1
|
Park SJ, Suh JW, Haam S. Transient hyperthermia during ex vivo lung perfusion has no protective effect in rat model. Transpl Immunol 2023; 77:101800. [PMID: 36841512 DOI: 10.1016/j.trim.2023.101800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/30/2023] [Accepted: 02/21/2023] [Indexed: 02/27/2023]
Abstract
BACKGROUND Although ex vivo lung perfusion (EVLP) is a useful technique for evaluating and repairing donor lungs for transplantation, EVLP itself can lead to inflammation in the lung. Heat shock proteins (HSPs) have anti-inflammatory effects and can reduce ischemic reperfusion injury in the donor's lungs after transplantation. In this study, the effects of transient hyperthermia during EVLP on the expression of HSPs and inflammatory pathways were examined. METHODS Fifteen male Sprague-Dawley rats were randomly divided into three groups: sham (n = 5), normothermic EVLP (37 °C, n = 5), and transient hyperthermia during EVLP (42 °C, n = 5). Lung function analyses regarding PaO2/FiO2 ratio, compliance, and pulmonary vascular resistance were conducted. The expression levels of HSPs and inflammatory cytokines were also evaluated. The degree of lung injury was histopathologically evaluated. Transcriptome analysis was performed on lung tissues from the sham (n = 2), normothermic EVLP (n = 2), and heat stress-EVLP (n = 2) groups. RESULTS There were no significant differences in functional or histological parameters between the three groups. The expression of HSPs had significantly increased, especially that of HSPs 40 and 60 in the heat stress EVLP group; this was consistent with the inflammatory response. Inflammatory cytokine levels were significantly higher during EVLP and intensified with transient hyperthermia. CONCLUSION Transient hyperthermia during EVLP has no protective effect on the donor lung graft or activation of the inflammatory pathway at the gene level.
Collapse
Affiliation(s)
- Soo Jin Park
- Department of Thoracic and Cardiovascular Surgery, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jee Won Suh
- Department of Thoracic and Cardiovascular Surgery, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Gyeonggi-do, Republic of Korea.
| | - Seokjin Haam
- Department of Thoracic and Cardiovascular Surgery, Ajou University Hospital, Ajou University School of Medicine, 164 World cup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea.
| |
Collapse
|
2
|
Yazdanian M, Barzegar PEF, Ranjbar R, Tahmasebi E, Tebyaniyan H, Barzegar KEF, Hayati FZ, Farjanikish G. The comparative effects of erythromycin and amikacin on acute respiratory Pseudomonas aeruginosa infection. Vet Med Sci 2023; 9:867-875. [PMID: 36318447 PMCID: PMC10029875 DOI: 10.1002/vms3.991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND One of the most common causes of pneumonia is Pseudomonas aeruginosa (P. aeruginosa). As with other microbial pathogens, this bacterium tends to develop resistance to various antibiotics. Amikacin and erythromycin, which are from the aminoglycoside and macrolide antibiotic families, are used to treat respiratory infections caused by P. aeruginosa. OBJECTIVES This study explored whether amikacin, erythromycin or a combination of both works better against P. aeruginosa acute lung infection. METHODS For this study, 32 rats were used. The trachea of rats was exposed aseptically and their lung was infected with P. aeruginosa through trachea. Then, according to the group, they received amikacin, erythromycin or a combination of both for 1 week. Finally, they were euthanised on the 3rd and 7th days post-infection. The macroscopic and microscopic evaluations of the lungs, kidney and liver were performed. The right lung was collected for in vivo bacteriological analysis. RESULTS The amikacin group (A group) had a statistically significantly lower macroscopic and microscopic scores than the other groups (p < 0.05). In vivo bacteriological test revealed that the A group had significantly lower lung bacterial load (p < 0.05). CONCLUSIONS In summary, it was concluded that amikacin could help alleviate the respiratory infection caused by P. aeruginosa solely, and it was more effective than erythromycin.
Collapse
Affiliation(s)
- Mohsen Yazdanian
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Reza Ranjbar
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
- School of Dentistry, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elahe Tahmasebi
- Research Center for Prevention of Oral and Dental Diseases, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | | | - Farhad Zadeh Hayati
- Faculty of Veterinary Medicine, Department of Clinical Sciences, Lorestan University, Khorramabad, Iran
| | - Ghasem Farjanikish
- Department of Pathobiology, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| |
Collapse
|
3
|
Nayak SS, Sundararajan V. Robust anti-inflammatory activity of genistein against neutrophil elastase: a microsecond molecular dynamics simulation study. J Biomol Struct Dyn 2023; 41:11612-11628. [PMID: 36705087 DOI: 10.1080/07391102.2023.2170919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/26/2022] [Indexed: 01/28/2023]
Abstract
Human Neutrophil Elastase (HNE) is one of the major causes of tissue destruction in numerous chronic and inflammatory disorders and has been reported as a therapeutic target for inflammatory diseases. Overexpression of this enzyme plays a critical role in the pathogenesis of rheumatoid arthritis (RA). The focus of this study is to identify potent natural inhibitors that could target the active site of the HNE through the use of computational methods. The molecular structure of small molecules was retrieved from several natural compound databases. This was followed by structure-based virtual screening, molecular docking, ADMET property predictions and molecular dynamic simulation studies to screen potential HNE inhibitors. In total, 1881 natural compounds were extracted and subjected to molecular docking studies, and 10 compounds were found to have good interactions, exhibiting the best docking scores. Genistein showed higher binding efficacy (-10.28 Kcal/mol) to HNE in comparison to other natural compounds. The conformational stability of the docked complex of the ELANE gene (HNE) with genistein was assessed using 1-microsecond molecular dynamic simulation (MDs), which reliably revealed the unique stereochemical alteration of the complex, indicating its conformational stability and flexibility. Alterations in the enzyme structure upon complex formation were further characterized through clustering analysis and linear interaction energy (LIE) calculation. The outcomes of this research propose novel potential candidates against target HNE.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Smruti Sudha Nayak
- Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India
| | - Vino Sundararajan
- Department of Bio-Sciences, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamilnadu, India
| |
Collapse
|
4
|
Hong S, Jian C, Wang H, Wang X, Xing L, Qiao L. Effects of different doses of methylprednisolone therapy on acute respiratory distress syndrome: results from animal and clinical studies. BMC Pulm Med 2022; 22:348. [PMID: 36114531 PMCID: PMC9482269 DOI: 10.1186/s12890-022-02148-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/09/2022] [Indexed: 11/23/2022] Open
Abstract
Background The optimal dose of glucocorticoids for acute respiratory distress syndrome (ARDS) is uncertain. This study aimed to evaluate the effects of different doses of methylprednisolone on sepsis-induced acute lung injury (ALI) rats and a cohort of moderate and severe ARDS patients. Methods ALI rats, challenged with lipopolysaccharide, were randomly received intraperitoneal injection of normal saline (model group) and different doses of methylprednisolone (0.5, 2, 8 mg/kg, named as low-, moderate- and high-dose group, respectively) for 5 days. The body weight changes of rats, inflammatory factors in bronchoalveolar lavage fluid (BALF), lung wet/dry ratio, histopathological score, and the mRNA expressions of glucocorticoid receptor α (GRα), GRβ and nuclear factor-κB (NF-κB) were measured. Forty moderate and severe ARDS patients were treated with standard of care or plus different doses of methylprednisolone (40, 80, 120 mg/day, named as low-, moderate- and high-dose group, respectively) for 5 days. Clinical outcomes were PaO2/FiO2 ratio and C-reactive protein (CRP) level at day 5, intubation rate, hospital stay, 28-day mortality, and adverse events rate. Results In animal experiment, different doses of methylprednisolone could increase the body weight of rats, and reduce inflammatory factors in BALF and the degree of lung injury compared with model group. The efficacy of methylprednisolone at moderate-dose was better than that at low-dose, but was equivalent to that at high-dose, which was consistent with the differential changes in the mRNA expression of GRα, GRβ and NF-κB. In clinical study, the moderate-dose group was associated with higher PaO2/FiO2 ratio and lower CRP level. No significant difference in other clinical outcomes among groups was detected. Conclusions This study showed that the efficacy of methylprednisolone in ARDS treatment was not always dose-dependent due to the differential regulation of related receptors. The moderate-dose of methylprednisolone may be the potential optimal dose for ARDS treatment, which needs to be further verified by larger clinical trials.
Collapse
|
5
|
Overexpression of CXCL14 Alleviates Ventilator-Induced Lung Injury through the Downregulation of PKM2-Mediated Cytokine Production. Mediators Inflamm 2020; 2020:7650978. [PMID: 32774150 PMCID: PMC7396076 DOI: 10.1155/2020/7650978] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/06/2020] [Indexed: 12/30/2022] Open
Abstract
Ventilator-induced lung injury (VILI) is one of the most common complications of mechanical ventilation (MV), which strongly impacts the outcome of ventilated patients. Current evidences indicated that inflammation is a major contributor to the pathogenesis of VILI. Our results showed that MV induced excessive proinflammatory cytokine productions together with decreased CXCL14 and increased PKM2 expressions in injured lungs. In addition, CXCL14 overexpression downregulated PKM2 expression and attenuated VILI with reduced inflammation. Moreover, the overexpression of PKM2 markedly diminished the protective effects of CXCL14 against VILI as reflected by worsened morphology and increased cytokine production, whereas PKM2 knockdown decreased cytokine production and attenuated VILI. Collectively, these results suggested that CXCL14 overexpression attenuates VILI through the downregulation of PKM2-mediated proinflammatory cytokine production.
