han W, Fan C. Targeting Notch-activated M1 macrophages attenuate lung tissue damage in a rat model of ventilator induced lung injury. Int J Mol Med 2019; 44:1388-1398. [PMID: 31432103 PMCID: PMC6713421 DOI: 10.3892/ijmm.2019.4315] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 06/27/2019] [Indexed: 12/26/2022] Open
Abstract
Ventilator induced lung injury (VILI) may be involved in the activation of alveolar macrophages. The purpose of this study was to investigate the relationship between the Notch signaling pathway and macrophage polarization in VILI. The VILI model was established using rats. Hematoxylineosin staining was used to test the lung tissue morphology. Bicinchoninic acid assay and ELISA were performed to detect protein and tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-10 levels in bronchoalveolar lavage fluids (BALF), respectively. The ratio of alveolar M1 and M2 macrophages was detected by flow cytometry. The mRNA and protein expression levels of Notch pathway-related proteins were determined using reverse transcription-quantitative PCR and western blotting. The present study found that high-frequency mechanical ventilation could cause pulmonary edema and increase the levels of protein, TNF-α and IL-6 in BALF while decreasing the level of IL-10 in BALF. High-frequency mechanical ventilation also induced polarization of alveolar macrophages to M1. The results also showed a significant increase in the levels of Notch pathway-related proteins including notch intracellular domain, Hes1, Hes5 and Hey1. Injection of N-[N-(3,5-difluorophenylacetyl)-1-alanyl] phenylglycine t-butyl ester could inhibit the Notch pathway and such an inhibition protected lung tissue and reduced lung inflammation caused by mechanical ventilation. After the Notch pathway was inhibited, the level of M1 polarization of macrophages caused by high-frequency mechanical ventilation was reduced. VILI caused pulmonary inflammation and macrophages to polarize to M1 and upregulated the expression levels of Notch pathway-related proteins. The inhibition of Notch pathway also reduced the proportion of M1 macrophages and inflammatory responses.
Collapse
Affiliation(s)
- Danping Yin
- Department of Disease Prevention and Control, No. 960 Hospital of PLA, Jinan, Shandong 250031, P.R. China
| | - Weiming Wang
- Electrocardiogram Room, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong 264001, P.R. China
| | - Wei Han
- Department of Training, No. 960 Hospital of PLA, Jinan, Shandong 250031, P.R. China
| | - Chen Fan
- Department of Laboratory Diagnosis, No. 960 Hospital of PLA, Jinan, Shandong 250031, P.R. China
| |
Collapse
|
15
|
Wan B, Xu WJ, Zhan P, Jin JJ, Xi GM, Chen MZ, Hu YB, Zhu SH, Liu HB, Wang XX, Zhang XW, Lv TF, Song Y. Topotecan alleviates ventilator-induced lung injury via NF-κB pathway inhibition. Cytokine 2018; 110:381-388. [DOI: 10.1016/j.cyto.2018.04.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/26/2018] [Accepted: 04/15/2018] [Indexed: 11/25/2022]
|
16
|
Kiss T, Silva PL, Huhle R, Moraes L, Santos RS, Felix NS, Santos CL, Morales MM, Capelozzi VL, Kasper M, Pelosi P, Gama de Abreu M, Rocco PRM. Comparison of different degrees of variability in tidal volume to prevent deterioration of respiratory system elastance in experimental acute lung inflammation. Br J Anaesth 2018; 116:708-15. [PMID: 27106975 DOI: 10.1093/bja/aew093] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2015] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Variable ventilation improves respiratory function, but it is not known whether the amount of variability in tidal volume (VT) can be reduced in recruited lungs without a deterioration of respiratory system elastance. METHODS Acute lung inflammation was induced by intratracheal instillation of lipopolysaccharide in 35 Wistar rats. Twenty-eight animals were anaesthetized and ventilated in volume-controlled mode. Lungs were recruited by random variation of VT (mean 6 ml kg(-1), coefficient of variation 30%, normal distribution) for 30 min. Animals were randomly assigned to different amounts of VT variability (n=7 for 90 min per group): 30, 15, 7.5, or 0%. Lung function, diffuse alveolar damage, and gene expression of biological markers associated with cell mechanical stress, inflammation, and fibrogenesis were assessed. Seven animals were not ventilated and served as controls for post-mortem analyses. RESULTS A VT variability of 30%, but not 15, 7.5, or 0%, prevented deterioration of respiratory system elastance [Mean (SD) -7.5 (8.7%), P<0.05; 21.1 (9.6%), P<0.05; 43.3 (25.9), P<0.05; and 41.2 (16.4), P<0.05, respectively]. Diffuse alveolar damage was lower with a VT variability of 30% than with 0% and without ventilation, because of reduced oedema and haemorrhage. A VT variability of 30, 15, or 7.5% reduced the gene expression of amphiregulin, cytokine-induced neutrophil chemoattractant-1, and tumour necrosis factor α compared with a VT variability of 0%. CONCLUSIONS In this model of acute lung inflammation, a VT variability of 30%, compared with 15 and 7.5%, was necessary to avoid deterioration of respiratory system elastance and was not associated with lung histological damage.
Collapse
Affiliation(s)
- T Kiss
- Pulmonary Engineering Group, Department of Anesthesiology and Intensive Care Therapy, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| | - P L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| | - R Huhle
- Pulmonary Engineering Group, Department of Anesthesiology and Intensive Care Therapy, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - L Moraes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| | - R S Santos
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| | - N S Felix
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| | - C L Santos
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| | - M M Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| | - V L Capelozzi
- Department of Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - M Kasper
- Institute of Anatomy, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - P Pelosi
- Department of Surgical Sciences and Integrated Diagnostics, IRCCS AOU San Martino-IST, University of Genoa, Genoa, Italy
| | - M Gama de Abreu
- Pulmonary Engineering Group, Department of Anesthesiology and Intensive Care Therapy, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - P R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Chen L, Xia HF, Shang Y, Yao SL. Molecular Mechanisms of Ventilator-Induced Lung Injury. Chin Med J (Engl) 2018; 131:1225-1231. [PMID: 29553050 PMCID: PMC5956775 DOI: 10.4103/0366-6999.226840] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Mechanical ventilation (MV) has long been used as a life-sustaining approach for several decades. However, researchers realized that MV not only brings benefits to patients but also cause lung injury if used improperly, which is termed as ventilator-induced lung injury (VILI). This review aimed to discuss the pathogenesis of VILI and the underlying molecular mechanisms. DATA SOURCES This review was based on articles in the PubMed database up to December 2017 using the following keywords: "ventilator-induced lung injury", "pathogenesis", "mechanism", and "biotrauma". STUDY SELECTION Original articles and reviews pertaining to mechanisms of VILI were included and reviewed. RESULTS The pathogenesis of VILI was defined gradually, from traditional pathological mechanisms (barotrauma, volutrauma, and atelectrauma) to biotrauma. High airway pressure and transpulmonary pressure or cyclic opening and collapse of alveoli were thought to be the mechanisms of barotraumas, volutrauma, and atelectrauma. In the past two decades, accumulating evidence have addressed the importance of biotrauma during VILI, the molecular mechanism underlying biotrauma included but not limited to proinflammatory cytokines release, reactive oxygen species production, complement activation as well as mechanotransduction. CONCLUSIONS Barotrauma, volutrauma, atelectrauma, and biotrauma contribute to VILI, and the molecular mechanisms are being clarified gradually. More studies are warranted to figure out how to minimize lung injury induced by MV.
