101
|
Frick AE, Orlitová M, Vanstapel A, Ordies S, Claes S, Schols D, Heigl T, Kaes J, Saez-Gimenez B, Vos R, Verleden GM, Vanaudenaerde B, Verleden SE, Van Raemdonck DE, Neyrinck AP. A novel experimental porcine model to assess the impact of differential pulmonary blood flow on ischemia-reperfusion injury after unilateral lung transplantation. Intensive Care Med Exp 2021; 9:4. [PMID: 33543363 PMCID: PMC7862464 DOI: 10.1186/s40635-021-00371-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 01/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Primary graft dysfunction (PGD) remains a major obstacle after lung transplantation. Ischemia-reperfusion injury is a known contributor to the development of PGD following lung transplantation. We developed a novel approach to assess the impact of increased pulmonary blood flow in a large porcine single-left lung transplantation model. MATERIALS Twelve porcine left lung transplants were divided in two groups (n = 6, in low- (LF) and high-flow (HF) group). Donor lungs were stored for 24 h on ice, followed by left lung transplantation. In the HF group, recipient animals were observed for 6 h after reperfusion with partially clamping right pulmonary artery to achieve a higher flow (target flow 40-60% of total cardiac output) to the transplanted lung compared to the LF group, where the right pulmonary artery was not clamped. RESULTS Survival at 6 h was 100% in both groups. Histological, functional and biological assessment did not significantly differ between both groups during the first 6 h of reperfusion. injury was also present in the right native lung and showed signs compatible with the pathophysiological hallmarks of ischemia-reperfusion injury. CONCLUSIONS Partial clamping native pulmonary artery in large animal lung transplantation setting to study the impact of low versus high pulmonary flow on the development of ischemia reperfusion is feasible. In our study, differential blood flow had no effect on IRI. However, our findings might impact future studies with extracorporeal devices and represent a specific intra-operative problem during bilateral sequential single-lung transplantation.
Collapse
Affiliation(s)
| | | | - Arno Vanstapel
- BREATHE, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), Leuven Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Sofie Ordies
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Sandra Claes
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Tobias Heigl
- BREATHE, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), Leuven Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Janne Kaes
- BREATHE, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), Leuven Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Berta Saez-Gimenez
- BREATHE, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), Leuven Lung Transplant Unit, KU Leuven, Leuven, Belgium.,Lung Transplant Unit, Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | - Robin Vos
- BREATHE, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), Leuven Lung Transplant Unit, KU Leuven, Leuven, Belgium.,Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Geert M Verleden
- BREATHE, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), Leuven Lung Transplant Unit, KU Leuven, Leuven, Belgium.,Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Bart Vanaudenaerde
- BREATHE, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), Leuven Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Stijn E Verleden
- BREATHE, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), Leuven Lung Transplant Unit, KU Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- BREATHE, Department of Chronic Diseases, Metabolism and Ageing (Chrometa), Leuven Lung Transplant Unit, KU Leuven, Leuven, Belgium.,Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Arne P Neyrinck
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.,Department of Anesthesiology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
102
|
Lan CC, Wu YK, Peng CK, Huang KL, Wu CP. Surfactant Attenuates Air Embolism-Induced Lung Injury by Suppressing NKCC1 Expression and NF-κB Activation. Inflammation 2021; 44:57-67. [PMID: 33089374 PMCID: PMC7796887 DOI: 10.1007/s10753-020-01266-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Excessive amounts of air can enter the lungs and cause air embolism (AE)-induced acute lung injury (ALI). Pulmonary AE can occur during diving, aviation, and iatrogenic invasive procedures. AE-induced lung injury presents with severe hypoxia, pulmonary hypertension, microvascular hyper-permeability, and severe inflammatory responses. Pulmonary AE-induced ALI is a serious complication resulting in significant morbidity and mortality. Surfactant is abundant in the lungs and its function is to lower surface tension. Earlier studies have explored the beneficial effects of surfactant in ALI; however, none have investigated the role of surfactant in pulmonary AE-induced ALI. Therefore, we conducted this study to determine the effects of surfactant in pulmonary AE-induced ALI. Isolated-perfused rat lungs were used as a model of pulmonary AE. The animals were divided into four groups (n = 6 per group): sham, air embolism (AE), AE + surfactant (0.5 mg/kg), and AE+ surfactant (1 mg/kg). Surfactant pretreatment was administered before the induction of pulmonary AE. Pulmonary AE was induced by the infusion of 0.7 cc air through a pulmonary artery catheter. After induction of air, pulmonary AE was presented with pulmonary edema, pulmonary microvascular hyper-permeability, and lung inflammation with neutrophilic sequestration. Activation of NF-κB was observed, along with increased expression of pro-inflammatory cytokines, and Na-K-Cl cotransporter isoform 1 (NKCC1). Surfactant suppressed the activation of NF-κB and decreased the expression of pro-inflammatory cytokines and NKCC1, thereby attenuating AE-induced lung injury. Therefore, AE-induced ALI presented with pulmonary edema, microvascular hyper-permeability, and lung inflammation. Surfactant suppressed the expressions of NF-κB, pro-inflammatory cytokines, and NKCC1, thereby attenuating AE-induced lung injury.