Collapse
|
6
|
Lin H, Chen M, Tian F, Tikkanen J, Ding L, Andrew Cheung HY, Nakajima D, Wang Z, Mariscal A, Hwang D, Cypel M, Keshavjee S, Liu M. α 1 -Anti-trypsin improves function of porcine donor lungs during ex-vivo lung perfusion. J Heart Lung Transplant 2018; 37:656-666. [DOI: 10.1016/j.healun.2017.09.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 09/15/2017] [Accepted: 09/26/2017] [Indexed: 11/28/2022] Open
|
7
|
MicroRNA-34a Suppresses Autophagy in Alveolar Type II Epithelial Cells in Acute Lung Injury by Inhibiting FoxO3 Expression. Inflammation 2018; 40:927-936. [PMID: 28321785 PMCID: PMC7101993 DOI: 10.1007/s10753-017-0537-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Excessive autophagic activity of alveolar type II epithelial (AT-II) cells is one of the main causes of acute lung injury (ALI); however, the underlying molecular mechanism remains to be determined. The microRNAs (miRNAs) are involved with autophagy in many diseases. The objective of this study was therefore to investigate the relationship between the miRNA expression and the autophagic activity of the AT-II cells in the pathogenesis of ALI and its molecular mechanism. A mouse model of ALI and AT-II cell injury was induced using lipopolysaccharide (LPS) in vivo and in vitro, and the expression of miR-34a and the autophagy-related proteins LC3 II/I and p62 were determined. Moreover, the autophagic activity was investigated after miR-34a overexpression and inhibition. The effects of miR-34a on its target gene, FoxO3, in regulating autophagic activity in AT-II cells were also determined. LPS induced autophagic activity and increased the expression of miR-34a in lung tissues and in AT-II cells. The in vitro results showed that the upregulation of miR-34a suppressed, whereas the inhibition of miR-34a promoted, autophagy in AT-II cells. Moreover, miR-34a could directly bind to the 3'-untranslated region of the autophagy-related gene, FoxO3, to decrease its expression. In addition, the knockdown of FoxO3 expression inhibited the autophagic activity in AT-II cells. Together, this study suggested that miR-34a might suppress the excessive autophagic activity in AT-II cells via targeting FoxO3 to reduce the damage of LPS-induced ALI.
Collapse
|
8
|
Tian YG, Zhang J. Protective effect of SIRT3 on acute lung injury by increasing manganese superoxide dismutase-mediated antioxidation. Mol Med Rep 2018; 17:5557-5565. [PMID: 29363727 DOI: 10.3892/mmr.2018.8469] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/19/2016] [Indexed: 11/06/2022] Open
Abstract
Prolonged exposure to hyperoxia results in acute lung injury (ALI). Pulmonary damage caused by oxygen toxicity occurs due to the generation of reactive oxygen species and subsequent formation of more potent oxidants. The present study demonstrated that sirtuin 3 (SIRT3) may attenuate hyperoxia‑induced ALI due to its potential antioxidative effect. In the present study, a hyperoxia‑induced acute lung injury mouse model, reverse transcription‑quantitative polymerase chain reaction, western blotting, retroviral mediated gene over‑expression and knockdown assays revealed that the expression of SIRT3 in the lung tissue of mice with hyperoxia‑induced ALI was decreased and overexpression of SIRT3 may significantly reduce hyperoxia‑induced ALI, as reflected by decreases in protein concentration, infiltrated neutrophils in bronchoalveolar lavage (BAL) fluid and wet/dry ratio of lung tissues. Furthermore, overexpression of SIRT3 increased the protein levels and enzymatic activity of manganese superoxide dismutase (MnSOD), and inhibited oxidative stress in the lungs of ALI mice. Additionally, the current study demonstrated that SIRT3 promoted the expression of MnSOD, and this regulation was crucial for the protective effect of SIRT3 on hyperoxia‑induced ALI. In summary, the results of the current study indicated that SIRT3 overexpression may effectively ameliorate hyperoxia‑induced ALI in mice, which indicates a potential application for SIRT3‑based gene therapy to treat clinical adult respiratory distress syndrome.
Collapse
Affiliation(s)
- Yong Gang Tian
- Department of Critical Care Medicine, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| | - Jian Zhang
- Department of Critical Care Medicine, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| |
Collapse
|
9
|
Sun LC, Zhang HB, Gu CD, Guo SD, Li G, Lian R, Yao Y, Zhang GQ. Protective effect of acacetin on sepsis-induced acute lung injury via its anti-inflammatory and antioxidative activity. Arch Pharm Res 2017; 41:1199-1210. [PMID: 29243040 PMCID: PMC7101724 DOI: 10.1007/s12272-017-0991-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 11/19/2017] [Indexed: 01/14/2023]
Abstract
Sepsis is a clinical syndrome with no effective protective or therapeutic treatments. Acacetin, a natural flavonoid compound, has anti-oxidative and anti-inflammatory effects which can potentially work to reduce sepsis. We investigated the potential protective effect of acacetin on sepsis-induced acute lung injury (ALI) ALI and dissect out the underlying mechanisms. Mice were divided into five groups: a sham group, a sepsis-induced ALI group, and three sepsis groups pre-treated with 20, 40, and 80 mg/kg body weight of acacetin. We found that acacetin significantly attenuated sepsis-induced ALI, in histological examinations and lung edema. Additionally, acacetin treatment decreased protein and inflammatory cytokine concentration and the number of infiltrated inflammatory cells in BALF compared with that in the non-treated sepsis mice. Pulmonary myeloperoxidase (MPO) activity was lower in the acacetin-pre-treated sepsis groups than in the sepsis group. The mechanism underlying the protective effect of acacetin on sepsis is related to the regulation of certain antioxidation genes, including inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), superoxide dismutases (SODs), and heme oxygenase 1 (HO-1).Taken together, our results indicate that acacetin pre-treatment inhibits sepsis-induced ALI through its anti-inflammatory and antioxidative activity, suggesting that acacetin may be a potential protective agent for sepsis-induced ALI.
Collapse
Affiliation(s)
- Li-Chao Sun
- Emergency Department of China-Japan Friendship Hospital, 2 Yinghua Dongjie, Chaoyang District, Hepingli, Beijing, 100029, China
| | - Hong-Bo Zhang
- Emergency Department of China-Japan Friendship Hospital, 2 Yinghua Dongjie, Chaoyang District, Hepingli, Beijing, 100029, China
| | - Cheng-Dong Gu
- Emergency Department of China-Japan Friendship Hospital, 2 Yinghua Dongjie, Chaoyang District, Hepingli, Beijing, 100029, China
| | - Shi-Dong Guo
- Emergency Department of China-Japan Friendship Hospital, 2 Yinghua Dongjie, Chaoyang District, Hepingli, Beijing, 100029, China
| | - Gang Li
- Emergency Department of China-Japan Friendship Hospital, 2 Yinghua Dongjie, Chaoyang District, Hepingli, Beijing, 100029, China
| | - Rui Lian
- Emergency Department of China-Japan Friendship Hospital, 2 Yinghua Dongjie, Chaoyang District, Hepingli, Beijing, 100029, China
| | - Yao Yao
- Emergency Department of China-Japan Friendship Hospital, 2 Yinghua Dongjie, Chaoyang District, Hepingli, Beijing, 100029, China
| | - Guo-Qiang Zhang
- Emergency Department of China-Japan Friendship Hospital, 2 Yinghua Dongjie, Chaoyang District, Hepingli, Beijing, 100029, China.
| |
Collapse
|
10
|
Ji Z, Liu H, Fang L, Yu Y, Zhou Z. Use of immunoproteomics to identify immunogenic proteins in a rat model of acute respiratory distress syndrome. Mol Med Rep 2017; 16:7625-7632. [PMID: 28944852 DOI: 10.3892/mmr.2017.7557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/17/2017] [Indexed: 11/06/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common and life‑threatening clinical syndrome, and seeking biomarkers of ARDS has been an area of continuing research. The present study hypothesized that alterations to certain immunogenic substances occur in injured lungs and are able to specifically bind with corresponding proteins in the blood, and that these proteins may be readily detected. To investigate this hypothesis, a rat model of ARDS was established by cecal ligation and puncture surgery, and an immunoproteomics approach, using serum as the primary antibody in a western blot analysis, was used with the aim of identifying immunogenic proteins in the injured lungs. Ingenuity Pathway Analysis (IPA) was used for bioinformatics analysis, and mass spectrometric analysis was used to identify a total of 38 differentially expressed immunogenic proteins. Bioinformatics analysis revealed that the top canonical pathways in which the identified proteins may be involved were gluconeogenesis I, glycolysis I, choline degradation I, NADH repair and heme degradation. IPA Biomarker Filter analysis with the terms 'acute respiratory distress syndrome/acute lung injury' was used to screen 13 proteins as candidate biomarkers. These proteins were described as antigens, and suggested that paired antibodies may be detected in the plasma of patients at high risk of ARDS. Analysis of these identified proteins may provide novel insights into the potential pathological mechanisms of ARDS.
Collapse
Affiliation(s)
- Zongshu Ji
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Haiyan Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Linsen Fang
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Youxin Yu
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Zheng Zhou
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
11
|
Apocynin suppressed the nuclear factor-κB pathway and attenuated lung injury in a rat hemorrhagic shock model. J Trauma Acute Care Surg 2017; 82:566-574. [DOI: 10.1097/ta.0000000000001337] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Ischemic post-conditioning attenuates acute lung injury induced by intestinal ischemia-reperfusion in mice: role of Nrf2. J Transl Med 2016; 96:1087-104. [PMID: 27501050 DOI: 10.1038/labinvest.2016.87] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/03/2016] [Accepted: 07/03/2016] [Indexed: 01/22/2023] Open
Abstract
Intestinal ischemic post-conditioning (IPo) protects against lung injury induced by intestinal ischemia-reperfusion (IIR) partly through promotion of expression and function of heme oxygenase-1 (HO-1). NF-E2-related factor-2 (Nrf2) is a key transcription factor that interacts with HO-1 and regulates antioxidant defense. However, the role of Nrf2 in IPo protection of IIR-induced pulmonary injury is not completely understood. Here we show that IPo significantly attenuated IIR-induced lung injury and suppressed oxidative stress and systemic inflammatory responses. IPo also increased the expression of both Nrf2 and HO-1. Consistently, the beneficial effects of IPo were abolished by ATRA and Brusatol, potent inhibitors of Nrf2. Moreover, the Nrf2 agonist t-BHQ showed similar activity as IPo. Taken together, our data suggest that Nrf2 activity, along with HO-1, plays an important role in the protective effects of IPo against IIR-induced acute lung injury.
