1
|
Salazar-Puerta AI, Rincon-Benavides MA, Cuellar-Gaviria TZ, Aldana J, Martinez GV, Ortega-Pineda L, Das D, Dodd D, Spencer CA, Deng B, McComb DW, Englert JA, Ghadiali S, Zepeda-Orozco D, Wold LE, Gallego-Perez D, Higuita-Castro N. Engineered Extracellular Vesicles Derived from Dermal Fibroblasts Attenuate Inflammation in a Murine Model of Acute Lung Injury. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210579. [PMID: 37119468 PMCID: PMC10573710 DOI: 10.1002/adma.202210579] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/29/2023] [Indexed: 06/06/2023]
Abstract
Acute respiratory distress syndrome (ARDS) represents a significant burden to the healthcare system, with ≈200 000 cases diagnosed annually in the USA. ARDS patients suffer from severe refractory hypoxemia, alveolar-capillary barrier dysfunction, impaired surfactant function, and abnormal upregulation of inflammatory pathways that lead to intensive care unit admission, prolonged hospitalization, and increased disability-adjusted life years. Currently, there is no cure or FDA-approved therapy for ARDS. This work describes the implementation of engineered extracellular vesicle (eEV)-based nanocarriers for targeted nonviral delivery of anti-inflammatory payloads to the inflamed/injured lung. The results show the ability of surfactant protein A (SPA)-functionalized IL-4- and IL-10-loaded eEVs to promote intrapulmonary retention and reduce inflammation, both in vitro and in vivo. Significant attenuation is observed in tissue damage, proinflammatory cytokine secretion, macrophage activation, influx of protein-rich fluid, and neutrophil infiltration into the alveolar space as early as 6 h post-eEVs treatment. Additionally, metabolomics analyses show that eEV treatment causes significant changes in the metabolic profile of inflamed lungs, driving the secretion of key anti-inflammatory metabolites. Altogether, these results establish the potential of eEVs derived from dermal fibroblasts to reduce inflammation, tissue damage, and the prevalence/progression of injury during ARDS via nonviral delivery of anti-inflammatory genes/transcripts.
Collapse
Affiliation(s)
- Ana I. Salazar-Puerta
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
| | - María A. Rincon-Benavides
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States
| | | | - Julian Aldana
- Biochemistry Program, The Ohio State University, Columbus, Ohio, United States
| | - Gabriela Vasquez Martinez
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States
| | - Lilibeth Ortega-Pineda
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Devleena Das
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Daniel Dodd
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biomedical Science Graduate Program, The Ohio State University, Columbus, Ohio, United States
| | - Charles A. Spencer
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| | - Binbin Deng
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, Ohio, United States
| | - David W. McComb
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, Ohio, United States
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Joshua A. Englert
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Samir Ghadiali
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Diana Zepeda-Orozco
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States
- Division of Pediatric Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, Ohio, United States
| | - Loren E. Wold
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States
- Division of General Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States
- Division of General Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
2
|
Zhou W, Hou Y, Yu T, Wang T, Ding Y, Nie H. Submersion and hypoxia inhibit alveolar epithelial Na + transport through ERK/NF-κB signaling pathway. Respir Res 2023; 24:117. [PMID: 37095538 PMCID: PMC10127099 DOI: 10.1186/s12931-023-02428-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/19/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Hypoxia is associated with many respiratory diseases, partly due to the accumulation of edema fluid and mucus on the surface of alveolar epithelial cell (AEC), which forms oxygen delivery barriers and is responsible for the disruption of ion transport. Epithelial sodium channel (ENaC) on the apical side of AEC plays a crucial role to maintain the electrochemical gradient of Na+ and water reabsorption, thus becomes the key point for edema fluid removal under hypoxia. Here we sought to explore the effects of hypoxia on ENaC expression and the further mechanism related, which may provide a possible treatment strategy in edema related pulmonary diseases. METHODS Excess volume of culture medium was added on the surface of AEC to simulate the hypoxic environment of alveoli in the state of pulmonary edema, supported by the evidence of increased hypoxia-inducible factor-1 expression. The protein/mRNA expressions of ENaC were detected, and extracellular signal-regulated kinase (ERK)/nuclear factor κB (NF-κB) inhibitor was applied to explore the detailed mechanism about the effects of hypoxia on epithelial ion transport in AEC. Meanwhile, mice were placed in chambers with normoxic or hypoxic (8%) condition for 24 h, respectively. The effects of hypoxia and NF-κB were assessed through alveolar fluid clearance and ENaC function by Ussing chamber assay. RESULTS Hypoxia (submersion culture mode) induced the reduction of protein/mRNA expression of ENaC, whereas increased the activation of ERK/NF-κB signaling pathway in parallel experiments using human A549 and mouse alveolar type 2 cells, respectively. Moreover, the inhibition of ERK (PD98059, 10 µM) alleviated the phosphorylation of IκB and p65, implying NF-κB as a downstream pathway involved with ERK regulation. Intriguingly, the expression of α-ENaC could be reversed by either ERK or NF-κB inhibitor (QNZ, 100 nM) under hypoxia. The alleviation of pulmonary edema was evidenced by the administration of NF-κB inhibitor, and enhancement of ENaC function was supported by recording amiloride-sensitive short-circuit currents. CONCLUSIONS The expression of ENaC was downregulated under hypoxia induced by submersion culture, which may be mediated by ERK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Tong Yu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Tingyu Wang
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Zhao W, Zhang C, Mu D, Cui F, Jia H. Muscular tissue desaturation and pneumonia in patients receiving lung cancer surgery: a cohort study. Chin Med J (Engl) 2023; 136:65-72. [PMID: 36780417 PMCID: PMC10106230 DOI: 10.1097/cm9.0000000000002497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Post-operative pneumonia (POP) is a common complication of lung cancer surgery, and muscular tissue oxygenation is a root cause of post-operative complications. However, the association between muscular tissue desaturation and POP in patients receiving lung cancer surgery has not been specifically studied. This study aimed to investigate the potential use of intra-operative muscular tissue desaturation as a predictor of POP in patients undergoing lung cancer surgery. METHODS This cohort study enrolled patients (≥55 years) who had undergone lobectomy with one-lung ventilation. Muscular tissue oxygen saturation (SmtO 2 ) was monitored in the forearm (over the brachioradialis muscle) and upper thigh (over the quadriceps) using a tissue oximeter. The minimum SmtO 2 was the lowest