Collapse
Affiliation(s)
- Chou-Chin Lan
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu-Chi University, Hualien, Taiwan
| | - Yao-Kuang Wu
- Division of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu-Chi University, Hualien, Taiwan
| | - Chung-Kan Peng
- Division of Pulmonary Medicine, Tri-Service General Hospital, Taipei, Taiwan
- Institute of Undersea and Hyperbaric Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Division of Pulmonary Medicine, Tri-Service General Hospital, Taipei, Taiwan
- Institute of Undersea and Hyperbaric Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Pyng Wu
- Department of Critical Care Medicine, Landseed International Hospital, Tao-Yuan, Taiwan.
| |
Collapse
|
103
|
Calabrese DR, Aminian E, Mallavia B, Liu F, Cleary SJ, Aguilar OA, Wang P, Singer JP, Hays SR, Golden JA, Kukreja J, Dugger D, Nakamura M, Lanier LL, Looney MR, Greenland JR. Natural killer cells activated through NKG2D mediate lung ischemia-reperfusion injury. J Clin Invest 2021; 131:137047. [PMID: 33290276 PMCID: PMC7852842 DOI: 10.1172/jci137047] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022] Open
Abstract
Pulmonary ischemia-reperfusion injury (IRI) is a clinical syndrome of acute lung injury that occurs after lung transplantation or remote organ ischemia. IRI causes early mortality and has no effective therapies. While NK cells are innate lymphocytes capable of recognizing injured cells, their roles in acute lung injury are incompletely understood. Here, we demonstrated that NK cells were increased in frequency and cytotoxicity in 2 different IRI mouse models. We showed that NK cells trafficked to the lung tissue from peripheral reservoirs and were more mature within lung tissue. Acute lung ischemia-reperfusion injury was blunted in a NK cell-deficient mouse strain but restored with adoptive transfer of NK cells. Mechanistically, NK cell NKG2D receptor ligands were induced on lung endothelial and epithelial cells following IRI, and antibody-mediated NK cell depletion or NKG2D stress receptor blockade abrogated acute lung injury. In human lung tissue, NK cells were increased at sites of ischemia-reperfusion injury and activated NK cells were increased in prospectively collected human bronchoalveolar lavage in subjects with severe IRI. These data support a causal role for recipient peripheral NK cells in pulmonary IRI via NK cell NKG2D receptor ligation. Therapies targeting NK cells may hold promise in acute lung injury.
Collapse
Affiliation(s)
- Daniel R. Calabrese
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Emily Aminian
- Department of Medicine, University of California, San Francisco, California
| | - Benat Mallavia
- Department of Medicine, University of California, San Francisco, California
| | - Fengchun Liu
- Department of Medicine, University of California, San Francisco, California
| | - Simon J. Cleary
- Department of Medicine, University of California, San Francisco, California
| | - Oscar A. Aguilar
- Department of Microbiology and Immunology, University of California, San Francisco, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Ping Wang
- Department of Medicine, University of California, San Francisco, California
| | - Jonathan P. Singer
- Department of Medicine, University of California, San Francisco, California
| | - Steven R. Hays
- Department of Medicine, University of California, San Francisco, California
| | - Jeffrey A. Golden
- Department of Medicine, University of California, San Francisco, California
| | - Jasleen Kukreja
- Department of Surgery, University of California, San Francisco, California
| | - Daniel Dugger
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Mary Nakamura
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, California
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco, California
| | - John R. Greenland
- Department of Medicine, University of California, San Francisco, California
- Medical Service, Veterans Affairs Health Care System, San Francisco, California
| |
Collapse
|
104
|
Shimada M, Hoashi T, Imai K, Ichikawa H. Renal replacement therapy with a cytokine-adsorbing hemofilter after neonatal open-heart surgery. J Artif Organs 2021; 24:398-401. [PMID: 33416978 DOI: 10.1007/s10047-020-01241-5] [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] [Received: 11/18/2020] [Accepted: 12/17/2020] [Indexed: 11/28/2022]
Abstract
Two neonates with right atrial isomerism, single right ventricle, common atrioventricular valve regurgitation, and obstructive extra-cardiac total anomalous pulmonary venous connection underwent emergent total anomalous pulmonary venous connection repair combined with adjustment of pulmonary blood flow soon after birth. After the operation, both patients developed serious capillary leak syndrome, acute kidney injury, and lethal lactic acidosis with hemodynamic instability. Continuous renal replacement therapy was initiated with a cytokine-adsorbing hemofilter of polymethyl methacrylate membrane and a double lumen dialysis catheter inserted directly into the atrium. Elevated serum lactate levels were successfully decreased, and blood pressure was immediately increased.