Collapse
Affiliation(s)
- Lin Chen
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Hai-Fa Xia
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - You Shang
- Department of Critical Care Medicine, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Shang-Long Yao
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| |
Collapse
|
18
|
Chen H, Sun X, Yang X, Hou Y, Yu X, Wang Y, Wu J, Liu D, Wang H, Yu J, Yi W. Dexmedetomidine reduces ventilator-induced lung injury (VILI) by inhibiting Toll-like receptor 4 (TLR4)/nuclear factor (NF)-κB signaling pathway. Bosn J Basic Med Sci 2018; 18:162-169. [PMID: 29510084 DOI: 10.17305/bjbms.2018.2400] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/14/2017] [Accepted: 11/14/2017] [Indexed: 12/30/2022] Open
Abstract
Mechanical ventilation (MV) may lead to ventilator-induced lung injury (VILI). Previous research has shown that dexmedetomidine attenuates pulmonary inflammation caused by MV, but the underlying mechanisms remain unclear. Our study aims to test whether dexmedetomidine has a protective effect against VILI and to explore the possible molecular mechanisms using the rat model. Thirty adult male Wistar rats weighing 200-250 g were randomly assigned to 5 groups (n = 6): control, low tidal volume MV (LMV), high tidal volume (HVT) MV (HMV), HVT MV + dexmedetomidine (DEX), HVT MV + dexmedetomidine + yohimbine (DEX+Y). Rats were euthanized after being ventilated for 4 hours. Pathological changes, lung wet/dry (W/D) weight ratio, lung myeloperoxidase (MPO) activity, levels of inflammatory cytokines (i.e., interleukin [IL]-1β, tumor necrosis factor alpha [TNF-α], and IL-6) in the bronchoalveolar lavage fluid (BALF) and lung tissues, expression of Toll-like receptor 4 (TLR4) and nuclear factor (NF)-κB, and activation of NF-κB in lung tissues were measured. Compared with HMV, DEX group showed fewer pathological changes, lower W/D ratios and decreased MPO activity of the lung tissues and lower concentrations of the inflammatory cytokines in the BALF and lung tissues. Dexmedetomidine significantly inhibited the expression of TLR4 and NF-κB and activation of NF-κB. Yohimbine partly alleviated the effects of dexmedetomidine. Dexmedetomidine reduced the inflammatory response to HVT-MV and had a protective effect against VILI, with the inhibition of the TLR4/NF-κB signaling pathway, at least partly via α2-adrenoceptors.
Collapse
Affiliation(s)
- Hongli Chen
- Department of Anesthesiology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Gu C, Dai C, Sun Y, Liu M, Wang Y, Wu X. P120 regulates beta-catenin nuclear translocation through E-cadherin endocytosis in ventilator-induced lung injury. Oncotarget 2018; 7:83859-83868. [PMID: 27911872 PMCID: PMC5356630 DOI: 10.18632/oncotarget.13724] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 11/21/2016] [Indexed: 12/15/2022] Open
Abstract
Mechanical stretch induces epithelial barrier dysfunction by altering the location and degradation of cellular junction proteins. p120-catenin (p120) is a cell-cell junction protein known to protect against ventilator-induced lung injury (VILI) that results from improper ventilation of patients. In this study, we sought to determine the role of p120 in VILI and its relationship with the cellular response to mechanical stretch. Mouse lung epithelial cells (MLE-12) transfected with p120 siRNA, p120 cDNA, or E-cadherin siRNA were subjected to 20% cyclic stretch for 2 or 4 hours. Wild-type male C57BL/6 mice were transfected with p120 siRNA-liposome complex to delete p120 in vivo and then subjected to mechanical ventilation. Cyclic stretch induced p120 degradation and the endocytosis of E-cadherin, which induced β-catenin translocation into the nucleus, a key event in lung injury progress and repair. These findings reveal that by reducing β-catenin nuclear translocation through inhibition of E-cadherin endocytosis, p120 protects against ventilator-induced lung injury.
Collapse
Affiliation(s)
- Changping Gu
- Department of Anesthesiology, Qianfoshan Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Chenyang Dai
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Yongtao Sun
- Department of Anesthesiology, Qianfoshan Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Mengjie Liu
- Department of Anesthesiology, Qianfoshan Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Yuelan Wang
- Department of Anesthesiology, Qianfoshan Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Xinyi Wu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
20
|
SN50, a Cell-Permeable Inhibitor of Nuclear Factor-κB, Attenuates Ventilator-Induced Lung Injury in an Isolated and Perfused Rat Lung Model. Shock 2018; 46:194-201. [PMID: 26780513 DOI: 10.1097/shk.0000000000000563] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High tidal volume (VT) ventilation causes the release of various mediators and results in ventilator-induced lung injury (VILI). SN50, a cell-permeable nuclear factor-κB (NF-κB) inhibitory peptide, attenuates inflammation and acute respiratory distress syndrome. However, the mechanisms associated with the effects of SN50 in VILI have not been fully elucidated. We investigated the cellular and molecular mechanisms for the effects of SN50 treatment in VILI. An isolated and perfused rat lung model was exposed to low (5 mL/kg) or high (15 mL/kg) VT ventilation for 6 h. SN50 was administered in the perfusate at the onset of the high-stretch mechanical ventilation. The hemodynamics, lung histological changes, inflammatory responses, and activation of apoptotic pathways were evaluated. VILI was demonstrated by increased pulmonary vascular permeability and lung weight gain, as well as by increased levels of interleukin (IL)-1β, tumor necrosis factor (TNF)-α, myeloperoxidase (MPO), hydrogen peroxide, and macrophage inflammatory protein-2 in the bronchoalveolar lavage fluid. The lung tissue expression of TNF-α, IL-1β, mitogen-activated protein kinases (MAPKs), caspase-3, and phosphorylation of serine/threonine-specific protein kinase (p-AKT) was greater in the high VT group than in the low VT group. Upregulation and activation of NF-κB was associated with increased lung injury in VILI. SN50 attenuated the inflammatory responses, including the expression of IL-1β, TNF-α, MPO, MAPKs, and NF-κB. In addition, the downregulation of apoptosis was evaluated using caspase-3 and p-AKT expression. Furthermore, SN50 mitigated the increases in the lung weights, pulmonary vascular permeability, and lung injury. In conclusion, VILI is associated with inflammatory responses and activation of NF-κB. SN50 inhibits the activation of NF-κB and attenuates VILI.