intra-operative measurement at any time point. Muscular tissue desaturation was defined as a minimum baseline SmtO 2 of <80% for >15 s. The area under or above the threshold was the product of the magnitude and time of desaturation. The primary outcome was the association between intra-operative muscular tissue desaturation and POP within seven post-operative days using multivariable logistic regression. The secondary outcome was the correlation between SmtO 2 in the forearm and that in the thigh. RESULTS We enrolled 174 patients. The overall incidence of muscular desaturation (defined as SmtO 2 < 80% in the forearm at baseline) was approximately 47.1% (82/174). The patients with muscular desaturation had a higher incidence of pneumonia than those without desaturation (28.0% [23/82] vs. 12.0% [11/92]; P = 0.008). The multivariable analysis revealed that muscular desaturation was associated with an increased risk of pneumonia (odds ratio: 2.995, 95% confidence interval: 1.080-8.310, P = 0.035) after adjusting for age, American Society of Anesthesiologists status, Assess Respiratory Risk in Surgical Patients in Catalonia score, smoking, use of peripheral nerve block, propofol, and study center. CONCLUSION Muscular tissue desaturation, defined as a baseline SmtO 2 < 80% in the forearm, may be associated with an increased risk of POP. TRIAL REGISTRATION No. ChiCTR-ROC-17012627.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Caijuan Zhang
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Department of Anesthesiology, Peking University First Hospital, Beijing 100034, China
- Department of Anesthesiology, Tangshan Gongren Hospital, Tangshan, Hebei 063000, China
| | - Dongliang Mu
- Department of Anesthesiology, Peking University First Hospital, Beijing 100034, China
| | - Fan Cui
- Department of Anesthesiology, Peking University First Hospital, Beijing 100034, China
| | - Huiqun Jia
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
4
|
Belhaj A, Dewachter L, Hupkens E, Remmelink M, Galanti L, Rorive S, Melot C, Naeije R, Rondelet B. Tacrolimus Prevents Mechanical and Humoral Alterations in Brain Death-Induced Lung Injury in Pigs. Am J Respir Crit Care Med 2022; 206:584-595. [PMID: 35549669 DOI: 10.1164/rccm.202201-0033oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Donor brain death-induced lung injury may compromise graft function after transplantation. Establishing strategies to attenuate lung damage remains a challenge because the underlying mechanisms remain uncertain. OBJECTIVES The effects of tacrolimus pretreatment were evaluated in an experimental model of brain death-induced lung injury. METHODS Brain death was induced by slow intracranial infusion of blood in anesthetized pigs after randomization to tacrolimus (orally administered at 0.25 mg. kg-1 BID the day before the experiment and intravenously at 0.05 mg. kg-1 one hour before the experiment; n=8) or placebo (n=9) pretreatment. Hemodynamic measurements were performed 1, 3, 5 and 7 hours after brain death. After euthanasia of the animals, lung tissue was sampled for pathobiological and histological analysis, including lung injury scoring (LIS). MEASUREMENTS AND MAIN RESULTS Tacrolimus pretreatment prevented increases in pulmonary artery pressure, pulmonary vascular resistance and pulmonary capillary pressure and decreases in systemic artery pressure and thermodilution cardiac output associated with brain death. After brain death, the ratio of the partial arterial O2 pressure to the inspired O2 fraction (PaO2/FiO2) decreased, which was prevented by tacrolimus. Tacrolimus pretreatment prevented increases in the interleukin (IL)-6-to-IL-10 ratio, vascular cell adhesion molecule-1, circulating levels of IL-1β, IL-6-to-IL-10 ratio and glycocalyx-derived molecules. Tacrolimus partially decreased apoptosis [Bax-to-Bcl2 ratio (p=0.07) and the number of apoptotic cells in the lungs (p<0.05)] but failed to improve LIS. CONCLUSIONS Immunomodulation through tacrolimus pretreatment prevented pulmonary capillary hypertension as well as the activation of inflammatory and apoptotic processes in the lungs after brain death; however, LIS did not improve.
Collapse
Affiliation(s)
- Asmae Belhaj
- CHU UCL Namur, 82470, cardiovascular, thoracic surgery and lung transplantation, Yvoir, Belgium.,Université Libre de Bruxelles, 26659, Laboratory of Physiology and Pharmacology, Faculty of Medicine, Bruxelles, Belgium;
| | - Laurence Dewachter
- Université Libre de Bruxelles, 26659, Laboratory of Physiology and Pharmacology, Faculty of Medicine, Bruxelles, Belgium
| | - Emeline Hupkens
- Université Libre de Bruxelles, 26659, Laboratory of Physiology and Pharmacology, Faculty of Medicine, Bruxelles, Belgium
| | - Myriam Remmelink
- Université Libre de Bruxelles, 26659, Department of Pathology, Hôpital Erasme, Brussels, Belgium
| | - Laurence Galanti
- CHU UCL Namur, 82470, Department of Clinical Biology, Yvoir, Belgium
| | - Sandrine Rorive
- Université Libre de Bruxelles, 26659, Department of Pathology, Hôpital Erasme, Brussels, Belgium
| | - Christian Melot
- Université Libre de Bruxelles, 26659, Laboratory of Physiology and Pharmacology, Faculty of Medicine, Bruxelles, Belgium
| | - Robert Naeije
- Department of Pathophysiology, Free University of Brussels, Brussels, Belgium
| | - Benoît Rondelet
- CHU UCL Namur, 82470, cardiovascular, thoracic surgery and lung transplantation, Yvoir, Belgium.,Université Libre de Bruxelles, 26659, Laboratory of Physiology and Pharmacology, Faculty of Medicine, Bruxelles, Belgium
| |
Collapse
|
5
|
Li H, Yao C, Shi K, Zhao Y, Du J, Hu D, Liu Z. Astragaloside IV attenuates hypoxia/reoxygenation injury-induced apoptosis of type II alveolar epithelial cells through miR-21-5p. Bioengineered 2021; 12:7747-7754. [PMID: 34617873 PMCID: PMC8806943 DOI: 10.1080/21655979.2021.1982845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
We aimed to explore the role of miR-21-5p in the inhibitory effects of astragaloside IV (As-IV) on hypoxia/reoxygenation injury-induced apoptosis of type II alveolar epithelial cells. Rat type II alveolar epithelial cells RLE-6TN were cultured in vitro and randomly divided into control (C), hypoxia/reoxygenation injury (H/R), As-IV and miR-21-5p-siRNA + As-IV groups (n = 6). H/R model was established by 24 h of hypoxia and 4 h of reoxygenation. As-IV group was given 1 nmol/L As-IV and incubated for 1 h before modeling. MiR-21-5p-siRNA + As-IV group was transfected with 50 nmol/L miR-21-5p-siRNA. After 48 h, they were incubated with 1 nmol/L As-IV for 1 h before modeling. Cell viability was detected by cell counting kit-8 assay, and apoptosis rate was detected by flow cytometry. The expression levels of TLR4 and NF-κB were measured by immunofluorescence assay. The targeting relationship between miR-21-5p and TLR4 was determined by luciferase assay. Compared with H/R group, the cell viability, miR-21-5p, bax and cleaved caspase-3 expressions of As-IV group increased, apoptosis rate and Bcl-2 expression decreased, and TLR4 and NF-κB expressions were down-regulated (P < 0.05). Compared with As-IV group, the cell viability, miR-21-5p, bax and cleaved caspase-3 expressions of miR-21-5p-siRNA + As-IV group decreased, apoptosis rate and Bcl-2 expression increased, and the expressions of TLR4 and NF-κB were up-regulated (P < 0.05). As-IV up-regulates miR-21-5p expression, inhibits the TLR4/NF-κB signaling pathway and suppresses the apoptosis of type II alveolar epithelial cells during hypoxia/reoxygenation injury.