Collapse
Affiliation(s)
- Masatoshi Shimada
- Department of Pediatric Cardiovascular Surgery, National Cerebral and Cardiovascular Center, 6-1, Kishibe-shimmachi, Suita, Osaka, 564-8565, Japan
| | - Takaya Hoashi
- Department of Pediatric Cardiovascular Surgery, National Cerebral and Cardiovascular Center, 6-1, Kishibe-shimmachi, Suita, Osaka, 564-8565, Japan.
| | - Kenta Imai
- Department of Pediatric Cardiovascular Surgery, National Cerebral and Cardiovascular Center, 6-1, Kishibe-shimmachi, Suita, Osaka, 564-8565, Japan
| | - Hajime Ichikawa
- Department of Pediatric Cardiovascular Surgery, National Cerebral and Cardiovascular Center, 6-1, Kishibe-shimmachi, Suita, Osaka, 564-8565, Japan
| |
Collapse
|
105
|
Ischemia-reperfusion Injury in the Transplanted Lung: A Literature Review. Transplant Direct 2021; 7:e652. [PMID: 33437867 PMCID: PMC7793349 DOI: 10.1097/txd.0000000000001104] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 02/07/2023] Open
Abstract
Lung ischemia-reperfusion injury (LIRI) and primary graft dysfunction are leading causes of morbidity and mortality among lung transplant recipients. Although extensive research endeavors have been undertaken, few preventative and therapeutic treatments have emerged for clinical use. Novel strategies are still needed to improve outcomes after lung transplantation. In this review, we discuss the underlying mechanisms of transplanted LIRI, potential modifiable targets, current practices, and areas of ongoing investigation to reduce LIRI and primary graft dysfunction in lung transplant recipients.
Collapse
|
106
|
Oshima Y, Otsuki A, Endo R, Nakasone M, Harada T, Takahashi S, Inagaki Y. The Effects of Volatile Anesthetics on Lung Ischemia-Reperfusion Injury: Basic to Clinical Studies. J Surg Res 2020; 260:325-344. [PMID: 33373852 DOI: 10.1016/j.jss.2020.11.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/01/2020] [Indexed: 02/08/2023]
Abstract
Case reports from as early as the 1970s have shown that intravenous injection of even a small dose of volatile anesthetics result in fatal lung injury. Direct contact between volatile anesthetics and pulmonary vasculature triggers chemical damage in the vessel walls. A wide variety of factors are involved in lung ischemia-reperfusion injury (LIRI), such as pulmonary endothelial cells, alveolar epithelial cells, alveolar macrophages, neutrophils, mast cells, platelets, proinflammatory cytokines, and surfactant. With a constellation of factors involved, the assessment of the protective effect of volatile anesthetics in LIRI is difficult. Multiple animal studies have reported that with regards to LIRI, sevoflurane demonstrates an anti-inflammatory effect in immunocompetent cells and an anti-apoptotic effect on lung tissue. Scattered studies have dismissed a protective effect of desflurane against LIRI. While a single-center randomized controlled trial (RCT) found that volatile anesthetics including desflurane demonstrated a lung-protective effect in thoracic surgery, a multicenter RCT did not demonstrate a lung-protective effect of desflurane. LIRI is common in lung transplantation. One study, although limited due to its small sample size, found that the use of volatile anesthetics in organ procurement surgery involving "death by neurologic criteria" donors did not improve lung graft survival. Future studies on the protective effect of volatile anesthetics against LIRI must examine not only the mechanism of the protective effect but also differences in the effects of different types of volatile anesthetics, their optimal dosage, and the appropriateness of their use in the event of marked alveolar capillary barrier damage.
Collapse
Affiliation(s)
- Yoshiaki Oshima
- Department of Anesthesiology, Yonago Medical Center, Yonago, Tottori, Japan.
| | - Akihiro Otsuki
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Ryo Endo
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Masato Nakasone
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Tomomi Harada
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Shunsaku Takahashi
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Yoshimi Inagaki
- Division of Anesthesiology and Critical Care Medicine, Department of Surgery, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| |
Collapse
|
107
|
Abstract
Cardiothoracic surgery posits an arrangement of large, significant hemodynamic, and physiologic alterations upon the human body, which predisposes a patient to develop pathology. The care of these patients in the postoperative realm requires an astute physician with deep understanding of the cardiopulmonary system, who is able to address subtle developing problems promptly, before the patient suffers further sequelae. In this review, we describe the presentation and management of an assortment of important complications which occur in the pulmonary system. In addition, we aim to shed better light upon how the physiology of a patient responds to the condition of cardiothoracic surgery.
Collapse
|
108
|
Pan X, Xu S, Zhou Z, Wang F, Mao L, Li H, Wu C, Wang J, Huang Y, Li D, Wang C, Pan J. Fibroblast growth factor-2 alleviates the capillary leakage and inflammation in sepsis. Mol Med 2020; 26:108. [PMID: 33187467 PMCID: PMC7662026 DOI: 10.1186/s10020-020-00221-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023] Open
Abstract
Background Acute lung injury (ALI), which is induced by numerous pathogenic factors, especially sepsis, can generate alveolar damage, pulmonary edema and vascular hyper-permeability ultimately leading to severe hypoxemia. Fibroblast growth factor-2 (FGF2) is an important member of the FGF family associated with endothelial cell migration and proliferation, and injury repairment. Here, we conducted this study aiming to evaluate the therapeutic effect of FGF2 in sepsis-induced ALI. Methods Recombinant FGF2 was abdominally injected into septic mice induced by cecal ligation and puncture (CLP), and then the inflammatory factors of lung tissue, vascular permeability and lung injury-related indicators based on protein levels and gene expression were detected. In vitro, human pulmonary microvascular endothelial cells (HPMEC) and mouse peritoneal macrophages (PMs) were challenged by lipopolysaccharides (LPS) with or without FGF2 administration in different groups, and then changes in inflammation indicators and cell permeability ability were tested. Results The results revealed that FGF2 treatment reduced inflammation response, attenuated pulmonary capillary leakage, alleviated lung injury and improved survival in septic mice. The endothelial injury and macrophages inflammation induced by LPS were inhibited by FGF2 administration via AKT/P38/NF-κB signaling pathways. Conclusion These findings indicated a therapeutic role of FGF2 in ALI through ameliorating capillary leakage and inflammation.