Collapse
|
21
|
Fang XZ, Ge YL, Li M, Huang TF, Yang Z, Gao J. Preconditioning of physiological cyclic stretch inhibits the inflammatory response induced by pathologically mechanical stretch in alveolar epithelial cells. Exp Ther Med 2017; 15:2172-2176. [PMID: 29434821 DOI: 10.3892/etm.2017.5611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 04/28/2017] [Indexed: 01/09/2023] Open
Abstract
The aim of the present study was to investigate the effects of preconditioning of physiological cyclic stretch (CS) on the overexpression of early pro-inflammatory cytokines [including tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-8] during the inflammatory response induced by pathologically mechanical stretch in lung epithelial cells, and to determine its molecular mechanism of action. Cells were subjected to 5% CS for various durations (0, 15, 30, 60 and 120 min) prior to 6 h treatment with pathological 20% CS. In a separate experiment, cells were preconditioned with physiological 5% CS or incubated with a nuclear factor (NF)-κB inhibitor, pyrroldine dithiocarbamate (PDTC). The expression levels of inflammatory mediators were measured using reverse transcription-quantitative polymerase chain reaction. NF-κB was quantified using western blot analysis. Preconditioning with physiological 5% CS for 30, 60 and 120 min was demonstrated to significantly attenuate the release of pathologically mechanical stretch-induced early pro-inflammatory cytokines (TNF-α, IL-1β and IL-8) in alveolar epithelial cells (P<0.05) and significantly reduce the expression of NF-κB (P<0.05). Peak suppression was observed in cells preconditioned for 60 min. In the second set of experiments, it was demonstrated that mechanical stretch-induced release of TNF-α, IL-1β and IL-8 was significantly inhibited by both PDTC pretreatment and 5% CS pretreatment alone (all P<0.05). Furthermore, significant inhibition was also observed when both 5% CS pretreatment and PDTC pretreatment was used on mechanical stretch-induced cells (P<0.05), which was markedly greater than the inhibition induced by either pretreatment alone. The present findings suggest that preconditioning with physiological 5% CS is able to inhibit the inflammatory response induced by pathologically mechanical stretch in alveolar epithelial cells. These anti-inflammatory effects are induced, at least in part, by suppressing the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xiang-Zhi Fang
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - Ya-Li Ge
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - Min Li
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Jiangsu 212000, P.R. China
| | - Tian-Feng Huang
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - Zhang Yang
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| | - Ju Gao
- Department of Anesthesiology, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, P.R. China
| |
Collapse
|
22
|
Du B, Gao M, Cao L, Song J, Shi X. Role of notch signaling pathway in mechanical ventilation induced lung injury. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8106-8118. [PMID: 31966663 PMCID: PMC6965473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/28/2017] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To investigate the expression of Notch signaling pathway after mechanical ventilation and its role in mechanical ventilation and lung injury. METHODS VILI model was established by mechanical ventilation at high tidal volume (VT = 20 ml/kg) for 4 hours. Lung injury and inflammatory response were evaluated. Hes-1 and Hes-5 mRNA levels were measured after mechanical ventilation, and the levels of NICD, Hes-1, and Hes-5 protein were measured. Furthermore, the Notch signaling pathway inhibitor (DAPT 100 mg/kg) was administered intraperitoneally before mechanical ventilation to assess the role of Notch signaling in VILI. RESULTS Compared with the control group, hypertonic mechanical ventilation induced VILI and increased the expression of inflammatory factors such as TNF-α, IL-6 and MIP-2 in the cell lavage fluid. The level of Hes-5 gene was significantly up-regulated in pulmonary macrophages with high tidal volume ventilation, and the protein levels of NICD and Hes-5 were up-regulated in pulmonary macrophages. The levels of TNF-α, IL-6, MIP-2 and other inflammatory cytokines in the alveolar lavage fluid decreased, and the levels of NICD and Hes-5 were also decreased, after DAPT pretreatment compared with the high tidal volume. The upregulation of p-IκBα and the degradation of IκBα protein in alveolar macrophages after mechanical ventilation was also observed. CONCLUSION The expression of Notch signaling pathway protein in alveolar macrophages is upregulated after high tidal volume mechanical ventilation, and is involved in the regulation of mechanical ventilation and lung injury. Our results suggested that NF-κB pathway is likely involved in Notch-mediated mechanisms that underlie mechanical ventilation induced lung injury.
Collapse
Affiliation(s)
- Boxiang Du
- Department of Anesthesiology, The Second Affiliated Hospital of Nantong UniversityNantong, Jiangsu, China
| | - Min Gao
- Department of Anesthesiology, The First Affiliated Hospital of Medical School of Zhejiang UniversityZhejiang, China
| | - Liang Cao
- Department of ICU, The Second Affiliated Hospital of Nantong UniversityNantong, Jiangsu, China
| | - Jie Song
- Department of Anesthesiology, The Second Affiliated Hospital of Nantong UniversityNantong, Jiangsu, China
| | - Xueyin Shi
- Department of Anesthesiology and SICU, Xinhua Hospital, Shanghai Jiao Tong UniversityShanghai, China
| |
Collapse
|
23
|
Li Q, Ge YL, Li M, Fang XZ, Yuan YP, Liang L, Huang SQ. miR-127 contributes to ventilator-induced lung injury. Mol Med Rep 2017; 16:4119-4126. [PMID: 28765901 DOI: 10.3892/mmr.2017.7109] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 04/27/2017] [Indexed: 11/06/2022] Open
Abstract
Although it is essential in critical care medicine, mechanical ventilation often results in ventilator‑induced lung injury (VILI). Treating mice with lipopolysaccharide has been reported to upregulate the expression of miR‑127, which has been implicated in the modulation of immune responses. However, the putative roles of miR‑127 during the development of VILI have yet to be elucidated. The present study demonstrated that challenging mice with mechanical ventilation for 6 h significantly upregulated the expression of miR‑127 in bronchoalveolar lavage fluid, serum and lung tissue samples. Conversely, following the downregulation of miR‑127 expression in vivo using an adenovirus delivery system, VILI‑associated pathologies, including alterations in the pulmonary wet/dry ratio, pulmonary permeability, lung neutrophil infiltration and levels of pro‑inflammatory cytokines, were significantly attenuated. In addition, miR‑127 knockdown inhibited the ventilation‑induced activation of nuclear factor (NF)‑κB and p38 mitogen‑activated protein kinase (MAPK). These findings suggested that the upregulation of miR‑127 expression may contribute to the development of VILI, through the modulation of pulmonary permeability, the induction of histopathological alterations, and the potentiation of inflammatory responses involving NF‑κB and p38 MAPK‑associated signaling pathways.