Collapse
Affiliation(s)
- Hang Li
- Department of Cardiothoracic Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.,Department of Cardiothoracic Surgery, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Chang Yao
- Department of Breast Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Kaihu Shi
- Department of Cardiothoracic Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.,Department of Cardiothoracic Surgery, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yang Zhao
- Department of Cardiothoracic Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.,Department of Cardiothoracic Surgery, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Jin Du
- Department of Cardiothoracic Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.,Department of Cardiothoracic Surgery, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Dinghui Hu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.,Department of Cardiothoracic Surgery, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Zuntao Liu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.,Department of Cardiothoracic Surgery, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu Province, China
| |
Collapse
|
6
|
Liu PL, Chong IW, Lee YC, Tsai JR, Wang HM, Hsieh CC, Kuo HF, Liu WL, Chen YH, Chen HL. Anti-inflammatory Effects of Resveratrol on Hypoxia/Reoxygenation-Induced Alveolar Epithelial Cell Dysfunction. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:9480-9487. [PMID: 26466890 DOI: 10.1021/acs.jafc.5b01168] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Reducing oxidative stress is crucial to prevent hypoxia-reoxygenation (H/R)-induced lung injury. Resveratrol has excellent antioxidant and anti-inflammatory effects, and this study investigated its role in H/R-induced type II pneumocyte dysfunction. H/R conditions increased expression of inflammatory cytokines including interleukin (IL)-1β (142.3 ± 21.2%, P < 0.05) and IL-6 (301.9 ± 35.1%, P < 0.01) in a type II alveolar epithelial cell line (A549), while the anti-inflammatory cytokine IL-10 (64.6 ± 9.8%, P < 0.05) and surfactant proteins (SPs) decreased. However, resveratrol treatment effectively inhibited these effects. H/R significantly activated an inflammatory transcription factor, nuclear factor (NF)-κB, while resveratrol significantly inhibited H/R-induced NF-κB transcription activities. To the best of our knowledge, this is the first study showing resveratrol-mediated reversal of H/R-induced inflammatory responses and dysfunction of type II pneumocyte cells in vitro. The effects of resveratrol were partially mediated by promoting SP expression and inhibiting inflammation with NF-κB pathway involvement. Therefore, our study provides new insights into mechanisms underlying the action of resveratrol in type II pneumocyte dysfunction.
Collapse
Affiliation(s)
- Po-Len Liu
- Department of Respiratory Therapy, Department of Fragrance and Cosmetic Science, College of Medicine, Kaohsiung Medical University , Kaohsiung 807, Taiwan
| | - Inn-Wen Chong
- Department of Respiratory Therapy, Department of Fragrance and Cosmetic Science, College of Medicine, Kaohsiung Medical University , Kaohsiung 807, Taiwan
- Department of Pediatrics, Department of Internal Medicine, Department of Chest Surgery, Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Medical University Hospital , Kaohsiung 807, Taiwan
| | - Yi-Chen Lee
- Department of Respiratory Therapy, Department of Fragrance and Cosmetic Science, College of Medicine, Kaohsiung Medical University , Kaohsiung 807, Taiwan
| | - Jong-Rung Tsai
- Department of Respiratory Therapy, Department of Fragrance and Cosmetic Science, College of Medicine, Kaohsiung Medical University , Kaohsiung 807, Taiwan
- Department of Pediatrics, Department of Internal Medicine, Department of Chest Surgery, Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Medical University Hospital , Kaohsiung 807, Taiwan
| | - Hui-Min Wang
- Department of Respiratory Therapy, Department of Fragrance and Cosmetic Science, College of Medicine, Kaohsiung Medical University , Kaohsiung 807, Taiwan
| | - Chong-Chao Hsieh
- Department of Pediatrics, Department of Internal Medicine, Department of Chest Surgery, Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Medical University Hospital , Kaohsiung 807, Taiwan
| | - Hsuan-Fu Kuo
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University , Kaohsiung 801, Taiwan
| | - Wei-Lun Liu
- Department of Intensive Care Medicine, Chi Mei Medical Center , Tainan 736, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University , Taichung 404, Taiwan
- Department of Medical Research, China Medical University Hospital , Taichung 404, Taiwan
- Department of Psychology, College of Medical and Health Science, Asia University , Taichung 413, Taiwan
| | - Hsiu-Lin Chen
- Department of Respiratory Therapy, Department of Fragrance and Cosmetic Science, College of Medicine, Kaohsiung Medical University , Kaohsiung 807, Taiwan
- Department of Pediatrics, Department of Internal Medicine, Department of Chest Surgery, Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Medical University Hospital , Kaohsiung 807, Taiwan
| |
Collapse
|
7
|
Merry HE, Phelan P, Doaks M, Zhao M, Mulligan MS. Functional roles of tumor necrosis factor-alpha and interleukin 1-Beta in hypoxia and reoxygenation. Ann Thorac Surg 2015; 99:1200-5. [PMID: 25686667 DOI: 10.1016/j.athoracsur.2014.11.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/23/2014] [Accepted: 11/17/2014] [Indexed: 11/16/2022]
Abstract