Collapse
Affiliation(s)
- Xiaojun Pan
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P. R. China
| | - Shunyao Xu
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P. R. China
| | - Zhen Zhou
- Department of Intensive Care Unit, Hangzhou Third Hospital, Hangzhou, 310000, Zhejiang, P. R. China
| | - Fen Wang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P. R. China
| | - Lingjie Mao
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P. R. China
| | - Hao Li
- Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P. R. China
| | - Caixia Wu
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P. R. China
| | - Junfeng Wang
- The Yiwu Affiliated Hospital of Wenzhou Medical University, Jinhua, 322000, Zhejiang, P. R. China
| | - Yueyue Huang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P. R. China
| | - Dequan Li
- Department of Traumatology Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P. R. China.
| | - Cong Wang
- Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P. R. China.
| | - Jingye Pan
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, P. R. China.
| |
Collapse
|
109
|
Creatine Supply Attenuates Ischemia-Reperfusion Injury in Lung Transplantation in Rats. Nutrients 2020; 12:nu12092765. [PMID: 32927837 PMCID: PMC7551831 DOI: 10.3390/nu12092765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) is one of the factors limiting the success of lung transplantation (LTx). IRI increases death risk after transplantation through innate immune system activation and inflammation induction. Some studies have shown that creatine (Cr) protects tissues from ischemic damage by its antioxidant action. We evaluated the effects of Cr supplementation on IRI after unilateral LTx in rats. Sixty-four rats were divided into four groups: water + 90 min of ischemia; Cr + 90 min of ischemia; water + 180 min of ischemia; and Cr + 180 min of ischemia. Donor animals received oral Cr supplementation (0.5 g/kg/day) or vehicle (water) for five days prior to LTx. The left lung was exposed to cold ischemia for 90 or 180 min, followed by reperfusion for 2 h. We evaluated the ventilatory mechanics and inflammatory responses of the graft. Cr-treated animals showed a significant decrease in exhaled nitric oxide levels and inflammatory cells in blood, bronchoalveolar lavage fluid and lung tissue. Moreover, edema, cell proliferation and apoptosis in lung parenchyma were reduced in Cr groups. Finally, TLR-4, IL-6 and CINC-1 levels were lower in Cr-treated animals. We concluded that Cr caused a significant decrease in the majority of inflammation parameters evaluated and had a protective effect on the IRI after LTx in rats.
Collapse
|
110
|
Lv N, Li X. Isoflurane suppresses lung ischemia-reperfusion injury by inactivating NF-κB and inhibiting cell apoptosis. Exp Ther Med 2020; 20:74. [PMID: 32968431 PMCID: PMC7500037 DOI: 10.3892/etm.2020.9202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 02/25/2020] [Indexed: 12/13/2022] Open
Abstract
Patients with lung ischemia-reperfusion injury (LIRI), involving cytokines, including interleukin (IL)-6 and IL-8, display poor clinical outcomes. Isoflurane displays protective effects against ischemia-reperfusion injury in numerous organs. In the present study, the effects of isoflurane on LIRI were investigated in vitro using a hypoxia-reoxygenation (HR) cell model. The mRNA expression levels of specific genes were analyzed by reverse transcription-quantitative PCR and protein expression levels were measured by ELISA and western blotting. Cell apoptosis and proliferation were assessed by flow cytometry and the Cell Counting Kit-8 assay, respectively. Isoflurane pretreatment decreased HR-induced IL-6 and IL-8 expression levels in A549 cells. Isoflurane pretreatment also inhibited HR-induced cell apoptosis and Bax expression, and reversed HR-induced downregulation of Bcl-2 expression. Moreover, isoflurane pretreatment decreased HR-induced NF-κB phosphorylated-p65 protein expression and NF-κB activation. Furthermore, HR-induced increases in malondialdehyde concentration and decreases in superoxide dismutase activity were reversed by isoflurane pretreatment. In conclusion, the results indicated that isoflurane suppressed LIRI by inhibiting the activation of NF-κB and the induction of cell apoptosis.