Collapse
|
13
|
Guo N, Xu Y, Cao Z. Absinthin attenuates LPS-induced ALI through MIP-1α-mediated inflammatory cell infiltration. Exp Lung Res 2016; 41:514-24. [PMID: 26495959 DOI: 10.3109/01902148.2015.1093566] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Acute lung injury (ALI) is characterized by severe lung inflammation, and anti-inflammatory treatment is proposed to be a pertinent therapeutic strategy for the disease. Absinthin is a triterpene, extracted from a Chinese herb, with anti-inflammatory properties. The aim of this study was to evaluate whether absinthin can attenuate ALI in a mouse model of lung injury. Mice were treated with various concentrations (20 mg/kg, 40 mg/kg, and 80mg/kg) of absinthin, and lipopolysaccharide (LPS) to induce ALI. We found that the administration of absinthin relieved LPS-induced acute lung injury, as suggested by reduced histological scores, wet-to-dry ratio, myeloperoxidase activity, and accumulation of inflammatory cells in lung bronchoalveolar lavage fluid. Moreover, we demonstrated that absinthin significantly enhanced the expression of matrix metalloproteinase-8 (MMP-8); this effect could inhibit the accumulation of inflammatory cells in lung tissues through a mechanism dependent on MMP-8-mediated inactivation of macrophage inflammatory protein-1α. Therefore, we propose that absinthin is a promising novel therapeutic candidate for the treatment of ALI.
Collapse
Affiliation(s)
- Nailiang Guo
- a Department of Anesthesiology, Huadong Hospital Affiliated to Fudan University , Shanghai , China
| | - Yinghua Xu
- a Department of Anesthesiology, Huadong Hospital Affiliated to Fudan University , Shanghai , China
| | - Zhongqiang Cao
- a Department of Anesthesiology, Huadong Hospital Affiliated to Fudan University , Shanghai , China
| |
Collapse
|
14
|
Ulinastatin attenuates pulmonary endothelial glycocalyx damage and inhibits endothelial heparanase activity in LPS-induced ARDS. Biochem Biophys Res Commun 2016; 478:669-75. [PMID: 27498004 DOI: 10.1016/j.bbrc.2016.08.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 08/02/2016] [Indexed: 11/22/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a syndrome of acute respiratory failure characterized by major pathologic mechanisms of increased microvascular permeability and inflammation. The glycocalyx lines on the endothelial surface, which determines the vascular permeability, and heparanase play pivotal roles in the degradation of heparan sulfate (HS). HS is the major component of the glycocalyx. The aim of this study is to examine the effects of Ulinastatin (UTI) on vascular permeability and pulmonary endothelial glycocalyx dysfunction induced by lipopolysaccharide (LPS). In our study, C57BL/6 mice and human umbilical vein endothelial cells were stimulated with LPS to induce injury models. After 6 h of LPS stimulation, pulmonary pathological changes, pulmonary edema, and vascular permeability were notably attenuated by UTI. UTI inhibited LPS-induced endothelial glycocalyx destruction and significantly decreased the production of HS as determined by ELISA and immunofluorescence. UTI also reduced the active form of heparanase (50 kDa) expression and heparanase activity. Moreover, lysosome pH was investigated because heparanase (65 kDa) can be reduced easily in its active form at 50 kDa in a low pH environment within lysosome. Results showed that UTI could inhibit LPS-induced pH elevation in lysosome. In conclusion, UTI protects pulmonary endothelial glycocalyx integrity and inhibits heparanase activity during LPS-induced ARDS.
Collapse
|
15
|
Kong G, Huang X, Wang L, Li Y, Sun T, Han S, Zhu W, Ma M, Xu H, Li J, Zhang X, Liu X, Wang X. Astilbin alleviates LPS-induced ARDS by suppressing MAPK signaling pathway and protecting pulmonary endothelial glycocalyx. Int Immunopharmacol 2016; 36:51-58. [PMID: 27111514 DOI: 10.1016/j.intimp.2016.03.039] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/25/2016] [Accepted: 03/29/2016] [Indexed: 01/19/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a devastating disorder that is characterized by increased vascular endothelial permeability and inflammation. Unfortunately, no effective treatment beyond supportive care is available for ARDS. Astilbin, a flavonoid compound isolated from Rhizoma Smilacis Glabrae, has been used for anti-hepatic, anti-arthritic, and anti-renal injury treatments. This study examined the effects of Astilbin on pulmonary inflammatory activation and endothelial cell barrier dysfunction caused by Gram-negative bacterial endotoxin lipopolysaccharide (LPS). Endothelial cells from human umbilical veins or male Kunming mice were pretreated with Astilbin 24h before LPS stimulation. Results showed that Astilbin significantly attenuated the pulmonary histopathological changes and neutrophil infiltration 6h after the LPS challenge. Astilbin suppressed the activities of myeloperoxidase and malondialdehyde, as well as the expression of tumor necrosis factor-α and interleukin-6 in vivo and in vitro. As indices of pulmonary edema, lung wet-to-dry weight ratios, were markedly decreased by Astilbin pretreatment. Western blot analysis also showed that Astilbin inhibited LPS-induced activation of mitogen-activated protein kinase (MAPK) pathways in lung tissues. Furthermore, Astilbin significantly inhibited the activity of heparanase and reduced the production of heparan sulfate in the blood serum as determined by ELISA. These findings indicated that Astilbin can alleviate LPS-induced ARDS, which potentially contributed to the suppression of MAPK pathway activation and the degradation of endothelial glycocalyx.
Collapse
Affiliation(s)
- Guiqing Kong
- Department of Intensive Care Unit Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong Province, China; Department of Biotechnology, Binzhou Medical University, Yantai 264003, Shandong Province, China
| | - Xiao Huang
- Department of Intensive Care Unit Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong Province, China
| | - Lipeng Wang
- Department of Intensive Care Unit Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong Province, China
| | - Yan Li
- Department of Respiratory Medicine, Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong Province, China
| | - Ting Sun
- Department of Respiratory Medicine, Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong Province, China
| | - Shasha Han
- Department of Respiratory Medicine, Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong Province, China
| | - Weiwei Zhu
- Department of Intensive Care Unit Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong Province, China
| | - Mingming Ma
- Department of Intensive Care Unit, Hulunbeier People's Hospital, Hulunbeier 021000, Inner Mongolia Autonomous Region, China
| | - Haixiao Xu
- Department of Intensive Care Unit Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong Province, China
| | - Jiankui Li
- Department of Intensive Care Unit Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong Province, China
| | - Xiaohua Zhang
- Department of Biotechnology, Binzhou Medical University, Yantai 264003, Shandong Province, China.
| | - Xiangyong Liu
- Department of Cell Biology, Binzhou Medical University, Yantai 264003, Shandong Province, China.
| | - Xiaozhi Wang
- Department of Intensive Care Unit Affiliated Hospital of Binzhou Medical University, Binzhou 256603, Shandong Province, China.
| |
Collapse
|
16
|
Hsin MKY, Iskender I, Nakajima D, Chen M, Kim H, dos Santos PR, Sakamoto J, Lee J, Hashimoto K, Harmantas C, Hwang D, Waddell T, Liu M, Keshavjee S, Cypel M. Extension of donor lung preservation with hypothermic storage after normothermic ex vivo lung perfusion. J Heart Lung Transplant 2016; 35:130-136. [DOI: 10.1016/j.healun.2015.05.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 03/26/2015] [Accepted: 05/28/2015] [Indexed: 01/11/2023] Open
|
17
|
Ma H, Feng X, Ding S. Hesperetin attenuates ventilator-induced acute lung injury through inhibition of NF-κB-mediated inflammation. Eur J Pharmacol 2015; 769:333-41. [PMID: 26610718 DOI: 10.1016/j.ejphar.2015.11.038] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 01/01/2023]
Abstract
Hesperetin, a major bioflavonoid in sweet oranges and lemons, has been reported to have anti-inflammatory properties. However, the effect of hesperetin on ventilator-induced acute lung injury has not been studied. In present study, we investigated the protective effect of hesperetin on ventilator-induced acute lung injury in rats. Rats were orally administered hesperetin (10, 20, or 40mg/kg) two hour before acute lung injury was induced by mechanical ventilation. Rats were then randomly divided into six groups: the lung protective ventilation group (n=20, LV group), injurious ventilation group (n=20, HV group), vehicle-treated injurious ventilation group (n=20, LV+vehicle group), hesperetin (10mg/kg)-treated acute lung injury group (n=20, HV+Hsp (10mg)), hesperetin (20mg/kg)-treated acute lung injury group (n=20, HV+Hsp (20mg)), and hesperetin (40mg/kg)-treated acute lung injury group (n=20, HV+Hsp (40mg)). The lung tissues and bronchoalveolar lavage fluid were isolated for subsequent measurements. Treatment with hesperetin dramatically improved the histology of lung tissue, and reduced the wet/dry ratio, myeloperoxidase activity, protein concentration, and production of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β, and MIP-2 in the bronchoalveolar lavage fluid of rats with ventilator-induced acute lung injury. Additionally, our study indicated that this protective effect of hesperetin results from its ability to increase the expression of peroxisome proliferator-activated receptor (PPAR)-γ and inhibit the activation of the nuclear factor (NF)-κB pathway. These results suggest that hesperetin may be a potential novel therapeutic candidate for protection against ventilator-induced acute lung injury.
Collapse
Affiliation(s)
- Hongzhong Ma
- Department of Anesthesia, YanTai Yu Huang Ding Hospital, YanTai city, ShanDong Province 264000, China.
| | - Xiaoli Feng
- Department of Pathology, YanTai Yu Huang Ding Hospital, China
| | - Suchun Ding
- Department of Anesthesia, YanTai Yu Huang Ding Hospital, YanTai city, ShanDong Province 264000, China
| |
Collapse
|
18
|
Zhu WW, Kong GQ, Ma MM, Li Y, Huang X, Wang LP, Peng ZY, Zhang XH, Liu XY, Wang XZ. Short communication: Camel milk ameliorates inflammatory responses and oxidative stress and downregulates mitogen-activated protein kinase signaling pathways in lipopolysaccharide-induced acute respiratory distress syndrome in rats. J Dairy Sci 2015; 99:53-6. [PMID: 26601576 DOI: 10.3168/jds.2015-10005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/26/2015] [Indexed: 12/21/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a complex syndrome disorder with high mortality rate. Camel milk (CM) contains antiinflammatory and antioxidant properties and protects against numerous diseases. This study aimed to demonstrate the function of CM in lipopolysaccharide (LPS)-induced ARDS in rats. Camel milk reduced the lung wet:dry weight ratio and significantly reduced LPS-induced increases in neutrophil infiltration, interstitial and intra-alveolar edema, thickness of the alveolar wall, and lung injury scores of lung tissues. It also had antiinflammatory and antioxidant effects on LPS-induced ARDS. After LPS stimulation, the levels of proinflammatory cytokines (tumor necrosis factor-α, IL-10, and IL-1β) in serum and oxidative stress markers (malondialdehyde, myeloperoxidase, and total antioxidant capacity) in lung tissue were notably attenuated by CM. Camel milk also downregulated mitogen-activated protein kinase signaling pathways. Given these results, CM is a potential complementary food for ARDS treatment.