Collapse
Affiliation(s)
- Qian Li
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, P.R. China
| | - Ya-Li Ge
- Department of Anesthesiology, Subei People's Hospital of Jiangsu, Yangzhou, Jiangsu 225001, P.R. China
| | - Min Li
- Department of Anesthesiology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Xiang-Zhi Fang
- Department of Anesthesiology, Subei People's Hospital of Jiangsu, Yangzhou, Jiangsu 225001, P.R. China
| | - Yan-Ping Yuan
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, P.R. China
| | - Lei Liang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Shao-Qiang Huang
- Department of Anesthesiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, P.R. China
| |
Collapse
|
24
|
Wu NC, Liao FT, Cheng HM, Sung SH, Yang YC, Wang JJ. Intravenous superoxide dismutase as a protective agent to prevent impairment of lung function induced by high tidal volume ventilation. BMC Pulm Med 2017; 17:105. [PMID: 28747201 PMCID: PMC5530466 DOI: 10.1186/s12890-017-0448-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 07/19/2017] [Indexed: 12/12/2022] Open
Abstract
Background Positive-pressure mechanical ventilation is essential in assisting patients with respiratory failure in the intensive care unit and facilitating oxygenation in the operating room. However, it was also recognized as a primary factor leading to hospital-acquired pulmonary dysfunction, in which pulmonary oxidative stress and lung inflammation had been known to play important roles. Cu/Zn superoxide dismutase (SOD) is an important antioxidant, and possesses anti-inflammatory capacity. In this study, we aimed to study the efficacy of Cu/Zn SOD, administered intravenously during high tidal volume (HTV) ventilation, to prevent impairment of lung function. Methods Thirty-eight male Sprague-Dawley rats were divided into 3 groups: 5 h ventilation with (A) low tidal volume (LTV; 8 mL/kg; n = 10), (B) high tidal volume (HTV; 18 mL/kg; n = 14), or (C) HTV and intravenous treatment of Cu/Zn SOD at a dose of 1000 U/kg/h (HTV + SOD; n = 14). Lung function was evaluated both at baseline and after 5-h ventilation. Lung injury was assessed by histological examination, lung water and protein contents in the bronchoalveolar lavage fluid (BALF). Pulmonary oxidative stress was examined by concentrations of methylguanidine (MG) and malondialdehyde (MDA) in BALF, and antioxidative activity by protein expression of glutathione peroxidase-1 (GPx-1) in the lung. Severity of lung inflammation was evaluated by white blood cell and differential count in BALF, and protein expression of inducible nitric oxide synthase (iNOS), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-α (TNF-α), matrix metalloproteinase-9 (MMP-9), and mRNA expression of nuclear factor-κB (NF-κB) in the lung. We also examined protein expression of surfactant protein (SP)-A and D and we measured hourly changes in serum nitric oxide (NO) level. Results Five hours of LTV ventilation did not induce a major change in lung function, whereas 5 h of HTV ventilation induced apparent combined restrictive and obstructive lung disorder, together with increased pulmonary oxidative stress, decreased anti-oxidative activity and increased lung inflammation (P < 0.05). HTV ventilation also decreased SP-A and SP-D expression and suppressed serum NO level during the time course of ventilation. Cu/Zn SOD administered intravenously during HTV ventilation effectively reversed associated pulmonary oxidative stress and lung inflammation (P < 0.05); moreover, it preserved SP-A and SP-D expressions in the lung and increased serum nitric oxide (NO) level, enhancing vascular NO bioavailability. Conclusions HTV ventilation can induce combined restrictive and obstructive lung disorders. Intravenous administration of Cu/Zn SOD during HTV ventilation can prevent lung function impairment and lung injury via reducing pulmonary oxidative stress and lung inflammation, preserving pulmonary surfactant expression, and enhancing vascular NO bioavailability.
Collapse
Affiliation(s)
- Nan-Chun Wu
- Division of Cardiovascular Surgery, Department of Surgery, Chi-Mei Foundation Hospital, 901, Chung Hwa Rd. Yung Kang, Tainan, Taiwan
| | - Fan-Ting Liao
- School of Medicine, Fu Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 24205, Taiwan
| | - Hao-Min Cheng
- Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Public Health and Community Medicine Research Center, National Yang-Ming University, Taipei, Taiwan.,Department of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Hsien Sung
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Public Health and Community Medicine Research Center, National Yang-Ming University, Taipei, Taiwan.,Department of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Chun Yang
- School of Medicine, Fu Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 24205, Taiwan
| | - Jiun-Jr Wang
- School of Medicine, Fu Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang Dist, New Taipei City, 24205, Taiwan.
| |
Collapse
|
25
|
Lung Functional and Biologic Responses to Variable Ventilation in Experimental Pulmonary and Extrapulmonary Acute Respiratory Distress Syndrome. Crit Care Med 2017; 44:e553-62. [PMID: 26963321 DOI: 10.1097/ccm.0000000000001611] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVES The biologic effects of variable ventilation may depend on the etiology of acute respiratory distress syndrome. We compared variable and conventional ventilation in experimental pulmonary and extrapulmonary acute respiratory distress syndrome. DESIGN Prospective, randomized, controlled experimental study. SETTINGS University research laboratory. SUBJECTS Twenty-four Wistar rats. INTERVENTIONS Acute respiratory distress syndrome was induced by Escherichia coli lipopolysaccharide administered intratracheally (pulmonary acute respiratory distress syndrome, n = 12) or intraperitoneally (extrapulmonary acute respiratory distress syndrome, n = 12). After 24 hours, animals were randomly assigned to receive conventional (volume-controlled ventilation, n = 6) or variable ventilation (n = 6). Nonventilated animals (n = 4 per etiology) were used for comparison of diffuse alveolar damage, E-cadherin, and molecular biology variables. Variable ventilation was applied on a breath-to-breath basis as a sequence of randomly generated tidal volume values (n = 600; mean tidal volume = 6 mL/kg), with a 30% coefficient of variation (normal distribution). After randomization, animals were ventilated for 1 hour and lungs were removed for histology and molecular biology analysis. MEASUREMENTS AND MAIN RESULTS Variable ventilation improved oxygenation and reduced lung elastance compared with volume-controlled ventilation in both acute respiratory distress syndrome etiologies. In pulmonary acute respiratory distress syndrome, but not in extrapulmonary acute respiratory distress syndrome, variable ventilation 1) decreased total diffuse alveolar damage (median [interquartile range]: volume-controlled ventilation, 12 [11-17] vs variable ventilation, 9 [8-10]; p < 0.01), interleukin-6 expression (volume-controlled ventilation, 21.5 [18.3-23.3] vs variable ventilation, 5.6 [4.6-12.1]; p < 0.001), and angiopoietin-2/angiopoietin-1 ratio (volume-controlled ventilation, 2.0 [1.3-2.1] vs variable ventilation, 0.7 [0.6-1.4]; p < 0.05) and increased relative angiopoietin-1 expression (volume-controlled ventilation, 0.3 [0.2-0.5] vs variable ventilation, 0.8 [0.5-1.3]; p < 0.01). In extrapulmonary acute respiratory distress syndrome, only volume-controlled ventilation increased vascular cell adhesion molecule-1 messenger RNA expression (volume-controlled ventilation, 7.7 [5.7-18.6] vs nonventilated, 0.9 [0.7-1.3]; p < 0.05). E-cadherin expression in lung tissue was reduced in volume-controlled ventilation compared with nonventilated regardless of acute respiratory distress syndrome etiology. In pulmonary acute respiratory distress syndrome, E-cadherin expression was similar in volume-controlled ventilation and variable ventilation; in extrapulmonary acute respiratory distress syndrome, however, it was higher in variable ventilation than in volume-controlled ventilation. CONCLUSIONS Variable ventilation improved lung function in both pulmonary acute respiratory distress syndrome and extrapulmonary acute respiratory distress syndrome. Variable ventilation led to more pronounced beneficial effects in biologic marker expressions in pulmonary acute respiratory distress syndrome compared with extrapulmonary acute respiratory distress syndrome but preserved E-cadherin in lung tissue only in extrapulmonary acute respiratory distress syndrome, thus suggesting lower damage to epithelial cells.