Collapse
Affiliation(s)
- Ning Lv
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin 300100, P.R. China
| | - Xiaoyun Li
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
111
|
Luna-Flores A, Olmos-Zuñiga JR, Jasso-Victoria R, Gaxiola-Gaxiola M, Aguirre-Pérez T, Ruiz V, García-Torrentera R, Silva-Martínez M, Zenteno E, Gutierrez-Ospina G, Santillan-Doherty P. Expression of Claudin-4 in Lung Ischemia-Reperfusion Injury in Experimental Lung Transplantation. J INVEST SURG 2020; 35:191-200. [PMID: 32900258 DOI: 10.1080/08941939.2020.1815253] [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: 10/23/2022]
Abstract
OBJECTIVE To assess the presence of CLDN4 in bronchoalveolar lavage fluid (BALF) and pulmonary tissue as an early indicator of LIRI and its relationship with changes in pulmonary physiology, edema formation and histology in an experimental porcine model of LTx with CIT of 50 min or 6 h. METHODS In 12 pigs, LIRI was produced by: group I (n = 6) LTx with 50 min of CIT (LTx-50 min-CIT); and group II (n = 6) LTx with 6 h of CIT (LTx-6h-CIT). The lung function, edema formation, macroscopic and microscopic changes were assessed. CLDN4 expression in BALF and pulmonary tissue were determined. RESULTS Both groups presented similar clinical, edema, and histological damage, as well as similar expression of CLDN4 in BALF and tissue (p > 0.05, RM-ANOVA). CONCLUSION CLDN4 expressed in BALF and the pulmonary tissue during the first 5 h within 72 h of the PGD window are not associated by the deterioration of lung function, edema and lung histological injury, in LTx with CIT 50 min or 6 h, CLDN4 does not seem to be a valuable indicator of LIRI.
Collapse
Affiliation(s)
- Antonia Luna-Flores
- Lung Transplantation Research Unit, Department of Surgical Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - J Raúl Olmos-Zuñiga
- Lung Transplantation Research Unit, Department of Surgical Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Rogelio Jasso-Victoria
- Department of Surgical Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Miguel Gaxiola-Gaxiola
- Department of Morphology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Teresa Aguirre-Pérez
- Bronchoscopy and Endoscopy Service, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Victor Ruiz
- Molecular Biology Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Rogelio García-Torrentera
- Respiratory Emergency Unit, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Mariana Silva-Martínez
- Lung Transplantation Research Unit, Department of Surgical Research, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Edgar Zenteno
- Department of Biochemistry, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Gabriel Gutierrez-Ospina
- Department of Cell Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Patricio Santillan-Doherty
- Medical Direction, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| |
Collapse
|
112
|
Zhao N, Li CC, Di B, Xu LL. Recent advances in the NEK7-licensed NLRP3 inflammasome activation: Mechanisms, role in diseases and related inhibitors. J Autoimmun 2020; 113:102515. [PMID: 32703754 DOI: 10.1016/j.jaut.2020.102515] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022]
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor containing pyrin domain 3 (NLRP3) inflammasome is a high-molecular-weight complex mediated by the activation of pattern-recognition receptors (PRRs) seed in innate immunity. Once NLRP3 is activated, the following recruitment of the adapter apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD) (ASC) and procaspase-1 would be initiated. Cleavage of procaspase-1 into active caspase-1 then leads to the maturation of the precursor forms of interleukin (IL)-1β and IL-18 into biologically active IL-1β and IL-18. The activation of NLRP3 inflammasome is thought to be tightly associated with a regulator never in mitosis A (NIMA)-related kinase 7 (NEK7), apart from other signaling events such as K+ efflux and reactive oxygen species (ROS). Plus, the NLRP3 inflammasome has been linked to various metabolic disorders, chronic inflammation and other diseases. In this review, we firstly describe the cellular/molecular mechanisms of the NEK7-licensed NLRP3 inflammasome activation. Then we detail the potential inhibitors that can selectively and effectively modulate either the NEK7-NLRP3 complex itself or the related molecular/cellular events. Finally, we describe some inhibitors as promising therapeutic strategies for diverse diseases driven by NLRP3 inflammasome.
Collapse
Affiliation(s)
- Ni Zhao
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Cui-Cui Li
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Bin Di
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China.
| | - Li-Li Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
113
|
Cytochrome P450 Epoxygenase 2J2 Protects Against Lung Ischemia/Reperfusion Injury by Activating the P13K/Akt/GSK-3-β/NF-kB Signaling Pathway During Deep Hypothermic Low Flow in Mice. J Surg Res 2020; 253:8-17. [PMID: 32305498 DOI: 10.1016/j.jss.2019.12.052] [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] [Received: 03/15/2019] [Revised: 12/03/2019] [Accepted: 12/27/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Cytochrome P450 epoxygenase 2J2 (CYP2J2) metabolizes arachidonic acid to epoxyeicosatrienoic acids, which exert anti-inflammatory effects and alleviate oxidative stress in the cardiovascular system. Our previous work revealed that CYP2J2 is expressed in pulmonary artery endothelial cells. It was therefore hypothesized that CYP2J2 overexpression may prevent lung ischemia/reperfusion injury (LIRI) in 3-week-old C57BL/6 mice during deep hypothermic low flow (DHLF). This study aimed to establish whether CYP2J2 protects against LIRI and the mechanisms of CYP2J2 overexpression during DHLF in mice. The aim of this study was to explore the effects of DHLF on lung tissue in mice and to find out the regularity of this process, so as to provide theoretical data for lung tissue protection in children undergoing this process in clinic. METHODS A 3-week-old C57BL/6 mouse model was used to mimic LIRI conditions during DHLF by clamping the left pulmonary artery and left main bronchus for 120 min, followed by reperfusion for 2 h. The body temperature of the mice was maintained between 18°C and 19°C to induce DHLF. RESULTS During DHLF, lung ischemia/reperfusion increased the left lung wet/dry weight, the left lung weight/body weight ratio, the protein concentration in bronchoalveolar lavage fluid, and the concentration of proinflammatory mediators in the lungs, including interleukin (IL)-1, IL-8, and necrosis factor (NF)-α, and decreased the concentration of the anti-inflammatory mediator IL-10. Furthermore, activation of NF-κB p65 and degradation of IKBα were remarkably increased in lung tissues after ischemia/reperfusion. The CYP2J2 overexpression group showed the opposite results (P < 0.05), and p-Akt1 and p-GSK-3β expression were significantly higher in the CYP2J2 overexpression group (P < 0.05). Moreover, the changes in IL-1, IL-8, tumor necrosis factor-α, IL-10, p-Akt1, p-GSK-3β, NF-κB p65, and IKBα were reversed in the Akt1 gene heterozygous knockout group, and lung damage was significantly higher in the Akt1 gene heterozygous knockout group than in the CYP2J2 overexpression group. CYP2J2 overexpression can protect against LIRI, whereas Akt1 gene heterozygous knockout in mice can abolish this protective effect. CONCLUSIONS CYP2J2 overexpression can protect against LIRI by activating the P13K/Akt/GSK-3β/NF-kB signaling pathway during DHLF. Thus, changing CYP2J2 expression can be a novel strategy for the prevention and treatment of LIRI during DHLF.