Collapse
Affiliation(s)
- Wei-Wei Zhu
- Department of Intensive Care Unit, Affiliated Hospital of Binzhou Medical University, Binzhou, 256603, Shandong Province, China
| | - Gui-Qing Kong
- Department of Intensive Care Unit, Affiliated Hospital of Binzhou Medical University, Binzhou, 256603, Shandong Province, China
| | - Ming-Ming Ma
- Department of Intensive Care Unit, Hulunbeier People's Hospital, Hulunbeier, 021000, Inner Mongolia Autonomous Region, China
| | - Yan Li
- Department of Respiratory Medicine, Affiliated Hospital of Binzhou Medical University, Binzhou, 256603, Shandong Province, China
| | - Xiao Huang
- Department of Intensive Care Unit, Affiliated Hospital of Binzhou Medical University, Binzhou, 256603, Shandong Province, China
| | - Li-Peng Wang
- Department of Intensive Care Unit, Affiliated Hospital of Binzhou Medical University, Binzhou, 256603, Shandong Province, China
| | - Zhen-Yi Peng
- Department of Ultrasound, Affiliated Hospital of Binzhou Medical University, Binzhou, 256603, Shandong Province, China
| | - Xiao-Hua Zhang
- Department of Biotechnology, Binzhou Medical University, Yantai, 264003, Shandong Province, China
| | - Xiang-Yong Liu
- Department of Cell Biology, Binzhou Medical University, Yantai, 264003, Shandong Province, China.
| | - Xiao-Zhi Wang
- Department of Intensive Care Unit, Affiliated Hospital of Binzhou Medical University, Binzhou, 256603, Shandong Province, China.
| |
Collapse
|
19
|
The therapeutic effect and mechanism of niacin on acute lung injury in a rat model of hemorrhagic shock. J Trauma Acute Care Surg 2015. [DOI: 10.1097/ta.0000000000000761] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
20
|
Role of Nrf2/ARE pathway in protective effect of electroacupuncture against endotoxic shock-induced acute lung injury in rabbits. PLoS One 2014; 9:e104924. [PMID: 25115759 PMCID: PMC4130631 DOI: 10.1371/journal.pone.0104924] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/15/2014] [Indexed: 12/03/2022] Open
Abstract
NF-E2 related factor 2 (Nrf2) is a major transcription factor and acts as a key regulator of antioxidant genes to exogenous stimulations. The aim of current study was to determine whether Nrf2/ARE pathway is involved in the protective effect of electroacupuncture on the injured lung in a rabbit model of endotoxic shock. A dose of lipopolysaccharide (LPS) 5 mg/kg was administered intravenously to replicate the model of acute lung injury induced by endotoxic shock. Electroacupuncture pretreatment was handled bilaterally at Zusanli and Feishu acupoints for five consecutive days while sham electroacupuncture punctured at non-acupoints. Fourty anesthetized New England male rabbits were randomized into normal control group (group C), LPS group (group L), electroacupuncture + LPS group (group EL) and sham electroacupuncture + LPS (group SEL). At 6 h after LPS administration, the animals were sacrificed and the blood samples were collected for biochemical measurements. The lungs were removed for calculation of wet-to-dry weight ratios (W/D), histopathologic examination, determination of heme oxygenase (HO)-1 protein and mRNA, Nrf2 total and nucleoprotein, as well as Nrf2 mRNA expression, and evaluation of the intracellular distribution of Nrf2 nucleoprotein. LPS caused extensive morphologic lung damage, which was lessened by electroacupuncture treatment. Besides, lung W/D ratios were significantly decreased, the level of malondialdehyde was inhibited, plasma levels of TNF-α and interleukin-6 were decreased, while the activities of superoxide dismutase, glutathione peroxidase and catalase were enhanced in the electroacupucnture treated animals. In addition, electroacupuncture stimulation distinctly increased the expressions of HO-1 and Nrf2 protein including Nrf2 total protein and nucleoprotein as well as mRNA in lung tissue, while these effects were blunted in the sham electroacupuncture group. We concluded that electroacupuncture treatment at ST36 and BL13 effectively attenuates lung injury in a rabbit model of endotoxic shock through activation of Nrf2/ARE pathway and following up-regulation of HO-1 expression.
Collapse
|
21
|
Proteomic analysis of lung tissue in a rat acute lung injury model: identification of PRDX1 as a promoter of inflammation. Mediators Inflamm 2014; 2014:469358. [PMID: 25024510 PMCID: PMC4082880 DOI: 10.1155/2014/469358] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/01/2014] [Accepted: 05/19/2014] [Indexed: 12/14/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) remains a high morbidity and mortality disease entity in critically ill patients, despite decades of numerous investigations into its pathogenesis. To obtain global protein expression changes in acute lung injury (ALI) lung tissues, we employed a high-throughput proteomics method to identify key components which may be involved in the pathogenesis of ALI. In the present study, we analyzed lung tissue proteomes of Pseudomonas aeruginosa-induced ALI rats and identified eighteen proteins whose expression levels changed more than twofold as compared to normal controls. In particular, we found that PRDX1 expression in culture medium was elevated by a lipopolysaccharide (LPS) challenge in airway epithelial cells in vitro. Furthermore, overexpression of PRDX1 increased the expression of proinflammatory cytokines interleukin-6 (IL-6), interleukin-8 (IL-8), and tumor necrosis factor-α (TNF-α), whereas knockdown of PRDX1 led to downregulated expression of cytokines induced by LPS. In conclusion, our findings provide a global alteration in the proteome of lung tissues in the ALI rat model and indicate that PRDX1 may play a critical role in the pathogenesis of ARDS by promoting inflammation and represent a novel strategy for the development of new therapies against ALI.
Collapse
|
22
|
Xu Z, Zhang C, Cheng L, Hu M, Tao H, Song L. The microRNA miR-17 regulates lung FoxA1 expression during lipopolysaccharide-induced acute lung injury. Biochem Biophys Res Commun 2014; 445:48-53. [PMID: 24486549 DOI: 10.1016/j.bbrc.2014.01.108] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 01/22/2014] [Indexed: 11/30/2022]
Abstract
Acute lung injury (ALI) is a severe pulmonary disease that causes a high number of fatalities worldwide. Studies have shown that FoxA1 expression is upregulated during ALI and may play an important role in ALI by promoting the apoptosis of alveolar type II epithelial cells. However, the mechanism of FoxA1 overexpression in ALI is unclear. In this study, an in vivo murine model of ALI and alveolar type II epithelial cells injury was induced using lipopolysaccharide (LPS). LPS upregulated FoxA1 in the lung tissue of the in vivo ALI model and in LPS-challenged type II epithelial cells. In contrast, miR-17 was significantly downregulated in these models. After miR-17 antagomir injection, the expression of FoxA1 was significantly increased in ALI mice. MiR-17 mimics could significantly inhibit FoxA1 mRNA and protein expression, whereas the miR-17 inhibitor could significantly increase FoxA1 mRNA and protein expression in LPS-induced type II epithelial cells. Thus, our results suggest that the downregulation of miR-17 expression could lead to FoxA1 overexpression in ALI.
Collapse
Affiliation(s)
- Zhaojun Xu
- Cardiothoracic Surgery of the First Affiliated Hospital, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 41007, China
| | - Caiping Zhang
- University of South China, College of Life Science, Department of Biochemistry and Molecular Biology, Hengyang, Hunan 421001, China
| | - Lijuan Cheng
- Department of Biochemistry and Molecular Biology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Mei Hu
- Department of Biochemistry and Molecular Biology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Huai Tao
- Department of Biochemistry and Molecular Biology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Lan Song
- Department of Biochemistry and Molecular Biology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; Division of Stem Cell Regulation and Application, State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan (Incubation), Hunan University of Chinese Medicine, Changsha, Hunan 410208, China; University of South China, College of Life Science, Department of Biochemistry and Molecular Biology, Hengyang, Hunan 421001, China.
| |
Collapse
|
23
|
Yu JB, Jianbo Y, Dong SA, Shuan D, Luo XQ, Xiaoqing L, Gong LR, Lirong G, Zhang Y, Yuan Z, Wang M, Man W, Cao XS, Xinshun C, Liu DQ, Daquan L. Role of HO-1 in protective effect of electro-acupuncture against endotoxin shock-induced acute lung injury in rabbits. Exp Biol Med (Maywood) 2013; 238:705-12. [PMID: 23918882 DOI: 10.1177/1535370213489487] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Heme oxygenase (HO)-1 has been reported to play a great role in attenuating lung injury during endotoxic shock in our previous research. Although electro-acupuncture has been explored to reduce oxidative stress and decrease inflammatory reaction in animals with endotoxic shock, the mechanism of this effect is still unclear. The aim of this study was to determine whether HO-1 is involved in the effect of electro-acupuncture on the injured lung during endotoxic shock in rabbits. Sixty New England white rabbits were randomly divided into groups C, Z, ES, EA, AP, and EAZ. Before inducing endotoxic shock, group ES received no electro-acupuncture, while group EA received electro-acupuncture at ST36 (zusanli) and BL13 (feishu) acupoints on both sides for five days and group AP received electro-acupuncture (EA) stimulation at a non-acupoint. Groups ES, AP, EA, and EAZ received LPS to replicate the experimental model of injured lung induced by endotoxic shock, and electro-acupuncture was performed throughout the procedure with the same parameter. Groups EAZ and Z received the HO-1 inhibitor, ZnPP-IX, intraperitoneally. The animals were sacrificed by blood-letting at 6 h after LPS administration. The blood samples were collected for serum examination, and the lungs were removed for pathology examination, detection of alveolaer epithelial cell apoptosis by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL assay), determination of wet to dry ratio, measurement of Evans blue (EB) contents, and determination of HO-1protein and mRNA expression. According to the results, EA at ST36 and BL13 could increase the expression of HO-1. At the same time, index of quantitative assessment (IQA) score and the number of TUNEL-positive cells decreased, while electro-acupuncture at the other points did not exert this effect, and pretreatment with ZnPP-IX in group EAZ suppressed the efficacy of electro-acupuncture preconditioning. In summary, electro-acupuncture stimulation at ST36 and BL13, while not the non-acupoint, could attenuate the lung injury during the endotoxic shock, and this effect was due to increased expression of HO-1.