BACKGROUND Intercellular signaling plays an important role in the development of lung ischemia-reperfusion injury. However, the role of specific mediators remains poorly characterized. Alveolar macrophages (AM) produce soluble mediators early in reperfusion, which modulate the responses of endothelial and epithelial cells to oxidative stress. There is a burst of proinflammatory cytokine production in a variety of cells; however, interleukin 1-beta (IL-1β) and tumor necrosis factor-alpha (TNF-α) localize to the AM. We hypothesized that these cytokines account for the costimulatory effects that AM exert on endothelial and epithelial cells. METHODS Activated AM media was placed on cultured rat type 2 pneumocytes and pulmonary artery endothelial cells, which were then subjected to hypoxia and reoxygenation. To assess the contributions of IL-1β and TNF-α, the cells were treated with control media or media that had been depleted of IL-1β or TNF-α. To deplete specific cytokines, activated media was passed through a column with immobilized IL-1β or TNF-α antibodies. Nuclear translocation of transcription factors, mitogen-activated protein kinase activation, and cytokine and chemokine production were assessed. RESULTS Depletion of IL-1β or TNF-α effectively eliminated the ability of AM media to enhance the response of endothelial and epithelial cells to oxidative stress. There were significant reductions in monocyte chemotactic protein 1 and cytokine-induced neutrophil chemoattractant (CINC) production (p < 0.05) at 4 hours of reperfusion. Additionally there was decreased nuclear translocation of nuclear factor-kappa B, and extracellular signal-regulated kinase phosphorylation. CONCLUSIONS Interleukin 1-beta and TNF-α are critical mediators in the intercellular communication pathways that allow the AM to enhance the response of surrounding cells to oxidative stress.
Collapse
Affiliation(s)
- Heather E Merry
- Division of Thoracic Surgery, Department of Surgery, University of Washington, Seattle, Washington
| | - Patrick Phelan
- Division of Thoracic Surgery, Department of Surgery, University of Washington, Seattle, Washington
| | - Matthew Doaks
- Division of Thoracic Surgery, Department of Surgery, University of Washington, Seattle, Washington
| | - Minqing Zhao
- Division of Thoracic Surgery, Department of Surgery, University of Washington, Seattle, Washington
| | - Michael S Mulligan
- Division of Thoracic Surgery, Department of Surgery, University of Washington, Seattle, Washington.
| |
Collapse
|
8
|
A murine model of lung ischemia and reperfusion injury: tricks of the trade. J Surg Res 2014; 194:659-666. [PMID: 25588947 DOI: 10.1016/j.jss.2014.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/19/2014] [Accepted: 12/03/2014] [Indexed: 11/23/2022]
Abstract
BACKGROUND Pulmonary ischemia-reperfusion injury (IRI) causes postoperative morbidity in patients undergoing lung transplantation, isolated lung perfusion, and cardiopulmonary bypass and may lead to potentially lethal pathologies such as respiratory shock. In-depth study of this pathology requires a reliable animal model. Mice are a popular species to develop experimental models because of their logistic advantages and the availability of knock outs. However, their small size warrants microsurgical techniques and a skilled surgeon. MATERIALS AND METHODS We developed a murine model of pulmonary anoxic IRI through hilar clamping using adult female Swiss mice. After left thoracotomy, we expose the pulmonary hilum keeping the ribs and the muscles of back and forepaw intact. A microvascular clamp is placed over the entire hilum, occluding bronchus, pulmonary artery, and vein. RESULTS Our model proved to be simple, reliable, and reproducible, showing minimal preoperative and postoperative mortality. Histopathologic analysis indicated all characteristic features of pulmonary IRI, such as an early recruitment of lymphocytes followed by neutrophil influx. CONCLUSIONS This article presents a murine surgery model for pulmonary IRI based on a muscle-sparing thoracotomy. The minimal approach limits manipulation of lung tissue, minimizing mortality and non-IRI-induced injury.
Collapse
|
9
|
McCourtie AS, Merry HE, Wolf PS, FitzSullivan E, Keech JC, Farivar AS, Mulligan MS. Synergistic protection in lung ischemia-reperfusion injury with calcineurin and thrombin inhibition. Ann Thorac Surg 2010; 89:1766-71. [PMID: 20494024 DOI: 10.1016/j.athoracsur.2010.02.068] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 02/18/2010] [Accepted: 02/22/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND Ischemia-reperfusion injury impairs lung transplant outcomes. The transcription factors, activator protein-1, and nuclear factor kappa B, are activated early in reperfusion and drive the development of injury. Thrombin inhibition with hirudin, and calcineurin inhibition with tacrolimus have independently been shown to ameliorate lung ischemia-reperfusion injury by reducing activator protein-1 and nuclear factor kappa B activation, respectively. However, high doses were required to achieve protection using individual agents, raising concerns about potential toxicities. We sought to determine if low-dose combination therapy reduced injury through synergistic inhibition of pretranscriptional signaling events. METHODS Rats were pretreated with either intravenous hirudin or tacrolimus at low doses or high doses, or both at low doses, prior to undergoing left lung ischemia and reperfusion. Lungs were assessed for markers of lung injury, including bronchoalveolar lavage cytokine-chemokine content and transcription factor transactivation of activator protein-1 and nuclear factor kappa B. RESULTS High-dose monotherapy with hirudin or tacrolimus reduced lung injury and transactivation of activator protein-1 and nuclear factor kappa B activation, respectively, whereas low-dose monotherapy with either agent did not alter transcription factor activation or lung injury compared with positive controls. Low-dose combination therapy was more protective than high-dose monotherapy with either drug, and correlated with a reduction in activation of both transcription factors and their associated cytokines. CONCLUSIONS The significant decrease in lung injury severity and transcription factor activation with combined pathway inhibition suggests pretranscriptional signaling redundancy between the calcineurin and thrombin dependent pathways in lung reperfusion injury.