Collapse
|
26
|
Huang C, Pan L, Lin F, Dai H, Fu R. Monoclonal antibody against Toll-like receptor 4 attenuates ventilator-induced lung injury in rats by inhibiting MyD88- and NF-κB-dependent signaling. Int J Mol Med 2017; 39:693-700. [PMID: 28204830 DOI: 10.3892/ijmm.2017.2873] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/04/2017] [Indexed: 11/06/2022] Open
Abstract
The mechanisms through which mechanical ventilation causes non-infectious inflammatory diseases and lung injury are poorly understood. Animals models of this type of injury suggest that it involves signaling mediated by Toll‑like receptor (TLR)4 and 9. In this study, in order to gain further insight into the involvement of TLR4 in this type of injury, we performed in vivo and in vitro experiments to determine the mechanisms through which TLR4 triggers inflammation. We also examined whether the use of TLR4 monoclonal antibody (mAb) can alleviate this type of injury. For this purpose, rats were tracheotomized and administered intratracheal injections of anti‑TLR4 mAb or saline, and then ventilated for 4 h at a high tidal volume (HTV) of 40 ml/ kg or allowed to breathe spontaneously for the same period of time (controls). Alveolar macrophages (AMs) were isolated from the bronchoalveolar lavage fluid (BALF) of the rats and stimulated for 16 h with tumor necrosis factor (TNF)‑α in the presence or absence of anti‑TLR4 mAb. Lung injury was assessed by examining lung histopathology, lung wet/dry weight ratio, BALF total protein and cytokine levels in BALF and plasma. The mRNA and protein expression levels of TLR4, TLR9, myeloid differentiation factor 88 (Myd88) and nuclear factor (NF)‑κB were measured in cultured macrophages. Compared to the controls (spontaneous breathing), the ventilated rats exhibited greater pulmonary permeability, more severe inflammatory cell infiltration/lung edema, and higher levels of interleukin (IL)‑1β, IL‑6 and TNF‑α in BALF and plasma. The AMs from the ventilated rats expressed higher mRNA and protein levels of TLR4, TLR9, Myd88 and NF‑κB compared with the macrophages from the spontaneously breathing rats. The ventilated rats pre‑treated with anti‑TLR4 mAb exhibited markedly attenuated signs of ventilation‑induced injury, such as less lung inflammation and pulmonary edema, fewer cells in BALF, and lower levels of ILs and TNF‑α in BALF and plasma. Similarly, the TNF‑α‑dependent increases in the mRNA and protein expression of TLR4, Myd88 and NF‑κB in AMs were attenuated when TNF‑α was co‑administered with anti‑TLR4 mAb than when TNF-α was administered alone. Co‑administering anti-TLR4 mAb also reduced the TNF‑α‑dependent secretion of ILs. On the whole, our data demonstrate that TLR4 contributes significantly to ventilation‑induced lung injury by activating the Myd88/NF‑κB pathway, and pre‑treating rats with anti‑TLR4 mAb partially protects them against this type of injury by inhibiting Myd88/NF-κB signaling.
Collapse
Affiliation(s)
- Cuiyuan Huang
- Department of Anesthesiology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Linghui Pan
- Department of Anesthesiology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Fei Lin
- Department of Anesthesiology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Huijun Dai
- Department of Anesthesiology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Ruili Fu
- Department of Anesthesiology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
27
|
Yang CH, Hsiao JL, Wu MF, Lu MH, Chang HM, Ko WS, Chiou YL. The declined levels of inflammatory cytokines related with weaning rate during period of septic patients using ventilators. CLINICAL RESPIRATORY JOURNAL 2016; 12:772-778. [PMID: 27925446 DOI: 10.1111/crj.12593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 11/04/2016] [Accepted: 11/11/2016] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Approximately 50% of patients with sepsis-induced acute lung injury and acute respiratory distress syndrome require mechanical ventilation. Patients with extended mechanical ventilator use routinely develop reinfections, which increases hospital stay, mortality, and health care cost. Some studies have pointed out inflammatory factors concentrations can affect ventilator weaning, but do not indicate changed inflammatory factors related to ventilator weaning during using ventilators. OBJECTIVES This study aimed to investigate during period of septic patients using ventilators, the inflammatory cytokines concentrations related with weaning rate. METHODS Blood was collected from 35 septic patients before and during ventilator use on days 1, 7, 14, and 21 (or weaning). RESULTS 58.3% (N = 20) of septic patients with mechanical ventilators were weaned successfully within 21 days (ventilator weaned group, VW), 16.7% (N = 6) did not wean within 21 days (ventilator dependent group, VD), and 25% died (death group) in hospital. Before ventilator use, higher C-reactive protein (CRP), IL-6, and IL-8 levels were measured in the death group than in all other groups (P < .05). During ventilator use, CRP, IL-6, and IL-8 concentrations declined significantly in VW and VD patients (P < .05). In addition, IL-6 concentrations in the VW group were significantly lower than in the VD group at 14 and 21 days (P < .05). CONCLUSION The factors of ventilators weaning successfully such as disease control, nutritional status, and so on. The declined levels of serum inflammatory cytokines, especially IL-6, improved inflammation status might be one factor of successfully weaning during septic patients on ventilators.