Collapse
Affiliation(s)
| | - Yu Jianbo
- Department of Anesthesiology, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, 300100, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Feng L, Zhu W, Huang C, Li Y. Direct interaction of ONO-5046 with human neutrophil elastase through ¹H NMR and molecular docking. Int J Biol Macromol 2012; 51:196-200. [PMID: 22579959 DOI: 10.1016/j.ijbiomac.2012.04.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 03/26/2012] [Accepted: 04/23/2012] [Indexed: 11/29/2022]
Abstract
Human neutrophil elastase (HNE) has been implicated as a major contributor in the pathogenesis of diseases, such as pulmonary emphysema, acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and other inflammatory diseases. Therefore, searching for appropriate and potential human neutrophil elastase inhibitors (HNEI) that would restore the balance between the free enzyme and the endogenous inhibitors would be of therapeutic interest. ONO-5046 is the first specific HNEI to improve respiratory function and protect lung tissues against various lung injuries. However, the mechanism of ONO-5046 to HNE is still unclear. In this study, the binding properties of ONO-5046 were investigated through (1)H NMR, molecular docking, and bioassay methods to understand the effect of ONO-5046 to HNE. The proton spin-lattice relaxation rate and molecular rotational correlation time results indicated that ONO-5046 has higher affinity with HNE. The molecular docking study showed that ONO-5046 is perfectly matched for the primary enzyme specificity pocket (S1 pocket), and is tightly bound to this pocket of HNE through hydrophobic and hydrogen bonding interactions. The results of both methods were validated through analysis of the HNE inhibitory activity bioassay of ONO-5046 with an IC(50) value of 87.05 nM. Our data suggested that ONO-5046 could bind to HNE through direct interaction, and that molecular docking and NMR methods are valid approaches to survey new HNEI.
Collapse
Affiliation(s)
- Li Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | | | | | | |
Collapse
|
25
|
Mannitol attenuates acute lung injury induced by infrarenal aortic occlusion-reperfusion in rats. Surg Today 2011; 41:955-65. [DOI: 10.1007/s00595-010-4394-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 06/03/2010] [Indexed: 11/25/2022]
|
26
|
Wang M, Liu T, Wang D, Zheng Y, Wang X, He J. Therapeutic effects of pyrrolidine dithiocarbamate on acute lung injury in rabbits. J Transl Med 2011; 9:61. [PMID: 21569464 PMCID: PMC3112441 DOI: 10.1186/1479-5876-9-61] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Accepted: 05/13/2011] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) is an early characteristic of multiple organ dysfunction, responsible for high mortality and poor prognosis in patients. The present study aims to evaluate therapeutic effects and mechanisms of pyrrolidine dithiocarbamate (PDTC) on ALI. METHODS Alveolar-arterial oxygen difference, lung tissue edema and compromise, NF-κB activation in polymorphonuclear neutrophil (PMN), and systemic levels of tumor necrosis factor-alpha (TNFa) and intercellular adhesion molecule-1 (ICAM-1) in rabbits induced by the intravenous administration of lipopolysaccharide (LPS) and treated with PDTC. Production of TNFa and IL-8, activation of Cathepsin G, and PMNs adhesion were also measured. RESULTS The intravenous administration of PDTC had partial therapeutic effects on endotoxemia-induced lung tissue edema and damage, neutrophil influx to the lung, alveolar-capillary barrier dysfunction, and high systemic levels of TNFa and ICAM-1 as well as over-activation of NF-κB. PDTC could directly and partially inhibit LPS-induced TNFa hyper-production and over-activities of Cathepsin G. Such inhibitory effects of PDTC were related to the various stimuli and enhanced through combination with PI3K inhibitor. CONCLUSION NF-κB signal pathway could be one of targeting molecules and the combination with other signal pathway inhibitors may be an alternative of therapeutic strategies for ALI/ARDS.
Collapse
Affiliation(s)
- Meitang Wang
- Department of Emergency Medicine, The Second Military University Changhai Hospital, China
| | | | | | | | | | | |
Collapse
|
27
|
A Tetracycline Analog Improves Acute Respiratory Distress Syndrome Survival in an Ovine Model. Ann Thorac Surg 2010; 90:419-26. [DOI: 10.1016/j.athoracsur.2010.04.052] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Revised: 04/09/2010] [Accepted: 04/12/2010] [Indexed: 11/22/2022]
|
28
|
Ball MK, Hillman NH, Kallapur SG, Polglase GR, Jobe AH, Pillow JJ. Body temperature effects on lung injury in ventilated preterm lambs. Resuscitation 2010; 81:749-54. [PMID: 20299144 DOI: 10.1016/j.resuscitation.2009.12.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 11/30/2009] [Accepted: 12/03/2009] [Indexed: 10/19/2022]
Abstract
AIMS Mechanical ventilation causes lung injury in premature infants. Hypothermia may protect against and hyperthermia may augment lung injury. We tested the effects of hypo- and hyperthermia on ventilation induced acute lung injury in preterm lambs. METHODS Twin sheep fetuses at 128 d GA (term 150 d) were surgically delivered and randomized to unventilated control (UVC), normothermia (38-39 degrees C) without lung injury (NTNI), or to 1 of 3 injurious ventilation groups: hypothermic (33-34 degrees C, LT), normothermic (38-39 degrees C, NT) or hyperthermic (40-41 degrees C, HT). NT, LT and HT groups had 15 min of injurious ventilation (PEEP 0 cmH(2)O, V(T) escalation to 15 mL/kg) following delivery and prior to surfactant. The animals were then gently ventilated (PEEP 5cmH(2)O, V(T) 7.5 mL/kg) for 2h 45 min. NTNI lambs received surfactant at birth prior to gentle ventilation. The lambs were then euthanized, and bronchoalveolar lavage (BAL) fluid and lung tissue were used to evaluate lung injury, inflammatory cell counts, inflammatory markers and cytokine mRNA. RESULTS Target temperatures were achieved by 15 min of age and maintained for 3h. All ventilated groups had increased BAL protein, lung inflammation and increased cytokine mRNA. HT animals developed acidosis, premature death, pneumothoraces, impaired lung function and increased inflammatory mRNA expression. LT animals remained clinically stable without pneumothoraces or death, had improved ventilatory efficiency and trended toward lower inflammatory mRNA expression than NT animals. CONCLUSION Hyperthermia exacerbated ventilator induced lung injury, while hypothermia may protect against lung injury in the preterm lamb.
Collapse
Affiliation(s)
- Molly K Ball
- Division of Perinatal Medicine, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Lu X, Li Y, Simovic MO, Peckham R, Wang Y, Tsokos GC, Dalle Lucca JJ. Decay-accelerating factor attenuates C-reactive protein-potentiated tissue injury after mesenteric ischemia/reperfusion. J Surg Res 2009; 167:e103-15. [PMID: 20189588 DOI: 10.1016/j.jss.2009.10.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 09/14/2009] [Accepted: 10/14/2009] [Indexed: 11/16/2022]
Abstract
BACKGROUND C-reactive protein (CRP) is an acute pro-inflammatory mediator that has been demonstrated to enhance ischemia/reperfusion (IR) injury by virtue of activating the complement system. CRP is able to interact with complement proteins such as C1q, complement factor H, and C4b-binding protein. Since complement activation is central in the expression of tissue injury following IR, we have investigated the effects of human decay-accelerating factor (DAF), a complement inhibitor, on CRP-potentiated complement activation and tissue injury in mice subjected to mesenteric IR. MATERIALS AND METHODS Male C57B1/6 mice were allocated into eight groups: (1) Sham-operated group without IR injury; (2) CRP+Sham group; (3) IR group; (4) CRP+IR group; (5) DAF group; (6) CRP+DAF group; (7) IR+DAF group, and (8) CRP+IR+DAF group. Intestinal and lung injury, neutrophil infiltration, myeloperoxidase (MPO) expression, complement component deposition, and interleukin-6 (IL-6) production were assessed for each treatment group of mice. RESULTS We report that administration of DAF significantly attenuates the CRP-enhanced intestinal injury as well as remote lung damages following acute mesenteric IR in mice, while DAF inhibits complement activation, suppresses neutrophil infiltration, and reduces IL-6 production. CONCLUSIONS Our study suggests that inhibition complement activation with DAF may prove useful for the treatment of post-ischemic inflammatory injuries associated with an increased production of CRP.
Collapse
Affiliation(s)
- Xinyue Lu
- Division of Military Casualty Research, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Cypel M, Rubacha M, Yeung J, Hirayama S, Torbicki K, Madonik M, Fischer S, Hwang D, Pierre A, Waddell TK, de Perrot M, Liu M, Keshavjee S. Normothermic ex vivo perfusion prevents lung injury compared to extended cold preservation for transplantation. Am J Transplant 2009; 9:2262-9. [PMID: 19663886 DOI: 10.1111/j.1600-6143.2009.02775.x] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Treatment of injured donor lungs ex vivo to accelerate organ recovery and ameliorate reperfusion injury could have a major impact in lung transplantation. We have recently demonstrated a feasible technique for prolonged (12 h) normothermic ex vivo lung perfusion (EVLP). This study was performed to examine the impact of prolonged EVLP on ischemic injury. Pig donor lungs were cold preserved in Perfadex for 12 h and subsequently divided into two groups: cold static preservation (CSP) or EVLP at 37 degrees C with Steen solution for a further 12 h (total 24 h preservation). Lungs were then transplanted and reperfused for 4 h. EVLP preservation resulted in significantly better lung oxygenation (PaO(2) 531 +/- 43 vs. 244 +/- 49 mmHg, p < 0.01) and lower edema formation rates after transplantation. Alveolar epithelial cell tight junction integrity, evaluated by zona occludens-1 protein staining, was disrupted in the cell membranes after prolonged CSP but not after EVLP. The maintenance of integrity of barrier function during EVLP translates into significant attenuation of reperfusion injury and improved graft performance after transplantation. Integrity of functional metabolic pathways during normothermic perfusion was confirmed by effective gene transfer and GFP protein synthesis by lung alveolar cells. In conclusion, EVLP prevents ongoing injury associated with prolonged ischemia and accelerates lung recovery.