Collapse
Affiliation(s)
- Anton S McCourtie
- Department of Surgery, Division of Cardiothoracic Surgery, University of Washington Medical Center, Seattle, Washington, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Ma L, Wu X, Chen W, Fujino Y. Propofol has anti-inflammatory effects on alveolar type II epithelial cells. Acta Anaesthesiol Scand 2010; 54:362-9. [PMID: 19764911 DOI: 10.1111/j.1399-6576.2009.02127.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND We investigated whether lipopolysaccharide (LPS) induced inflammation in alveolar epithelial type II (ATII) cells is through cluster of differentiation 14 (CD14) and Toll-like receptor 4 (TLR4) and the effect of different dosages of propofol on the inflammation in primary cultured rat ATII cells. METHODS Cultured ATII cells were randomly assigned to one of the following five groups: Group C: untreated group (control) cultured in the absence of propofol and LPS; Group LPS: treated with 1 microg/ml LPS; Group P1: treated with 1 microg/ml LPS and 25 microM propofol; Group P2: treated with 1 microg/ml LPS and 50 microM propofol; Group P3: treated with 1 microg/ml LPS and 100 microM propofol. ATII cells in all groups were cultured at 37 degrees C for 3 h. CD14 and TLR4 mRNA was detected using real-time polymerase chain reaction. Western blot was used to detect CD14 and TLR4 protein expression. CD14 and TLR4 expression on the ATII cells was imaged using immunofluorescence. Tumor necrosis factor-alpha (TNF-alpha) production was determined using an ELISA kit. RESULTS LPS stimulation resulted in an increased CD14 and TLR4 expression and increased TNF-alpha production in ATII cells. Propofol, at concentrations > or = 50 microM, significantly (P<0.05) and dose-dependently decreased CD14 and TLR4 mRNA expression and protein expression in ATII cells. This was accompanied by a decrease in TNF-alpha production (P<0.05). CONCLUSION These results suggest that propofol, at clinically relevant concentrations, can reduce inflammatory responses in LPS-induced ATII cells injury through downregulation of CD14 and TLR4 expression.
Collapse
Affiliation(s)
- L Ma
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | | | | | | |
Collapse
|
11
|
Hiramatsu M, Ninomiya H, Inamura K, Nomura K, Takeuchi K, Satoh Y, Okumura S, Nakagawa K, Yamori T, Matsuura M, Morikawa T, Ishikawa Y. Activation status of receptor tyrosine kinase downstream pathways in primary lung adenocarcinoma with reference of KRAS and EGFR mutations. Lung Cancer 2010; 70:94-102. [PMID: 20117855 DOI: 10.1016/j.lungcan.2010.01.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 12/02/2009] [Accepted: 01/05/2010] [Indexed: 01/05/2023]
Abstract
The activation status of signal transduction pathways involving receptor tyrosine kinases and its association with EGFR or KRAS mutations have been widely studied using cancer cell lines, although it is still uncertain in primary tumors. To study the activation status of main components of growth factor-induced pathways, phosphorylated Akt (pAkt), extracellular signal-regulated kinases 1 and 2 (pERK) and other downstream proteins were immunohistochemically examined using surgical samples of 193 primary lung adenocarcinomas. Also, thyroid transcription factor-1 (TTF-1) expression and mutation status of EGFR and KRAS were examined. Advanced tumor stages (p<0.001), negative TTF-1 expression (p<0.001) and Akt activation (p=0.015) were independent and significant poor prognostic markers. Akt activation related to advanced stage (p=0.021), invasiveness (p=0.004), and not to mutations. TTF-1 expression associated with never-smoker (p=0.013), pre- or minimally invasiveness (p<0.001) and EGFR mutations (p=0.017) as well as with pERK (p=0.039) expression. EGFR mutations did not correlated with pAkt and pERK expression, which was different from the results based on cultured cells, while KRAS mutations were solely and significantly linked to ERK activation (p=0.009). In lung adenocarcinoma, tumors with TTF-1 expression have distinct characteristics regarding mutations, signal protein activation and clinical issues. Moreover, this property was revealed to be important in outcome estimation at any tumor stage, whereas Akt activation is abnormally affected according to the tumor stage regardless of their cell origin. The signal proteins were differently related to mutation status from cultured cells.
Collapse
Affiliation(s)
- Miyako Hiramatsu
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research (JFCR), 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
McCourtie AS, Farivar AS, Woolley SM, Merry HE, Wolf PS, Mackinnon-Patterson B, Keech JC, Fitzsullivan E, Mulligan MS. Alveolar macrophage secretory products effect type 2 pneumocytes undergoing hypoxia-reoxygenation. Ann Thorac Surg 2009; 86:1774-9. [PMID: 19021974 DOI: 10.1016/j.athoracsur.2008.07.071] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 07/23/2008] [Accepted: 07/24/2008] [Indexed: 11/16/2022]
Abstract
BACKGROUND Activation of the alveolar macrophage is centrally important to the development of lung ischemia reperfusion injury. Alveolar macrophages and type 2 pneumocytes secrete a variety of proinflammatory mediators in response to oxidative stress. The manner in which they interact and how the macrophage may influence pneumocyte responses in lung ischemia reperfusion injury is unknown. Utilizing an in vitro model of hypoxia and reoxygenation, we sought to determine if the proinflammatory response of type 2 pneumocytes to oxidative stress would be amplified by alveolar macrophage secretory products. METHODS Cultured pneumocytes were exposed to control media or media from cultured macrophages exposed to hypoxia and reoxygenation. Pneumocytes were subsequently subjected to hypoxia and reoxygenation and assessed for both nuclear translocation of nuclear factor kappa B and inflammatory cytokine and chemokine secretion. To examine for any reciprocal interactions, we reversed the experiment, exposing macrophages to conditioned pneumocyte media. RESULTS In the presence of media from stimulated macrophages, production of proinflammatory mediators by type 2 pneumocytes was dramatically enhanced. In contrast, exposure of the macrophage to conditioned pneumocyte media had an inhibitory effect on macrophage responses subsequently exposed to hypoxia and reoxygenation. CONCLUSIONS The alveolar macrophage drives the development of lung reperfusion injury in part through amplification of the inflammatory response of type 2 pneumocytes subjected to hypoxia and reoxygenation.