Collapse
|
114
|
Jin Y, Sun M, Lv X, Wang X, Jiang G, Chen C, Wen Z. Extracellular histones play a pathogenic role in primary graft dysfunction after human lung transplantation. RSC Adv 2020; 10:12485-12491. [PMID: 35497627 PMCID: PMC9051052 DOI: 10.1039/d0ra00127a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/22/2020] [Indexed: 11/21/2022] Open
Abstract
Primary graft dysfunction (PGD) causes early mortality and late graft failure after lung transplantation. The mechanisms of PGD are not fully understood but ischemia/reperfusion (I/R) injury may be involved. Extracellular histones have recently been identified as major contributors to I/R injury. Hence, we investigated whether extracellular histones are associated with PGD after lung transplantation. In total, 65 lung transplant patients were enrolled into this study. Blood samples were collected from patients before and serially after transplantation (24 h, 48 h, and 72 h) and measured for extracellular histones, myeloperoxidase (MPO), lactate dehydrogenase (LDH), and multiple cytokines. Besides, the patients' sera were cultured with human pulmonary artery endothelial cells (HPAEC) and human monocyte cell line (THP1) cells, respectively, and cellular viability and cytokine production were determined. Heparin or anti-histone antibody were used to study the effects of histone-neutralized interventions. The results showed that extracellular histones increased markedly after lung transplantation, peaked by 24 h and tended to decrease thereafter, but still retained high levels up to 72 h. Extracellular histones were more abundant in patients with PGD (n = 8) than patients without PGD (n = 57) and linearly correlated with MPO, LDH, and most detected cytokines. Ex vivo studies showed that the patients' sera collected within 24 h after transplantation were very damaging to HPAEC cells and promoted cytokine production in cultured THP1 cells, which could be largely prevented by heparin or anti-histone antibodies. These data suggested a pathogenic role for extracellular histones in PGD after lung transplantation. Targeting extracellular histones may serve as a preventive and therapeutic strategy for PGD following lung transplantation.
Collapse
Affiliation(s)
- Yang Jin
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| | - Meng Sun
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| | - Xingan Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine Pittsburgh PA 15213 USA
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine Shanghai 200433 China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine Shanghai 200433 China
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| |
Collapse
|
115
|
Shade BC, Dudley S, McCabe T, Gray-DeAngelis K. Use of pulmonoplegia and delivery system during recipient surgery in lung transplantation. Perfusion 2020; 35:587-590. [PMID: 32081092 DOI: 10.1177/0267659120906078] [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: 11/16/2022]
Abstract
Lung transplantation in the United States has steadily grown over the last decade. Major attention has been with the understanding of lung ischemia-reperfusion injury and how it relates to primary graft dysfunction. In 2015, our institution implemented the use of a pulmonoplegia solution during recipient surgery of lung transplantation. A unique circuit utilizing the heart lung machine is used to deliver the pulmonoplegia solution. This system is considered to be a key contributing factor to the success of our lung transplant program.