Collapse
Affiliation(s)
- M Cypel
- Toronto Lung Transplant Program, Division of Thoracic Surgery, Latner Thoracic Laboratories, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Kuromitsu S, Yokota H, Hiramoto M, Morita S, Mita H, Yamada T. Increased concentration of neutrophil elastase in urine from patients with interstitial cystitis. ACTA ACUST UNITED AC 2009; 42:455-61. [PMID: 18609268 DOI: 10.1080/00365590802025881] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To identify proteins associated with interstitial cystitis (IC), protein profiles were analyzed using a proteomics-based approach. The study tested whether neutrophil elastase in urine correlates with the symptomatic condition of IC. MATERIAL AND METHODS Proteins in urine from IC patients and healthy subjects were analyzed through a comparative proteomics approach using two-dimensional difference in-gel electrophoresis and nano-liquid chromatography-tandem mass spectrometry. Neutrophil elastase activity was measured by the digestion of peptide substrate. RESULTS The urinary neutrophil elastase concentration was significantly higher in IC patients with pain than in healthy subjects. It was significantly increased in patients with small bladder capacity (median 6.31 ng/ml in IC with a bladder capacity < 200 ml vs 1.15 ng/ml in IC with a bladder capacity > or = 200 ml and 0.18 ng/ml in healthy bladders, p < 0.01). The concentration of neutrophil elastase did not correlate with the neutrophil count in the urine of IC patients. CONCLUSION The concentration of neutrophil elastase increased in the urine of the IC patient subset with bladder pain and small bladder capacity.
Collapse
Affiliation(s)
- Sadao Kuromitsu
- Molecular Medicine Laboratories, Astellas Pharma Inc., Tsukuba, Japan.
| | | | | | | | | | | |
Collapse
|
33
|
Effects of Tempol, a Membrane-Permeable Radical Scavenger, on Local and Remote Organ Injuries Caused by Intestinal Ischemia/Reperfusion in Rats. J Surg Res 2008; 149:259-71. [DOI: 10.1016/j.jss.2007.12.791] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Revised: 11/12/2007] [Accepted: 12/19/2007] [Indexed: 11/22/2022]
|
34
|
Teke Z, Sacar M, Yenisey C, Atalay AO, Bicakci T, Erdem E. Activated protein C attenuates intestinal reperfusion–induced acute lung injury: an experimental study in a rat model. Am J Surg 2008; 195:861-73. [DOI: 10.1016/j.amjsurg.2007.06.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Revised: 06/01/2007] [Accepted: 06/01/2007] [Indexed: 10/22/2022]
|
35
|
He X, Han B, Mura M, Xia S, Wang S, Ma T, Liu M, Liu Z. Angiotensin-converting enzyme inhibitor captopril prevents oleic acid-induced severe acute lung injury in rats. Shock 2007; 28:106-11. [PMID: 17510605 DOI: 10.1097/shk.0b013e3180310f3a] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Lack of specific and efficient therapy leads to the high mortality rate of acute lung injury (ALI) and acute respiratory distress (ARDS). Recent evidence implies that angiotensin-converting enzyme (ACE) plays an important role in the pathogenesis of ALI. Pharmaceutical inhibitors of ACE have been used clinically for hypertension but not for ALI/ARDS yet. The objective was to study the effects of ACE inhibition with captopril on severe lung injury induced by oleic acid (OA) in rats. Oleic acid was intravenously injected into Sprague Dawley rats, followed by i.p. administration of captopril or saline control. Lung injury, endothelium damage and related molecules, and disturbance of coagulation were examined in comparison between the treated and the nontreated groups. An OA-induced ALI was featured with thickening of the alveolar septa, alveolar hemorrhage, and infiltration of inflammatory cells. Comparing with the nontreated OA group, the administration of captopril prevented the rats from OA-induced severe lungs injury, with a significantly lower lung injury score, less albumin content and infiltrated cells in the alveoli, decreased wet/dry weight ratio of the lung tissues, and improved lung function (PaO2 per fraction of inspired oxygen). Captopril also dramatically reduced the expression of intercellular adhesion molecule-1 in the lung tissue and in the circulating endothelial cells in the blood, indicating a protective effect on endothelial cells activation/damage. Moreover, captopril treatment led to a blockage of nuclear factor kappaB activation in lung tissues and to the recovery of the fibrinolytic disturbance. Thus, our data suggest that the inhibition of ACE with its clinically used inhibitor offers protective effects on ALI/ARDS, implying the potential for therapeutic option.
Collapse
Affiliation(s)
- Xiaolin He
- Department of Emergency and Institute of Respiratory Disease, First Affiliated Hospital, China Medical University, Shenyang, Liaoning, China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kabay B, Teke Z, Aytekin FO, Yenisey C, Bir F, Sacar M, Erdem E, Ozden A. Pyrrolidine Dithiocarbamate Reduces Lung Injury Caused by Mesenteric Ischemia/Reperfusion in a Rat Model. World J Surg 2007; 31:1707-15. [PMID: 17551782 DOI: 10.1007/s00268-007-9112-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Pyrrolidine dithiocarbamate (PDTC) is a low-molecular thiol antioxidant and potent inhibitor of nuclear factor-kappaB (NF-kappaB) activation. It has been shown to attenuate local harmful effects of ischemia/reperfusion (I/R) injury in many organs. In this study, we aimed to study the effect of PDTC on lung reperfusion injury induced by superior mesenteric occlusion. METHODS Male Wistar-albino rats randomized into three groups: (1) sham-operated control group (n = 12), laparotomy without I/R injury; (2) intestinal ischemia/reperfusion (I/R) group (n = 12), 60 min of ischemia by superior mesenteric occlusion followed by 2 h of reperfusion; and (3) I/R+PDTC-treated group (n = 12), 100 mg/kg injection of PDTC intravenously, 30 min after the commencement of reperfusion. Evans blue dye was injected to half of rats in all groups before the induction of I/R. We assessed the degree of pulmonary tissue injury biochemically by measuring malondialdehyde (MDA), glutathione (GSH), and nitric oxide (NO) levels, and histopathologically by establishing pulmonary neutrophil sequestration and acute lung injury scoring. Pulmonary edema was evaluated by Evans blue dye extravasation, as well as lung tissue wet/dry weight ratios. RESULTS Pyrrolidine dithiocarbamate treatment significantly reduced the MDA and NO levels, and increased the GSH levels in the lung parenchyma, biochemically (p < 0.05), and atteneuated the pulmonary parenchymal damage, histopathologically (p < 0.05). However, pulmonary neutrophil sequestration was not affected by postischemic treatment with PDTC (p > 0.05). Pyrrolidine dithiocarbamate administration also significantly alleviated the formation of pulmonary edema, as evidenced by the decreased Evans blue dye extravasation and organ wet/dry weight ratios (p < 0.05). CONCLUSIONS This study showed that postischemic treatment with PDTC significantly attenuated the lung reperfusion injury. Further clinical studies are needed for better understanding of the specific mechanisms of PDTC protection against I/R-related organ injury and to clarify whether PDTC may be a useful therapeutic agent during particular operations where remote organ I/R injury occurs.
Collapse
Affiliation(s)
- Burhan Kabay
- Department of General Surgery, Pamukkale University, School of Medicine, 20070, Denizli, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Misumi T, Tanaka T, Mikawa K, Nishina K, Morikawa O, Obara H. Effects of sivelestat, a new elastase inhibitor, on IL-8 and MCP-1 production from stimulated human alveolar epithelial type II cells. J Anesth 2006; 20:159-65. [PMID: 16897233 DOI: 10.1007/s00540-006-0391-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2005] [Accepted: 01/29/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE The alveolar epithelial cell type II (AEC-II) is itself able to amplify lung inflammation by producing inflammatory cytokines and chemokines, leading to the activation and recruitment of phagocytes. Sivelestat, a new neutrophil elastase inhibitor, has been shown to attenuate acute lung injury in animal experiments. In the current study, we assessed the effects of sivelestat on the production of chemokines from cultured A549 cells, a human AEC-II-like cell line. METHODS A549 cells were stimulated with endotoxin or tumor necrosis factor-alpha in the presence of sivelestat (1-100 microg x ml(-1)). Culture supernatant levels of interleukin-8 (IL-8) and monocyte chemoattractant protein-1 (MCP-1) were determined by enzyme-linked immunosorbent assay. The expression of IL-8 and MCP-1 mRNAs in stimulated A549 cells in the presence of sivelestat (100 microg x ml(-1)) was quantified by real-time polymerase chain reaction. RESULTS Sivelestat, at 100 microg x ml(-1) reduced the accumulation of IL-8 and MCP-1 in the culture medium. The high dose of sivelestat significantly inhibited the expression of IL-8 mRNA in A549 cells. The drug also decreased MCP-1 mRNA expression, although not significantly. CONCLUSION These data suggest that a high dose of sivelestat regulates the production of IL-8 and MCP-1 in AEC-II.
Collapse
Affiliation(s)
- Takuyo Misumi
- Department of Anesthesia and Perioperative Medicine, Faculty of Medical Sciences, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Kobe 650-0017, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Fujita Y, Fujino Y, Maeda Y, Uchiyama A, Mashimo T, Nishimura M. Effect of Ono-EI-600 elastase inhibitor on high-tidal-volume-induced lung injury in rats. J Anesth 2006; 20:141-4. [PMID: 16633777 DOI: 10.1007/s00540-005-0369-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Accepted: 11/08/2005] [Indexed: 11/25/2022]
Abstract
We tested the effect of Ono-EI-600, an elastase inhibitor that suppresses cytokine release, on ventilator-induced lung injury in a rat model. After Wistar rats (aged 8-11 weeks) were anesthetized and tracheostomized, they were randomly assigned to four groups: high tidal volume (V(T)) group (H group: n = 10) receiving peak inspiratory pressure (PIP) 30 cmH(2)O for 240 min; high V(T) with drug group (HD group: n = 10) receiving the same ventilation settings as H group and also intravenous infusion 10 mg x kg(-1) x h(-1) of Ono-EI-600 during the protocol; the lower V(T) group (L group: n = 5) receiving PIP 10 cmH(2)O for 240 min; and control group (C group: n = 5) receiving the same ventilation as L group for 30 min. The cytokine levels (IL-6 and CINC-1) in the bronchoalveolar lavage fluid (BALF) of the H group were significantly higher than those of the C and L groups (P < 0.05). However, for the H and HD groups, no differences were found in arterial blood gas data, cytokine levels in BALF, and histological injury scores. Our experiment provided no evidence that elastase inhibitor Ono-EI-600 protects against lung injury induced by high V(T) ventilation.