Collapse
Affiliation(s)
- Anton S McCourtie
- Department of Surgery, Division of Cardiothoracic Surgery, University of Washington Medical Center, Seattle, Washington, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Abstract
BACKGROUND Although short-term findings after lung reperfusion have been extensively reported, in vivo animal studies have not described outcome beyond the immediate time period. Therefore, the authors evaluated lung injury 27 h after reperfusion. They also investigated whether attenuation of lung injury with the A3 adenosine receptor agonist MRS3558 was sustained beyond the immediate time period. METHODS In intact-chest, spontaneously breathing cats in which the left lower lung lobe was isolated and subjected to 2 h of ischemia and 3 h of reperfusion, MRS3558 was administered before reperfusion. Animals were killed 3 or 27 h after reperfusion. RESULTS When compared with 3 h of reperfusion, at 27 h the left lower lobe showed reduced apoptosis and no change in inflammation, but increased edema. Increased edema of the nonischemic right lung and hypoxemia were observed at 27 h after left lower lobe reperfusion. Increases in phosphorylated p38 levels were found at 3 h of reperfusion compared with control lung, with further increases at 27 h. The attenuation of injury observed with MRS3558 treatment at 3 h of reperfusion was sustained at 27 h. CONCLUSIONS Lung edema may worsen hours after the immediate postreperfusion period, even though lung apoptosis and inflammation are reduced or show no change, respectively. This was associated with further increases in phosphorylated p38 levels. The nonischemic lung may also be affected, suggesting a systemic response to reperfusion. In addition, early attenuation of injury is beneficial beyond the immediate period after reperfusion. Treatment aimed at inhibiting p38 activation, such as A3 receptor activation, should be further studied to explore its potential long-term beneficial effect.
Collapse
|
14
|
Waldow T, Witt W, Ulmer A, Janke A, Alexiou K, Matschke K. Preconditioning by inhaled nitric oxide prevents hyperoxic and ischemia/reperfusion injury in rat lungs. Pulm Pharmacol Ther 2008; 21:418-29. [PMID: 18453045 DOI: 10.1016/j.pupt.2007.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Since the generation of nitric oxide (NO) is an essential step in the trigger phase of ischemic preconditioning, short-term inhalation of NO before ischemia should ameliorate ischemia/reperfusion (I/R) injury of the lung. We tested this hypothesis in high oxygen (>99%) ventilated rats in order to additionally evaluate compatibility of NO and exposure to hyperoxia. Male adult Sprague-Dawley rats inhaled NO (15 ppm, 10 min) before the left lung hilum was clamped for 1 h, and the reperfusion phase was observed for 4 h (NO group). Animals in the I/R group underwent the same treatment, but without NO inhalation. A third group without I/R served as time-matched controls. Animals in the I/R group showed severe I/R injury in terms of arterial pO2 (apO2), which was reduced to 22% of surgical controls (SCs) at time point 30 min reperfusion, and increased endothelial permeability (Evans blue procedure). The pretreatment with NO attenuated these effects. The pO2 after 4 h reperfusion was still 3.0-fold higher in the NO group compared to I/R. In contrast, the I/R- and hyperoxia-induced invasion of leukocytes, as determined by measuring myeloperoxidase (MPO) activity, was not affected by NO. These data were correlated with the activity of major cellular signaling pathways by measuring the phosphorylation at activating and inhibitory sites of extracellular-signal regulated kinase (ERK), c-Jun N-terminal kinase (JNK), p38, protein kinase B (AKT), and glycogen synthase kinase 3beta (GSK-3beta), and by determination of cGMP in plasma and lung tissue. Inhalation of NO partly prevented the loss of activation by I/R and hyperoxic ventilation of ERK, JNK, and AKT, and it reduced the I/R-induced activation of GSK-3beta. The level of cGMP in plasma and lung tissue was increased in the NO group after 4 h reperfusion. In conclusion, application of inhaled NO in the preconditioning mode prevented I/R injury in the rat lung without interfering effects of hyperoxic ventilation. The effects of NO on cellular signaling pathways resemble mechanisms of ischemic preconditioning, but further studies have to evaluate the physiological relevance of these results.
Collapse
Affiliation(s)
- Thomas Waldow
- Clinic for Cardiac Surgery, University Hospital Dresden, Fetscherstr. 76, 01307 Dresden, Germany
| | | | | | | | | | | |
Collapse
|
15
|
Stress-activated protein kinase inhibition to ameliorate lung ischemia reperfusion injury. J Thorac Cardiovasc Surg 2008; 135:656-65. [DOI: 10.1016/j.jtcvs.2007.11.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Revised: 11/09/2007] [Accepted: 11/26/2007] [Indexed: 11/19/2022]
|
16
|
McCourtie AS, Farivar AS, Woolley SM, Merry HE, Wolf PS, Szabo C, Mulligan MS. Poly (ADP) ribose synthetase inhibition in alveolar macrophages undergoing hypoxia and reoxygenation. Exp Mol Pathol 2007; 84:141-4. [PMID: 18206870 DOI: 10.1016/j.yexmp.2007.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2007] [Revised: 11/25/2007] [Accepted: 11/26/2007] [Indexed: 11/30/2022]
Abstract
BACKGROUND Inhibition of the nuclear enzyme poly ribose synthetase (PARS) protects against in vivo lung ischemia reperfusion injury (LIRI). The effectiveness of intratracheal treatment suggests that PARS inhibition may primarily modulate alveolar macrophage (AM) activation. These studies attempted to characterize the effects of PARS on AM activation in response to oxidative stress. METHODS Primary cultures of AM were rendered hypoxic for 2 h and reoxygenated for up to 4 h. Cells were preincubated with INO-1001, a specific PARS inhibitor 1 h prior to hypoxia. Gel shift assays characterized nuclear factor kappa B (NFkappaB), and enzyme linked immunosorbent assay quantitated chemokine/cytokine protein secretion. RESULTS Hypoxia and reoxygenation resulted in an increase in the early nuclear translocation of NFkappaB, and an increase in the secretion of the cytokine tumor necrosis factor-alpha (TNF-alpha), chemokines macrophage inflammatory protein (MIP-1alpha), monocyte chemoattractant protein one (MCP-1) and cytokine induced neutrophil chemoattractant (CINC). Pretreatment of AM with INO-1001 decreased both the early translocation of NFkappaB and the production of TNF-alpha (p<0.05) and MIP-1alpha p=0.02, but did not affect CINC or MCP-1 production. CONCLUSIONS These findings indicate that PARS inhibition in the AM blunts their response to oxidative stress and may help explain the protective effects of intratracheal PARS inhibition in LIRI.