Collapse
Affiliation(s)
- Chao-Huei Yang
- Department of Pulmonary Medicine, Kuang-Tien General Hospital, Taichung, Taiwan, Republic of China
| | - Jung-Lung Hsiao
- Department of Critical Care Medicine, Kuang-Tien General Hospital, Taichung, Taiwan, Republic of China
| | - Ming-Feng Wu
- Department of Internal Medicine, Kuang-Tien General Hospital, Taichung, Taiwan, Republic of China
| | - Mei-Hua Lu
- Department of Nursing, Kuang-Tien General Hospital, Taichung, Taiwan, Republic of China
| | - Hui-Ming Chang
- Department of Nursing, Kuang-Tien General Hospital, Taichung, Taiwan, Republic of China
| | - Wang-Sheng Ko
- Department of Internal Medicine, Kuang-Tien General Hospital, Taichung, Taiwan, Republic of China.,Department of Nutrition and Institute of BioMedical Nutrition, Hungkuang University, Taichung, Taiwan, Republic of China
| | - Ya-Ling Chiou
- Department of Nutrition and Institute of BioMedical Nutrition, Hungkuang University, Taichung, Taiwan, Republic of China
| |
Collapse
|
28
|
Variable ventilation improves pulmonary function and reduces lung damage without increasing bacterial translocation in a rat model of experimental pneumonia. Respir Res 2016; 17:158. [PMID: 27887604 PMCID: PMC5124241 DOI: 10.1186/s12931-016-0476-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/22/2016] [Indexed: 01/08/2023] Open
Abstract
Background Variable ventilation has been shown to improve pulmonary function and reduce lung damage in different models of acute respiratory distress syndrome. Nevertheless, variable ventilation has not been tested during pneumonia. Theoretically, periodic increases in tidal volume (VT) and airway pressures might worsen the impairment of alveolar barrier function usually seen in pneumonia and could increase bacterial translocation into the bloodstream. We investigated the impact of variable ventilation on lung function and histologic damage, as well as markers of lung inflammation, epithelial and endothelial cell damage, and alveolar stress, and bacterial translocation in experimental pneumonia. Methods Thirty-two Wistar rats were randomly assigned to receive intratracheal of Pseudomonas aeruginosa (PA) or saline (SAL) (n = 16/group). After 24-h, animals were anesthetized and ventilated for 2 h with either conventional volume-controlled (VCV) or variable volume-controlled ventilation (VV), with mean VT = 6 mL/kg, PEEP = 5cmH2O, and FiO2 = 0.4. During VV, tidal volume varied randomly with a coefficient of variation of 30% and a Gaussian distribution. Additional animals assigned to receive either PA or SAL (n = 8/group) were not ventilated (NV) to serve as controls. Results In both SAL and PA, VV improved oxygenation and lung elastance compared to VCV. In SAL, VV decreased interleukin (IL)-6 expression compared to VCV (median [interquartile range]: 1.3 [0.3–2.3] vs. 5.3 [3.6–7.0]; p = 0.02) and increased surfactant protein-D expression compared to NV (2.5 [1.9–3.5] vs. 1.2 [0.8–1.2]; p = 0.0005). In PA, compared to VCV, VV reduced perivascular edema (2.5 [2.0–3.75] vs. 6.0 [4.5–6.0]; p < 0.0001), septum neutrophils (2.0 [1.0–4.0] vs. 5.0 [3.3–6.0]; p = 0.0008), necrotizing vasculitis (3.0 [2.0–5.5] vs. 6.0 [6.0–6.0]; p = 0.0003), and ultrastructural lung damage scores (16 [14–17] vs. 24 [14–27], p < 0.0001). Blood colony-forming-unit (CFU) counts were comparable (7 [0–28] vs. 6 [0–26], p = 0.77). Compared to NV, VCV, but not VV, increased expression amphiregulin, IL-6, and cytokine-induced neutrophil chemoattractant (CINC)-1 (2.1 [1.6–2.5] vs. 0.9 [0.7–1.2], p = 0.025; 12.3 [7.9–22.0] vs. 0.8 [0.6–1.9], p = 0.006; and 4.4 [2.9–5.6] vs. 0.9 [0.8–1.4], p = 0.003, respectively). Angiopoietin-2 expression was lower in VV compared to NV animals (0.5 [0.3–0.8] vs. 1.3 [1.0–1.5], p = 0.01). Conclusion In this rat model of pneumonia, VV improved pulmonary function and reduced lung damage as compared to VCV, without increasing bacterial translocation. Electronic supplementary material The online version of this article (doi:10.1186/s12931-016-0476-7) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Huang J, Wu B, Song J, Wu J. Mechanisms of hyperventilation-induced lung injuries in neonatal rats. Minerva Pediatr (Torino) 2016; 73:73-79. [PMID: 27270839 DOI: 10.23736/s2724-5276.16.04458-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The aim of this study was to investigate the mechanism of early inflammatory injury in neonatal ventilator-induced lung injuries (VILI). METHODS Newborn rats were randomly assigned to groups and administrated mechanical ventilation with different tidal volumes. Morphological changes in lung tissues were observed, and the levels of interleukin-6 (IL-6), cysteinyl leukotriene mRNA (CysLT1 mRNA), and nuclear factor-κB mRNA (NF-κBp65 mRNA) in lung tissues were analyzed. RESULTS The ventilation groups exhibited different degrees of inflammatory cell infiltration, which was aggravated as the tidal volume and ventilation time increased. The IL-6 levels of the hyperventilation 5H, conventional ventilation 5H, hyperventilation 3H, control, and normal lung-tissue group were 785.33±39.06, 701.6±33.65, 686.65±46.85, 637.63±40.55, and 635.02±65.78 pg/g, respectively. Hyperventilation increased the levels of IL-6 and NF-κBp65 mRNA as the ventilation time increased, and IL-6 was positively correlated with NF-κBp65 mRNA levels (r=0.72, P<0.01). Longer hyperventilation periods upregulate the level of CysLT1 mRNA. CysLT1 mRNA/GAPDH of the hyperventilation 5H group was 2.14±1.45 (P<0.01). CONCLUSIONS Mechanical ventilation with a large tidal volume can cause VILI, characterized at an early stage by inflammatory responses and particularly by the increased secretion and invasion of inflammatory cytokines and inflammatory cells. The activation of the NF-κB-IL-6 signaling pathway was an important mechanism for the initiation of VILI. Additionally, CysLT1 was involved in the inflammatory VILI damage, and its upregulation occurred later than that of IL-6.
Collapse
Affiliation(s)
- Jinjie Huang
- Department of Neonatology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Benqing Wu
- Department of Neonatology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen, China -
| | - Jinzhi Song
- Department of Neonatology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Jun Wu
- Department of Neonatology, Shenzhen People's Hospital, the Second Clinical Medical College of Jinan University, Shenzhen, China
| |
Collapse
|
30
|
Mishra V, DiAngelo SL, Silveyra P. Sex-specific IL-6-associated signaling activation in ozone-induced lung inflammation. Biol Sex Differ 2016; 7:16. [PMID: 26949510 PMCID: PMC4779258 DOI: 10.1186/s13293-016-0069-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 02/25/2016] [Indexed: 01/10/2023] Open
Abstract
Background Acute ozone (O3) exposure has known deleterious effects on the respiratory system and has been linked with respiratory disease and infection. Inflammatory lung disease induced by air pollution has demonstrated greater severity and poorer prognosis in women vs. men. Both severe damage to the bronchial-alveolar epithelium and malfunctioning of bronchial-blood barrier have been largely attributed to the pathobiology of O3-induced inflammatory response, but the associated mechanisms in the male and female lung remain unknown. Methods Here, we investigated sex-based differential regulation of lung interleukin-6 (IL-6) and its downstream signaling pathways JAK2/STAT3 and AKT1/NF-κB in response to O3 exposure in a mouse model. We exposed male and female mice (in different stages of the estrous cycle) to 2 ppm of O3 or filtered air (FA) for 3 h, and we harvested lung tissue for protein expression analysis by Western blot. Results We found significant up-regulation of IL-6 and IL-6R in females and IL-6 in males in response to O3 vs. FA. Ozone exposure induced a significant increase in STAT3-Y705 phosphorylation in both females and males. Males exposed to O3 had decreased levels of JAK2, but increased JAK2 (Y1007+Y1008) phosphorylation, while females exposed to O3 showed significant up-regulation of both proteins. Both NF-κB (p105/p50) and AKT1 protein levels were significantly increased only in females exposed to O3. In addition, females exposed to O3 during proestrus displayed increased expression of selected genes when compared to females exposed to O3 in other estrous cycle stages. Conclusions Together, our observations indicate a sex-based and estrous cycle-dependent differential lung inflammatory response to O3 and involvement of two converging JAK2/STAT3 and AKT1/NF-κB pathways. To our knowledge, this is the first study specifically addressing the impact of the estrous cycle in O3-associated lung inflammatory pathways. Electronic supplementary material The online version of this article (doi:10.1186/s13293-016-0069-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vikas Mishra
- Department of Pediatrics, The Pennsylvania State University College of Medicine, 500 University Drive, H085, Hershey, PA 17033 USA
| | - Susan L DiAngelo
- Department of Pediatrics, The Pennsylvania State University College of Medicine, 500 University Drive, H085, Hershey, PA 17033 USA
| | - Patricia Silveyra
- Department of Pediatrics, The Pennsylvania State University College of Medicine, 500 University Drive, H085, Hershey, PA 17033 USA ; Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| |
Collapse
|
31
|
Rizzo AN, Sammani S, Esquinca AE, Jacobson JR, Garcia JGN, Letsiou E, Dudek SM. Imatinib attenuates inflammation and vascular leak in a clinically relevant two-hit model of acute lung injury. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1294-304. [PMID: 26432864 DOI: 10.1152/ajplung.00031.2015] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 09/27/2015] [Indexed: 12/29/2022] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS), an illness characterized by life-threatening vascular leak, is a significant cause of morbidity and mortality in critically ill patients. Recent preclinical studies and clinical observations have suggested a potential role for the chemotherapeutic agent imatinib in restoring vascular integrity. Our prior work demonstrates differential effects of imatinib in mouse models of ALI, namely attenuation of LPS-induced lung injury but exacerbation of ventilator-induced lung injury (VILI). Because of the critical role of mechanical ventilation in the care of patients with ARDS, in the present study we pursued an assessment of the effectiveness of imatinib in a "two-hit" model of ALI caused by combined LPS and VILI. Imatinib significantly decreased bronchoalveolar lavage protein, total cells, neutrophils, and TNF-α levels in mice exposed to LPS plus VILI, indicating that it attenuates ALI in this clinically relevant model. In subsequent experiments focusing on its protective role in LPS-induced lung injury, imatinib attenuated ALI when given 4 h after LPS, suggesting potential therapeutic effectiveness when given after the onset of injury. Mechanistic studies in mouse lung tissue and human lung endothelial cells revealed that imatinib inhibits LPS-induced NF-κB expression and activation. Overall, these results further characterize the therapeutic potential of imatinib against inflammatory vascular leak.