Collapse
Affiliation(s)
- Yasuki Fujita
- Intensive Care Unit, Osaka University Hospital, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Johnson FJ, Reynolds LJ, Toward TJ. Elastolytic activity and alveolar epithelial type-1 cell damage after chronic LPS inhalation: effects of dexamethasone and rolipram. Toxicol Appl Pharmacol 2005; 207:257-65. [PMID: 16129118 DOI: 10.1016/j.taap.2005.01.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2004] [Revised: 01/10/2005] [Accepted: 01/12/2005] [Indexed: 11/17/2022]
Abstract
This study investigated whether a correlation between leukocyte-derived elastolytic activity, alveolar epithelial type-1 cell damage, and leukocyte infiltration of the airways existed in guinea-pigs chronically exposed to inhaled lipopolysaccharide (LPS). The airway pathology of this model, notably the neutrophilia, resembles chronic obstructive pulmonary disease (COPD). The effect of the corticosteroid, dexamethasone, or the phosphodiesterase-4 (PDE4)-inhibitor, rolipram, on these features was studied. Conscious guinea-pigs were exposed for 1 h to single or repeated (nine) doses of LPS (30 microg ml(-1)). Dexamethasone (20 mg kg(-1), ip) or rolipram (1 mg kg(-1), ip) was administered 24 and 0.5 h before the first exposure and daily thereafter. Bronchoalveolar lavage fluid (BALF) was removed and elastolytic activity determined as the elastase-like release of Congo Red from impregnated elastin. The presence of the specific epithelial cell type-1 protein (40-42 kDa) RT1(40) in BALF was identified by Western blotting using a rat monoclonal antibody and semi-quantified by dot-blot analysis. The antibody was found to identify guinea-pig RT1(40). BALF inflammatory cells, particularly neutrophils and macrophages, and elastolytic activity were increased in chronic LPS-exposed guinea-pigs, the latter by 90%. Chronic LPS exposure also increased (10.5-fold) RT1(40) levels, indicating significant alveolar epithelial type-1 cell damage. Dexamethasone or rolipram treatment reduced the influx of inflammatory cells, the elastolytic activity (by 40% and 38%, respectively), and RT1(40) levels (by 50% and 57%, respectively). In conclusion, chronic LPS-exposed guinea-pigs, like COPD, exhibit elastolytic lung damage. This was prevented by a PDE4 inhibitor and supports their development for suppressing this leukocyte-mediated pathology.
Collapse
Affiliation(s)
- Frederick J Johnson
- Division of Pharmacology, Welsh School of Pharmacy, Cardiff University, King Edward VII Avenue, Cathays Park, Cardiff, CF10 3XF, UK.
| | | | | |
Collapse
|
40
|
Kabay B, Aytekin FO, Aydin C, Ozer A, Kabay N, Tekin K, Sungurtekin U, Erdem E, Ozden A. Interleukin-10 gene therapy attenuates pulmonary tissue injury caused by mesenteric ischemia-reperfusion in a mouse model. TOHOKU J EXP MED 2005; 207:133-42. [PMID: 16141682 DOI: 10.1620/tjem.207.133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
To investigate the role of interleukin (IL)-10 gene therapy on the reperfusion-induced lung injury, we utilised the technique of liposomal gene delivery before the induction of intestinal ischemia. Plasmid DNA encoding human IL10 (hIL-10) or empy vector was injected intraperitoneally 24 h before the study. Male Balb/c mice randomized into three groups: Sham operated control (n = 12), empty plasmid vector (n = 12), and hIL-10 gene therapy group (n = 12). The ischemia was generated by selective occlusion of superior mesenteric artery for 60 min and followed by reperfusion for 30 min. Lung tissue neutrophil infiltration was determined by myeloperoxidase assay and neutrophil counts. For the determination of lung tissue microvascular permeability, Evans blue dye injection was made and the lung edema was assessed by wet/dry ratio. hIL-10 protein expression was studied by immunostaining and ELISA. We found that pre-ischemic hIL-10 overexpression attenuated dye extravasation, leukocyte sequestration and reduced pulmonary tissue injury compared to the empty vector-injected control. Our study indicates that pre-ischemic hIL-10 overexpression attenuates lung injury caused by intestinal ischemia-reperfusion.
Collapse
Affiliation(s)
- Burhan Kabay
- Department of General Surgery, Pamukkale University Faculty of Medicine, Denizli, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Abstract
Endotoxin causes acute lung injury (ALI) through many mediators of inflammatory and immune responses. Propofol is an antiinflammatory and immunosuppressive drug. We conducted this study to evaluate whether propofol attenuates ALI associated with endotoxemia. Thirty-two anesthetized rabbits were randomly divided into four groups (n = 8 each). ALI was induced by IV endotoxin 5 mg/kg over 30 min in 3 groups. In 2 of the ALI groups, IV administration of propofol (2 or 5 mg/kg as a bolus followed by continuous infusion at 4 or 15 mg x kg(-1) x h(-1)) was started 15 min before endotoxin. The other ALI group received soybean-oil emulsion. The nonlung injury control group received infusion of both vehicles. The lungs were mechanically ventilated with 40% oxygen for 6 h after endotoxin. Hemodynamics did not differ among groups. The large dose of propofol attenuated lung leukosequestration, pulmonary edema (as assessed by lung wet/dry weight ratio), and pulmonary hyperpermeability (as assessed by albumin levels in bronchoalveolar lavage fluid) and resulted in better oxygenation, lung mechanics, and histological change. The small dose of propofol failed to do so. Our findings suggest that a large dose of propofol successfully mitigates physiological, biochemical, and histological deterioration in ALI in endotoxemia.
Collapse
Affiliation(s)
- Yumiko Takao
- Department of Anesthesia & Perioperative Medicine, Faculty of Medical Sciences, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | |
Collapse
|
43
|
Jian MY, Koizumi T, Tsushima K, Fujimoto K, Kubo K. Effects of Granulocyte Colony-Stimulating Factor (G-CSF) and Neutrophil Elastase Inhibitor (ONO-5046) on Acid-Induced Lung Injury in Rats. Inflammation 2004; 28:327-36. [PMID: 16245075 DOI: 10.1007/s10753-004-6643-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
It has been suggested that neutrophils play an important role in acid-aspirated lung injury. We examined the effects of the high dose of granulocyte-colony stimulating factor (G-CSF), which is capable of increasing peripheral neutrophils, and a specific neutrophil elastase inhibitor (ONO-5046) on acid lung injury in rats. Animals were anesthetized and normal saline (NS, 2 mL kg(-1)) or hydrochloric acid (HCl, 0.1 N 2 mL kg(-1)) was then instilled into trachea. Thirty minutes before HCl instillation, G-CSF (150 microg kg(-1)) was injected subcutaneously or ONO-5046 (10 mg kg(-1) h(-1)) was infused continuously into the right jugular vein. Animals were ventilated during the experiments. Five hours after HCl or NS instillation, bronchoalveolar lavage fluid (BALF) and lung tissue samples were obtained. Total nuclear cell count, absorbance, albumin, tumor necrosis factor (TNF)-alpha, interleukin (IL)-6, cytokine-induced neutrophil chemoattractant (CINC), neutrophil elastase in BALF, wet-to-dry (W/D) ratio were measured. HCl aspiration markedly increased these values in BALF and W/D ratio. Both ONO-5046 and G-CSF attenuated the parameters increased by acid-induced lung injury in rats. The data suggests that neutrophils play an important role in acid-induced lung injury. However, high-dose G-CSF does not exacerbate acid-aspirated lung injury in rats, although this agent causes an increase in peripheral neutrophils.
Collapse
Affiliation(s)
- Ming-Yuan Jian
- First Department of Medicine, Shinshu University School of Medicine, 3-1-1 Asahi Matsumoto 390-8621, Japan
| | | | | | | | | |
Collapse
|
44
|
Zeiher BG, Matsuoka S, Kawabata K, Repine JE. Neutrophil elastase and acute lung injury: prospects for sivelestat and other neutrophil elastase inhibitors as therapeutics. Crit Care Med 2002; 30:S281-7. [PMID: 12004249 DOI: 10.1097/00003246-200205001-00018] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To review the evidence and rationale that suggest that neutrophil elastase (NE) may contribute to the development of acute lung injury (ALI) and the acute respiratory distress syndrome. To review selected preliminary data regarding the effectiveness of NE inhibition in animals, in in vitro models, and in patients with ALI. DATA SOURCES The published literature and observations provided by Ono Pharmaceutical and Eli Lilly investigators and their colleagues. DATA SUMMARY Taken en toto, the data suggest that NE could contribute to ALI and endothelial cell injury that is relevant to ALI. Moreover, the toxic effects of NE are greatly enhanced by increased oxidative stress, which commonly occurs in patients with ALI. In addition to neutrophils, xanthine oxidase, a constituent of endothelial cells, is a potential source of oxidative stress in ALI; xanthine oxidase-derived oxidants enhance NE toxicity in in vivo, isolated lung, and in vitro endothelial cell test systems. Not surprisingly, endogenous nonoxidatively sensitive NE inhibitors (e.g., eglin C) are more effective in combating the detrimental effects of NE than oxidatively sensitive NE inhibitors (e.g., alpha-1-proteinase inhibitor). In addition, a synthetic NE inhibitor, sivelestat (ONO-5046 and LY544349), is effective in reducing measures of inflammation and injury in multiple animal models of ALI. In a trial of ALI patients with systemic inflammatory response syndrome, conducted in Japan by Ono Pharmaceutical scientists, sivelestat treatment improved the investigator assessment of global improvement and the percentages of patients who were removed from ventilators and transferred out of the intensive care unit. CONCLUSIONS Further study of the role of NE inhibition as a treatment for ALI is warranted. Additional clinical and preclinical studies with sivelestat and various other NE inhibitors should not only clarify the clinical potential of this intervention strategy, but also better define the activities of NE in inflammatory disorders such as ALI and multiple organ failure.