Collapse
Affiliation(s)
- Anton S McCourtie
- Department of Surgery, Division of Cardiothoracic Surgery, University of Washington Medical Center, Seattle, WA 98195, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Rivo J, Zeira E, Galun E, Einav S, Linden J, Matot I. Attenuation of reperfusion lung injury and apoptosis by A2A adenosine receptor activation is associated with modulation of Bcl-2 and Bax expression and activation of extracellular signal-regulated kinases. Shock 2007; 27:266-73. [PMID: 17304107 DOI: 10.1097/01.shk.0000235137.13152.44] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Adenosine receptors (AR) and extracellular signal-regulated kinases (ERK) have been implicated in tissue protection and apoptosis regulation during ischemia/reperfusion (I/R) injury. This study tests the hypothesis that reduction of reperfusion lung injury after A2A AR activation is associated with attenuation of apoptosis, modulation of ERK activation, and alterations in antiapoptotic and proapoptotic protein expression (Bcl-2 and Bax, respectively). Experiments were performed in intact-chest, spontaneously breathing cats in which the arterial branch of the left lower lung lobe was occluded for 2 h and reperfused for 3 h (I/R group). Animals were treated with the selective A2A AR agonist ATL313 given 5 min before reperfusion alone or in combination with the selective A2A AR antagonist ZM241385. Western blot analysis showed significant reduction in expression of Bcl-2 and increase in expression of Bax after reperfusion, compared with control lungs. Phosphorylated ERK1/2 levels were also increased after reperfusion. Compared with the I/R group, ATL313 markedly (P < 0.01) attenuated indices of injury and apoptosis including the percentage of injured alveoli, wet-dry weight ratio, myeloperoxidase activity, in situ terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling-positive cells, and caspase 3 activity and expression. Furthermore, compared with reperfused lungs, in ATL313-pretreated lungs, Western blot analysis demonstrated substantial ERK1/2 activation, increased expression of Bcl-2, and attenuated expression of Bax. The protective effects of ATL313 were blocked by pretreatment with ZM241385. In summary, the present study shows that in vivo activation of A2A AR confers protection against reperfusion lung injury. This protection is associated with decreased apoptosis and involves ERK1/2 activation and alterations in antiapoptotic Bcl-2 and proapoptotic Bax proteins.
Collapse
Affiliation(s)
- Julia Rivo
- Department of Anesthesiology and Critical Care Medicine, Hadassah University Medical Center, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
18
|
Matot I, Weiniger CF, Zeira E, Galun E, Joshi BV, Jacobson KA. A3 adenosine receptors and mitogen-activated protein kinases in lung injury following in vivo reperfusion. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2006; 10:R65. [PMID: 16623960 PMCID: PMC1550918 DOI: 10.1186/cc4893] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2006] [Revised: 03/06/2006] [Accepted: 03/15/2006] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Although activation of A3 adenosine receptors attenuates reperfusion lung injury and associated apoptosis, the signaling pathway that mediates this protection remains unclear. Adenosine agonists activate mitogen-activated protein kinases, and these kinases have been implicated in ischemia/reperfusion injury; the purpose of this study was therefore to determine whether A3 adenosine receptor stimulation with reperfusion modulates expression of the different mitogen-activated protein kinases. In addition, we compared the effect of the A3 adenosine agonist IB-MECA with the newly synthesized, highly selective A3 adenosine receptor agonist MRS3558 on injury in reperfused lung. METHOD Studies were performed in an in vivo spontaneously breathing cat model, in which the left lower lobe of the lung was isolated and subjected to 2 hours of ischemia and 3 hours of reperfusion. The selective A3 adenosine receptor agonists IB-MECA (0.05 mg/kg, 0.1 mg/kg, or 0.3 mg/kg) and MRS3558 (0.05 mg/kg or 0.1 mg/kg) were administered before reperfusion. RESULTS Both A3 adenosine receptor agonists administered before reperfusion markedly (P < 0.01) attenuated indices of injury and apoptosis, including the percentage of injured alveoli, wet/dry weight ratio, myeloperoxidase activity, TUNEL (in situ TdT-mediated dUTP nick end labeling)-positive cells, and caspase 3 activity and expression. The more pronounced effects at low doses were observed with MRS3558. Increases in phosphorylated c-Jun amino-terminal protein kinase (JNK), p38, and extracellular signal-regulated kinase (ERK)1/2 levels were observed by the end of reperfusion compared with controls. Pretreatment with the A3 agonists upregulated phosphorylated ERK1/2 levels but did not modify phosphorylated JNK and p38 levels. CONCLUSION The protective effects of A3 adenosine receptor activation are mediated in part through upregulation of phosphorylated ERK. Also, MRS3558 was found to be more potent than IB-MECA in attenuating reperfusion lung injury. The results suggest not only that enhancement of the ERK pathway may shift the balance between cell death and survival toward cell survival, but also that A3 agonists have potential as an effective therapy for ischemia/reperfusion-induced lung injury.