Collapse
Affiliation(s)
- Brandon C Shade
- Department of Perfusion Services, Heart and Vascular Center, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Dudley
- Department of Perfusion Services, Heart and Vascular Center, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Tara McCabe
- Department of Perfusion Services, Heart and Vascular Center, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Kathryn Gray-DeAngelis
- Department of Perfusion Services, Heart and Vascular Center, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
116
|
Increased Arginase Expression and Decreased Nitric Oxide in Pig Donor Lungs after Normothermic Ex Vivo Lung Perfusion. Biomolecules 2020; 10:biom10020300. [PMID: 32075026 PMCID: PMC7072555 DOI: 10.3390/biom10020300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/30/2020] [Accepted: 02/11/2020] [Indexed: 01/12/2023] Open
Abstract
An established pig lung transplantation model was used to study the effects of cold ischemia time, normothermic acellular ex vivo lung perfusion (EVLP) and reperfusion after lung transplantation on l-arginine/NO metabolism in lung tissue. Lung tissue homogenates were analyzed for NO metabolite (NOx) concentrations by chemiluminescent NO-analyzer technique, and l-arginine, l-ornithine, l-citrulline and asymmetric dimethylarginine (ADMA) quantified using liquid chromatography-mass spectrometry (LC-MS/MS). The expression of arginase and nitric oxide synthase (NOS) isoforms in lung was measured by real-time polymerase chain reaction. EVLP preservation resulted in a significant decrease in concentrations of NOx and l-citrulline, both products of NOS, at the end of EVLP and after reperfusion following transplantation, compared to control, respectively. The ratio of l-ornithine over l-citrulline, a marker of the balance between l-arginine metabolizing enzymes, was increased in the EVLP group prior to reperfusion. The expression of both arginase isoforms was increased from baseline 1 h post reperfusion in EVLP but not in the no-EVLP group. These data suggest that EVLP results in a shift of the l-arginine balance towards arginase, leading to NO deficiency in the lung. The arginase/NOS balance may, therefore, represent a therapeutic target to improve lung quality during EVLP and, subsequently, transplant outcomes.
Collapse
|
117
|
Wong A, Zamel R, Yeung J, Bader GD, Dos Santos CC, Bai X, Wang Y, Keshavjee S, Liu M. Potential therapeutic targets for lung repair during human ex vivo lung perfusion. Eur Respir J 2020; 55:13993003.02222-2019. [DOI: 10.1183/13993003.02222-2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 01/15/2020] [Indexed: 12/20/2022]
Abstract
IntroductionThe ex vivo lung perfusion (EVLP) technique has been developed to assess the function of marginal donor lungs and has significantly increased donor lung utilisation. EVLP has also been explored as a platform for donor lung repair through injury-specific treatments such as antibiotics or fibrinolytics. We hypothesised that actively expressed pathways shared between transplantation and EVLP may reveal common mechanisms of injury and potential therapeutic targets for lung repair prior to transplantation.Materials and methodsRetrospective transcriptomics analyses were performed with peripheral tissue biopsies from “donation after brain death” lungs, with 46 pre-/post-transplant pairs and 49 pre-/post-EVLP pairs. Pathway analysis was used to identify and compare the responses of donor lungs to transplantation and to EVLP.Results22 pathways were enriched predominantly in transplantation, including upregulation of lymphocyte activation and cell death and downregulation of metabolism. Eight pathways were enriched predominantly in EVLP, including downregulation of leukocyte functions and upregulation of vascular processes. 27 pathways were commonly enriched, including activation of innate inflammation, cell death, heat stress and downregulation of metabolism and protein synthesis. Of the inflammatory clusters, Toll-like receptor/innate immune signal transduction adaptor signalling had the greatest number of nodes and was central to inflammation. These mechanisms have been previously speculated as major mechanisms of acute lung injury in animal models.ConclusionEVLP and transplantation share common molecular features of injury including innate inflammation and cell death. Blocking these pathways during EVLP may allow for lung repair prior to transplantation.
Collapse
|
118
|
Chan PG, Kumar A, Subramaniam K, Sanchez PG. Ex Vivo Lung Perfusion: A Review of Research and Clinical Practices. Semin Cardiothorac Vasc Anesth 2020; 24:34-44. [DOI: 10.1177/1089253220905147] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
End-stage lung disease is ultimately treated with lung transplantation. However, there is a paucity of organs with an increasing number of patients being diagnosed with end-stage lung disease. Ex vivo lung perfusion has emerged as a potential tool to assess the quality and to recondition marginal donor lungs prior to transplantation with the goal of increasing the donor pool. This technology has shown promise with similar results compared with the conventional technique of cold static preservation in terms of primary graft dysfunction and overall outcomes. This review provides an update on the results and uses of this technology. The review will also summarize clinical studies and techniques in reconditioning and assessing lungs on ex vivo lung perfusion. Last, we discuss how this technology can be applied to fields outside of transplantation such as thoracic oncology and bioengineering.
Collapse
|
119
|
Charles EJ, Chordia MD, Zhao Y, Zhang Y, Mehaffey JH, Glover DK, Dimastromatteo J, Chancellor WZ, Sharma AK, Kron IL, Pan D, Laubach VE. SPECT imaging of lung ischemia-reperfusion injury using [ 99mTc]cFLFLF for molecular targeting of formyl peptide receptor 1. Am J Physiol Lung Cell Mol Physiol 2020; 318:L304-L313. [PMID: 31800262 PMCID: PMC7052676 DOI: 10.1152/ajplung.00220.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Primary graft dysfunction after lung transplantation, a consequence of ischemia-reperfusion injury (IRI), is a major cause of morbidity and mortality. IRI involves acute inflammation and innate immune cell activation, leading to rapid infiltration of neutrophils. Formyl peptide receptor 1 (FPR1) expressed by phagocytic leukocytes plays an important role in neutrophil function. The cell surface expression of FPR1 is rapidly and robustly upregulated on neutrophils in response to inflammatory stimuli. Thus, we hypothesized that use of [99mTc]cFLFLF, a selective FPR1 peptide ligand, would permit in vivo neutrophil labeling and noninvasive imaging of IRI using single-photon emission computed tomography (SPECT). A murine model of left lung IRI was utilized. Lung function, neutrophil infiltration, and SPECT imaging were assessed after 1 h of ischemia and 2, 12, or 24 h of reperfusion. [99mTc]cFLFLF was injected 2 h before SPECT. Signal intensity by SPECT and total probe uptake by gamma counts were 3.9- and 2.3-fold higher, respectively, in left lungs after ischemia and 2 h of reperfusion versus sham. These values significantly decreased with longer reperfusion times, correlating with resolution of IRI as shown by improved lung function and decreased neutrophil infiltration. SPECT results were confirmed using Cy7-cFLFLF-based fluorescence imaging of lungs. Immunofluorescence microscopy confirmed cFLFLF binding primarily to activated neutrophils. These results demonstrate that [99mTc]cFLFLF SPECT enables noninvasive detection of lung IRI and permits monitoring of resolution of injury over time. Clinical application of [99mTc]cFLFLF SPECT may permit diagnosis of lung IRI for timely intervention to improve outcomes after transplantation.