Collapse
Affiliation(s)
- Alicia N Rizzo
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois; University of Illinois at Chicago, Department of Pharmacology, Chicago, Illinois
| | - Saad Sammani
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois
| | - Adilene E Esquinca
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois
| | - Jeffrey R Jacobson
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois
| | - Joe G N Garcia
- Arizona Health Sciences Center, University of Arizona, Tucson, Arizona
| | - Eleftheria Letsiou
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois
| | - Steven M Dudek
- University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois; University of Illinois at Chicago, Department of Pharmacology, Chicago, Illinois;
| |
Collapse
|
32
|
Biological Impact of Transpulmonary Driving Pressure in Experimental Acute Respiratory Distress Syndrome. Anesthesiology 2015; 123:423-33. [DOI: 10.1097/aln.0000000000000716] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Abstract
Background:
Ventilator-induced lung injury has been attributed to the interaction of several factors: tidal volume (VT), positive end-expiratory pressure (PEEP), transpulmonary driving pressure (difference between transpulmonary pressure at end-inspiration and end-expiration, ΔP,L), and respiratory system plateau pressure (Pplat,rs).
Methods:
Forty-eight Wistar rats received Escherichia coli lipopolysaccharide intratracheally. After 24 h, animals were randomized into combinations of VT and PEEP, yielding three different ΔP,L levels: ΔP,LLOW (VT = 6 ml/kg, PEEP = 3 cm H2O); ΔP,LMEAN (VT = 13 ml/kg, PEEP = 3 cm H2O or VT = 6 ml/kg, PEEP = 9.5 cm H2O); and ΔP,LHIGH (VT = 22 ml/kg, PEEP = 3 cm H2O or VT = 6 ml/kg, PEEP = 11 cm H2O). In other groups, at low VT, PEEP was adjusted to obtain a Pplat,rs similar to that achieved with ΔP,LMEAN and ΔP,LHIGH at high VT.
Results:
At ΔP,LLOW, expressions of interleukin (IL)-6, receptor for advanced glycation end products (RAGE), and amphiregulin were reduced, despite morphometric evidence of alveolar collapse. At ΔP,LHIGH (VT = 6 ml/kg and PEEP = 11 cm H2O), lungs were fully open and IL-6 and RAGE were reduced compared with ΔP,LMEAN (27.4 ± 12.9 vs. 41.6 ± 14.1 and 0.6 ± 0.2 vs. 1.4 ± 0.3, respectively), despite increased hyperinflation and amphiregulin expression. At ΔP,LMEAN (VT = 6 ml/kg and PEEP = 9.5 cm H2O), when PEEP was not high enough to keep lungs open, IL-6, RAGE, and amphiregulin expression increased compared with ΔP,LLOW (41.6 ± 14.1 vs. 9.0 ± 9.8, 1.4 ± 0.3 vs. 0.6 ± 0.2, and 6.7 ± 0.8 vs. 2.2 ± 1.0, respectively). At Pplat,rs similar to that achieved with ΔP,LMEAN and ΔP,LHIGH, higher VT and lower PEEP reduced IL-6 and RAGE expression.
Conclusion:
In the acute respiratory distress syndrome model used in this experiment, two strategies minimized ventilator-induced lung injury: (1) low VT and PEEP, yielding low ΔP,L and Pplat,rs; and (2) low VT associated with a PEEP level sufficient to keep the lungs open.
Collapse
|
33
|
Gu C, Liu M, Zhao T, Wang D, Wang Y. Protective role of p120-catenin in maintaining the integrity of adherens and tight junctions in ventilator-induced lung injury. Respir Res 2015; 16:58. [PMID: 25986488 PMCID: PMC4489357 DOI: 10.1186/s12931-015-0217-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 04/22/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Ventilator-induced lung injury (VILI) is one of the most common complications for patients with acute lung injury (ALI) or acute respiratory distress syndrome (ARDS). Although p120 is an important protein in the regulation of cell junctions, further mechanisms should be explored for prevention and treatment of VILI. METHODS Mouse lung epithelial cells (MLE-12), which were transfected with p120 small interfering (si)RNA, p120 cDNA, wild-type E-cadherin juxtamembrane domain or a K83R mutant juxtamembrane domain (K83R-JMD), were subjected to 20% cyclic stretches for 2 or 4 h. Furthermore, MLE-12 cells and mice, which were pretreated with the c-Src inhibitor PP2 or RhoA inhibitor Y27632, underwent 20% cyclic stretches or mechanical stretching, respectively. Moreover, wild-type C57BL/6 mice were transfected with p120 siRNA-liposome complexes before mechanical ventilation. Cell lysates and lung tissues were then analyzed to detect lung injury. RESULTS cyclic stretches of 20% actived c-Src, which induced degradation of E-cadherin, p120 and occludin. However, loss of p120 increased the degradation and endocytosis of E-cadherin. Immunoprecipitation and Immunofluorescence results showed a decrease in the association between p120 and E-cadherin, while gap formation increased in p120 siRNA and K83R-JMD groups after 20% cyclic stretches. Loss of p120 also reduced the occludin level and decreased the association of occludin and ZO-1 by enhancing RhoA activity. However, the altered levels of occludin and E-cadherin were reversed by PP2 or Y27632 treatments compared with the cyclic stretch group. Consistently, the expression, redistribution and disassociation of junction proteins were all restored in the p120 overexpression group after 20% cyclic stretches. Moreover, the role of p120 in VILI was confirmed by increased wet/dry weigh ratio and enhanced production of cytokines (tumor necrosis factor-α and interleukin-six) in p120-depleted mice under mechanical ventilation. CONCLUSIONS p120 protected against VILI by regulating both adherens and tight junctions. p120 inhibited E-cadherin endocytosis by increasing the association between p120 and juxtamembrane domain of E-cadherin. Furthermore, p120 reduced the degradation of occludin by inhibiting RhoA activity. These findings illustrated further mechanisms of p120 in the prevention of VILI, especially for patients with ALI or ARDS.