Collapse
Affiliation(s)
- Idit Matot
- Department of Anesthesiology & Critical Care Medicine, Hadassah University Medical Center, The Hebrew University, Jerusalem, Israel
| | - Carolyn F Weiniger
- Department of Anesthesiology & Critical Care Medicine, Hadassah University Medical Center, The Hebrew University, Jerusalem, Israel
| | - Evelyne Zeira
- Goldyne Savad Institute of Gene Therapy, Hadassah University Medical Center, The Hebrew University, Jerusalem, Israel
| | - Eithan Galun
- Goldyne Savad Institute of Gene Therapy, Hadassah University Medical Center, The Hebrew University, Jerusalem, Israel
| | - Bhalchandra V Joshi
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Lee SM, Lee CT, Kim YW, Han SK, Shim YS, Yoo CG. Hypoxia confers protection against apoptosis via PI3K/Akt and ERK pathways in lung cancer cells. Cancer Lett 2006; 242:231-8. [PMID: 16427189 DOI: 10.1016/j.canlet.2005.11.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2005] [Revised: 11/04/2005] [Accepted: 11/04/2005] [Indexed: 11/25/2022]
Abstract
Hypoxia confers protection against apoptosis in cancer cells, and this hypoxia-induced resistance to apoptosis has been suggested to be associated with genetic and adaptive changes. However, it is not clear whether survival signals, such as the PI3K/Akt and ERK pathways are involved. We investigated the roles of these pathways in hypoxia-induced protection against apoptosis in lung cancer cells. Treatment of cells with either ultraviolet (UV) or etoposide induced apoptosis time-dependently in A549 and NCI-H157 cells. However, though hypoxia alone neither induced apoptosis nor reduced cell survival, it suppressed the apoptosis induced by UV or etoposide. Moreover, hypoxia activated the PI3K/Akt and ERK pathways, and blocking the activation of either pathway reversed resistance to UV- and etoposide-induced apoptosis in response to hypoxia. These results suggest that hypoxia confers resistance to UV- or etoposide-mediated apoptosis in lung cancer cells via the activations of the PI3K/Akt and the ERK pathways.
Collapse
Affiliation(s)
- Sang-Min Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | | | | | | | | | | |
Collapse
|
20
|
Farivar AS, Mackinnon-Patterson BC, Barnes AD, McCourtie AS, Mulligan MS. Cyclosporine modulates the response to hypoxia-reoxygenation in pulmonary artery endothelial cells. Ann Thorac Surg 2005; 79:1010-6. [PMID: 15734424 DOI: 10.1016/j.athoracsur.2004.08.078] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/30/2004] [Indexed: 10/25/2022]
Abstract
BACKGROUND Depletion of macrophages, neutrophils, or lymphocytes confers only partial protection against experimental lung reperfusion injury, suggesting that inflammatory responses in other cell types contribute to tissue injury. Endothelial cell activation has previously been shown to be critical to the development of ischemia-reperfusion injury in other vascular beds. Furthermore, cyclosporine (CSA) reduces in vivo lung reperfusion injury through attenuated secretion of proinflammatory mediators. These studies determined whether pulmonary artery endothelial cells (PAEC), subjected to hypoxia and reoxygenation, promote inflammation and whether CSA afforded any modulation of that response. METHODS Isolated rat PAEC were subjected in vitro to 2 hours hypoxia followed by up to 4 hours reoxygenation. Cells were pretreated with CSA or a cremaphor vehicle. Differences in activation of signaling kinases and transcription factors were assessed, as was cytokine-chemokine protein secretion. RESULTS There was significant signaling kinase (extracellular signal regulated kinase [ERK 1/2]) activation by 15 minutes reoxygenation, which was temporally associated with marked activation of the transcription factors nuclear factor kappa B (NFkappaB) and early growth response one (EGR-1). At 4 hours reoxygenation there were significant increases in chemokine protein secretion. The CSA decreased ERK 1/2 phosphorylation and significantly attenuated transcription factor transactivation at 15 minutes reoxygenation. The CSA was found to be selective in reducing cytokine-chemokine elaboration at 4 hours reoxygenation. CONCLUSIONS Hypoxia-reoxygenation induces ERK 1/2 phosphorylation, as well as transactivation of the transcription factors NFkappaB and EGR-1 in PAEC. Cyclosporine selectively reduces proinflammatory mediator secretion, likely by transcriptional regulation through NFkappaB and EGR-1. This is the first demonstration of ERK 1/2 inhibition afforded by CSA.
Collapse
Affiliation(s)
- Alexander S Farivar
- University of Washington Medical Center, Department of Surgery, Division of Cardiothoracic Surgery, Seattle, Washington 98195, USA
| | | | | | | | | |
Collapse
|
21
|
Farivar AS, MacKinnon-Patterson B, Barnes AD, Mulligan MS. The effect of anti-inflammatory properties of mycophenolate mofetil on the development of lung reperfusion injury. J Heart Lung Transplant 2005; 24:2235-42. [PMID: 16364876 DOI: 10.1016/j.healun.2005.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2005] [Revised: 05/25/2005] [Accepted: 06/07/2005] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Lung ischemia-reperfusion injury (LIRI) is associated with an increased incidence of both primary graft failure and obliterative bronchiolitis. The immunosuppressant mycophenolate mofetil (MMF) has recently been shown to attenuate inflammatory injury in acute ischemia-reperfusion models via a mechanism that is presently unclear. These experiments studied the effects of MMF in a warm, in situ LIRI model, focusing on transcriptional regulation of pro-inflammatory mediators. METHODS Left lungs of rats were rendered ischemic for 90 minutes and reperfused for up to 4 hours. Treated animals received 10 mg/kg of intravenous MMF at 2 hours before ischemia. Left lung injury was quantitated by myeloperoxidase (MPO) content, permeability indices and bronchoalveolar lavage (BAL) inflammatory cell counts. Lungs were analyzed by electrophoretic mobility shift assay (EMSA) for transcription factor transactivation and by enzyme-linked immunoassay for BAL chemokine protein content. RESULTS MMF significantly reduced lung vascular permeability indices, MPO content and alveolar leukocyte counts at 4 hours of reperfusion. There was significant attenuation of activator protein 1 (AP-1) and early growth response 1 (EGR-1) transactivation, whereas nuclear factor-kappaB (NF-kappaB) was unaffected. Reductions in bronchoalveolar lavage monocyte chemoattractant protein 1 (MCP-1) and cytokine-induced neutrophil chemoattractant (CINC) protein content were found at 4 hours of reperfusion. CONCLUSIONS MMF limits lung ischemia-reperfusion-induced increases in vascular permeability and inflammatory cell sequestration in lung parenchyma and alveolar spaces. The protection is mediated at the transcriptional level via an attenuation of early EGR-1 and AP-1 transactivation, which was found to be associated with reduced late MCP-1 and CINC protein secretion. The use of MMF in concert with an agent that affects NF-kappaB activation may provide even further protection against lung reperfusion injury as multiple inflammatory pathways are inhibited.
Collapse
Affiliation(s)
- Alexander S Farivar
- Division of Thoracic Surgery, Department of Surgery, University of Washington Medical Center, Seattle, Washington 98195, USA
| | | | | | | |
Collapse
|