Collapse
Affiliation(s)
- Bernhardt G Zeiher
- Critical Care Clinical Research, Eli Lilly and Company, Indianapolis, IN 46285, USA.
| | | | | | | |
Collapse
|
45
|
Ueno M, Moriyama Y, Toda R, Yotsumoto G, Yamamoto H, Fukumoto Y, Sakasegawa K, Nakamura K, Sakata R. Effect of a neutrophil elastase inhibitor (ONO-5046 Na) on ischemia/reperfusion injury using the left-sided heterotopic canine heart transplantation model. J Heart Lung Transplant 2001; 20:889-96. [PMID: 11502411 DOI: 10.1016/s1053-2498(01)00281-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Ischemia/reperfusion injury is a major cause of transplanted heart dysfunction. Several reports have demonstrated that polymorphonuclear neutrophil (PMN) elastase derived from the activated neutrophils might play an important role in this injury. Herein, we investigated the protective effects of PMN elastase inhibitor (ONO-5046 Na) on ischemia/reperfusion injury using a left-sided canine heterotopic heart transplantation model. METHODS We used 10 pairs of adult beagle dogs. The donor heart was transplanted heterotopically into the left thoracic cavity of the recipient without cardiopulmonary bypass. A bolus of ONO-5046 Na (10 mg/kg) was introduced intravenously to 5 recipients (group II) at 15 minutes before reperfusion and was followed by continuous infusion (10 mg/kg per hour) for 180 minutes. Five dogs (group I) did not receive ONO-5046 Na and thus served as a control. After reperfusion, we evaluated transplanted heart function and obtained blood samples from the coronary sinus over a 360-minute period. RESULTS E(max) and pre-load recruitable stroke work in group II showed significantly better recovery than group I. Blood levels of PMN elastase, creatine kinase MB, lactate and inflammatory cytokines (tumor necrosis factor-alpha, interleukin-6, interleukin-8) were significantly lower in group II. Depletion of myocardial concentration of adenosine triphosphate at 120 minutes after reperfusion and myocardial water content was significantly lower in group II. CONCLUSIONS ONO-5046 Na, which inhibits PMN elastase, could reduce ischemia/reperfusion injury in heart transplantation. These results indicate that clinical application of ONO-5046 Na should be considered.
Collapse
Affiliation(s)
- M Ueno
- Second Department of Surgery, Faculty of Medicine, Kagoshima University, Kagoshima, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mathias MA, Tribble CG, Dietz JF, Nguyen RP, Shockey KS, Kern JA, Kron IL. Aprotinin improves pulmonary function during reperfusion in an isolated lung model. Ann Thorac Surg 2000; 70:1671-4. [PMID: 11093508 DOI: 10.1016/s0003-4975(00)01767-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND We hypothesized that the use of aprotinin would ameliorate the reperfusion injury observed after lung transplantation because of a reduction in the inflammatory response. METHODS We used an isolated, whole blood-perfused, ventilated rabbit lung model to study the effects of aprotinin during reperfusion. The control animals (group A, n = 8) underwent lung harvest after pulmonary arterial prostaglandin E1 injection and Euro-Collins preservation flush before saline storage for 18 hours at 4 degrees C. The experimental groups received either a low dose (3,000 KIU/mL; group B, n = 8) or a high dose (10,000 KIU/mL; group C, n = 8) of aprotinin added to the pulmonary flush before storage. Each lung was reperfused at 37 degrees C at a rate of 60 mL/min. RESULTS The arterial partial pressure of oxygen values of group B (low-dose aprotinin) were significantly higher than those of group A (control) after 10 minutes of reperfusion (69.19 +/- 5.69 mm Hg versus 264.30 +/- 48.59 mm Hg, respectively, p = 0.001). Similar results were recorded at 20 and at 30 minutes of reperfusion. Similarly, after 10 minutes of reperfusion, the differences between groups A and C were 69.19 +/- 5.69 mm Hg versus 235.91 +/- 28.63 mm Hg, respectively (p = 0.001). CONCLUSIONS The addition of aprotinin to the Euro-Collins pulmonary flush significantly improves arterial oxygenation in the early reperfusion period. The enhanced oxygenation suggests that aprotinin may offer protection against early reperfusion injury.
Collapse
Affiliation(s)
- M A Mathias
- Department of Surgery, University of Virginia Health Sciences Center, Charlottesville 22908, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Remarkable progress has been made in the past 10 years with regard to understanding the interplay of potent physiologic mediators in patients with acute lung injury. Because there are so many mediators and the interaction of these agents is complex, true insight into the process has been slow in coming. Clinical studies in ARDS, as well as sepsis, the leading cause of ARDS, have increased in number, size, and quality over this same period. Although none of these studies has produced an accepted new therapy for ARDS, each has laid the groundwork for more efficient and more elegant studies of the problem. The stage is now set for the real advances to be brought forward and put to rigorous, efficient clinical testing.
Collapse
Affiliation(s)
- B D Conner
- Center for Lung Research, Vanderbilt University, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|
48
|
Hayashi H, Koike H, Imanishi N, Tojo SJ. The effects of delayed treatment with sialyl Lewis X against lipopolysaccharide-induced acute lung injury in rabbits. Eur J Pharmacol 2000; 392:109-16. [PMID: 10748279 DOI: 10.1016/s0014-2999(00)00099-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The therapeutic effects of a selectin inhibitor against lipopolysaccharide-induced acute lung injury were studied in rabbits by using sialyl Lewis X-oligosaccharide. Lipopolysaccharide-induced acute lung injury, as characterized by an impairment of pulmonary gas exchange, clinically resembles that of the acute respiratory distress syndrome. Delayed treatments with sialyl Lewis X-oligosaccharide (55 mg kg(-1) i.v. bolus injection 0.5, 1 or 2 h after lipopolysaccharide administration+36 mg kg(-1) h(-1) i.v. infusion for 5.5, 5 or 4 h, respectively) prevented the lipopolysaccharide-induced impairments in pulmonary gas exchange, as well as the accumulation of polymorphonuclear leukocytes in the lung tissue. In contrast, this agent had no significant effects on lipopolysaccharide-induced systemic hypotension, the decrease in the number of circulating white blood cells and platelets or the decline in blood pH. This is the first demonstration that sialyl Lewis X-oligosaccharide is effective against the impairments in pulmonary gas exchange even if administered 0.5, 1 or 2 h following the lipopolysaccharide injection.
Collapse
Affiliation(s)
- H Hayashi
- Sumitomo Pharmaceuticals Research Center, 1-98 Kasugadenaka 3-chome, Konohana-ku, Osaka, Japan
| | | | | | | |
Collapse
|
49
|
Kishi M, Richard LF, Webster RO, Dahms TE. Role of neutrophils in xanthine/xanthine oxidase-induced oxidant injury in isolated rabbit lungs. J Appl Physiol (1985) 1999; 87:2319-25. [PMID: 10601184 DOI: 10.1152/jappl.1999.87.6.2319] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Reactive oxygen species have been shown to play an important role in the pathogenesis of lung injury. This study was designed to clarify the role of intrapulmonary neutrophils in the development of xanthine/xanthine oxidase (X/XO)-induced lung injury in isolated buffer-perfused rabbit lungs. We measured microvascular fluid filtration coefficient (K(f)) and wet-to-dry weight ratio to assess lung injury. X/XO induced a significant increase in K(f) and wet-to-dry weight ratio in neutrophil-replete lungs, whereas the lung injury was attenuated in neutrophil-depleted lungs. A neutrophil elastase inhibitor, ONO-5046, also attenuated the lung injury. In addition, X/XO induced a transient pulmonary arterial pressure (P(pa)) increase. The thromboxane inhibitor OKY-046 attenuated the P(pa) increase but did not alter the increase in permeability. Neutrophil depletion reduced the K(f) increase but had no effect on the P(pa) increase. These results suggest that intrapulmonary neutrophils activated by X/XO play a major role in development of the lung injury, that neutrophil elastase is involved in the injury, and that the X/XO-induced vasoconstriction is independent of intrapulmonary neutrophils.
Collapse
Affiliation(s)
- M Kishi
- Department of Anesthesiology, Saint Louis University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|
50
|
Hayashi H, Koike H, Kurata Y, Imanishi N, Tojo SJ. Protective effects of sialyl Lewis X and anti-P-selectin antibody against lipopolysaccharide-induced acute lung injury in rabbits. Eur J Pharmacol 1999; 370:47-56. [PMID: 10323279 DOI: 10.1016/s0014-2999(99)00068-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The prophylactic effects of selectin inhibitors on lipopolysaccharide-induced acute lung injury were studied in rabbits by using sialyl Lewis X-oligosaccharide and PB1.3, an anti-human P-selectin monoclonal antibody. Lipopolysaccharide-induced acute lung injury resembles that of the acute respiratory distress syndrome, in which there is a decrease in arterial blood oxygen tension (PaO2) and an increase in the difference between alveolar and arterial oxygen tension (A-aDO2). Prophylactic treatment with the selectin inhibitors, sialyl Lewis X-oligosaccharide (55 mg kg(-1) i.v. bolus injection immediately before lipopolysaccharide administration + 36 mg kg(-1) h(-1) i.v. infusion for 4 h) and PB1.3 (5 mg kg(-1) i.v. bolus injection immediately before lipopolysaccharide administration), prevented the lipopolysaccharide-induced impairments in pulmonary gas exchange. In contrast, these agents had no significant effects on lipopolysaccharide-induced systemic hypotension, the decrease in the number of circulating white blood cells and platelets, the decline in blood pH, or the increase in arterial CO2 tension (PaCO2). These results indicate that selectin inhibitors including sialyl Lewis X-oligosaccharide and the anti-P-selectin antibody, PB1.3, attenuate lipopolysaccharide-induced acute lung injury in rabbits. This is the first demonstration that P-selectin is directly involved in the development of lipopolysaccharide-induced impairments in pulmonary gas exchange.
Collapse
Affiliation(s)
- H Hayashi
- Sumitomo Pharmaceuticals Research Center, Osaka, Japan
| | | | | | | | | |
Collapse
|