Collapse
Affiliation(s)
- Changping Gu
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, 250014, Shandong Province, China.
| | - Mengjie Liu
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, 250014, Shandong Province, China.
| | - Tao Zhao
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, 250014, Shandong Province, China.
| | - Dong Wang
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, 250014, Shandong Province, China.
| | - Yuelan Wang
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, No. 16766 Jingshi Road, Jinan, 250014, Shandong Province, China.
| |
Collapse
|
34
|
Letsiou E, Rizzo AN, Sammani S, Naureckas P, Jacobson JR, Garcia JGN, Dudek SM. Differential and opposing effects of imatinib on LPS- and ventilator-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2014; 308:L259-69. [PMID: 25480336 DOI: 10.1152/ajplung.00323.2014] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Endothelial dysfunction underlies the pathophysiology of vascular disorders such as acute lung injury (ALI) syndromes. Recent work has identified the Abl family kinases (c-Abl and Arg) as important regulators of endothelial cell (EC) barrier function and suggests that their inhibition by currently available pharmaceutical agents such as imatinib may be EC protective. Here we describe novel and differential effects of imatinib in regulating lung pathophysiology in two clinically relevant experimental models of ALI. Imatinib attenuates endotoxin (LPS)-induced vascular leak and lung inflammation in mice but exacerbates these features in a mouse model of ventilator-induced lung injury (VILI). We next explored these discrepant observations in vitro through investigation of the roles for Abl kinases in cultured lung EC. Imatinib attenuates LPS-induced lung EC permeability, restores VE-cadherin junctions, and reduces inflammation by suppressing VCAM-1 expression and inflammatory cytokine (IL-8 and IL-6) secretion. Conversely, in EC exposed to pathological 18% cyclic stretch (CS) (in vitro model of VILI), imatinib decreases VE-cadherin expression, disrupts cell-cell junctions, and increases IL-8 levels. Downregulation of c-Abl expression with siRNA attenuates LPS-induced VCAM-1 expression, whereas specific reduction of Arg reduces VE-cadherin expression in 18% CS-challenged ECs to mimic the imatinib effects. In summary, imatinib exhibits pulmonary barrier-protective and anti-inflammatory effects in LPS-injured mice and lung EC; however, imatinib exacerbates VILI as well as dysfunction in 18% CS-EC. These findings identify the Abl family kinases as important modulators of EC function and potential therapeutic targets in lung injury syndromes.
Collapse
Affiliation(s)
- E Letsiou
- University of Illinois at Chicago, Division of Pulmonary, Critical Care, Sleep, and Allergy, Illinois; and
| | - A N Rizzo
- University of Illinois at Chicago, Division of Pulmonary, Critical Care, Sleep, and Allergy, Illinois; and
| | - S Sammani
- University of Illinois at Chicago, Division of Pulmonary, Critical Care, Sleep, and Allergy, Illinois; and
| | - P Naureckas
- University of Illinois at Chicago, Division of Pulmonary, Critical Care, Sleep, and Allergy, Illinois; and
| | - J R Jacobson
- University of Illinois at Chicago, Division of Pulmonary, Critical Care, Sleep, and Allergy, Illinois; and
| | - J G N Garcia
- Arizona Health Sciences Center, University of Arizona, Arizona
| | - S M Dudek
- University of Illinois at Chicago, Division of Pulmonary, Critical Care, Sleep, and Allergy, Illinois; and
| |
Collapse
|
35
|
Alvira CM. Nuclear factor-kappa-B signaling in lung development and disease: one pathway, numerous functions. ACTA ACUST UNITED AC 2014; 100:202-16. [PMID: 24639404 PMCID: PMC4158903 DOI: 10.1002/bdra.23233] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/28/2014] [Accepted: 01/29/2014] [Indexed: 01/04/2023]
Abstract
In contrast to other organs, the lung completes a significant portion of its development after term birth. During this stage of alveolarization, division of the alveolar ducts into alveolar sacs by secondary septation, and expansion of the pulmonary vasculature by means of angiogenesis markedly increase the gas exchange surface area of the lung. However, postnatal completion of growth renders the lung highly susceptible to environmental insults such as inflammation that disrupt this developmental program. This is particularly evident in the setting of preterm birth, where impairment of alveolarization causes bronchopulmonary dysplasia, a chronic lung disease associated with significant morbidity. The nuclear factor κ-B (NFκB) family of transcription factors are ubiquitously expressed, and function to regulate diverse cellular processes including proliferation, survival, and immunity. Extensive evidence suggests that activation of NFκB is important in the regulation of inflammation and in the control of angiogenesis. Therefore, NFκB-mediated downstream effects likely influence the lung response to injury and may also mediate normal alveolar development. This review summarizes the main biologic functions of NFκB, and highlights the regulatory mechanisms that allow for diversity and specificity in downstream gene activation. This is followed by a description of the pro and anti-inflammatory functions of NFκB in the lung, and of NFκB-mediated angiogenic effects. Finally, this review summarizes the clinical and experimental data that support a role for NFκB in mediating postnatal angiogenesis and alveolarization, and discusses the challenges that remain in developing therapies that can selectively block the detrimental functions of NFκB yet preserve the beneficial effects.
Collapse
Affiliation(s)
- Cristina M Alvira
- Division of Critical Care Medicine Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
36
|
Gao M, Liu D, Du Y, Sun R, Zhao L. Autophagy facilitates ventilator-induced lung injury partly through activation of NF-kappaB pathway. Med Sci Monit 2013; 19:1173-5. [PMID: 24343346 PMCID: PMC3871487 DOI: 10.12659/msm.889746] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Mechanical ventilation is an important supportive therapy in the intensive care unit (ICU) to assist the critically ill patients with respiratory failure. But longer ventilation time has been proven to contribute to the lung injury which has been recognized as ventilator-induced lung injury (VILI). Recently studies have suggested that NF-κB signaling pathways may play a critical role in the process of inflammation and autophagy, and autophagy can reduce the damage of VILI partly by activating the NF-κB pathways. Thus, we propose that autophagy may facilitate ventilator-induced lung injury partly through activation of NF-κB pathway, which might be a new potential therapeutic target for ventilator-induced lung injury. Although the exact mechanism of autophagy and its exact role in the VILI need to be further explored, at least it provides us a potential target in the future prevention of VILI.
Collapse
Affiliation(s)
- Min Gao
- Intensive Care Unit, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (mainland)
| | - Donglei Liu
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (mainland)
| | - Yuming Du
- Intensive Care Unit, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (mainland)
| | - Rongqing Sun
- Intensive Care Unit, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (mainland)
| | - Luosha Zhao
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China (mainland)
| |
Collapse
|