Collapse
Affiliation(s)
- Eric J. Charles
- 1Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Mahendra D. Chordia
- 2Department of Radiology and Medical Imaging, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Yunge Zhao
- 1Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Yi Zhang
- 5Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - J. Hunter Mehaffey
- 1Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - David K. Glover
- 3Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Julien Dimastromatteo
- 4Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia
| | - W. Zachary Chancellor
- 1Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Ashish K. Sharma
- 1Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Irving L. Kron
- 1Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Dongfeng Pan
- 2Department of Radiology and Medical Imaging, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Victor E. Laubach
- 1Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| |
Collapse
|
120
|
Moskowitzova K, Orfany A, Liu K, Ramirez-Barbieri G, Thedsanamoorthy JK, Yao R, Guariento A, Doulamis IP, Blitzer D, Shin B, Snay ER, Inkster JAH, Iken K, Packard AB, Cowan DB, Visner GA, Del Nido PJ, McCully JD. Mitochondrial transplantation enhances murine lung viability and recovery after ischemia-reperfusion injury. Am J Physiol Lung Cell Mol Physiol 2019; 318:L78-L88. [PMID: 31693391 PMCID: PMC6985877 DOI: 10.1152/ajplung.00221.2019] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The most common cause of acute lung injury is ischemia-reperfusion injury (IRI), during which mitochondrial damage occurs. We have previously demonstrated that mitochondrial transplantation is an efficacious therapy to replace or augment mitochondria damaged by IRI, allowing for enhanced muscle viability and function in cardiac tissue. Here, we investigate the efficacy of mitochondrial transplantation in a murine lung IRI model using male C57BL/6J mice. Transient ischemia was induced by applying a microvascular clamp on the left hilum for 2 h. Upon reperfusion mice received either vehicle or vehicle-containing mitochondria either by vascular delivery (Mito V) through the pulmonary artery or by aerosol delivery (Mito Neb) via the trachea (nebulization). Sham control mice underwent thoracotomy without hilar clamping and were ventilated for 2 h before returning to the cage. After 24 h recovery, lung mechanics were assessed and lungs were collected for analysis. Our results demonstrated that at 24 h of reperfusion, dynamic compliance and inspiratory capacity were significantly increased and resistance, tissue damping, elastance, and peak inspiratory pressure (Mito V only) were significantly decreased (P < 0.05) in Mito groups as compared with their respective vehicle groups. Neutrophil infiltration, interstitial edema, and apoptosis were significantly decreased (P < 0.05) in Mito groups as compared with vehicles. No significant differences in cytokines and chemokines between groups were shown. All lung mechanics results in Mito groups except peak inspiratory pressure in Mito Neb showed no significant differences (P > 0.05) as compared with Sham. These results conclude that mitochondrial transplantation by vascular delivery or nebulization improves lung mechanics and decreases lung tissue injury.
Collapse
Affiliation(s)
- Kamila Moskowitzova
- Department of Cardiac Surgery, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Arzoo Orfany
- Department of Cardiac Surgery, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Kaifeng Liu
- Department of Pulmonary and Respiratory Diseases, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Giovanna Ramirez-Barbieri
- Department of Cardiac Surgery, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Jerusha K Thedsanamoorthy
- Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, Massachusetts
| | - Rouan Yao
- Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, Massachusetts
| | - Alvise Guariento
- Department of Cardiac Surgery, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Ilias P Doulamis
- Department of Cardiac Surgery, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - David Blitzer
- Department of Cardiac Surgery, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Borami Shin
- Department of Cardiac Surgery, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Erin R Snay
- Department of Radiology, Division of Nuclear Medicine and Molecular imaging, Boston Children's Hospital, Boston, Massachusetts
| | - James A H Inkster
- Department of Radiology, Division of Nuclear Medicine and Molecular imaging, Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Khadija Iken
- Department of Pulmonary and Respiratory Diseases, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Alan B Packard
- Department of Radiology, Division of Nuclear Medicine and Molecular imaging, Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Douglas B Cowan
- Department of Anesthesiology, Critical Care and Pain Medicine, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Gary A Visner
- Department of Pulmonary and Respiratory Diseases, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Pedro J Del Nido
- Department of Cardiac Surgery, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - James D McCully
- Department of Cardiac Surgery, Harvard Medical School, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
121
|
Xu G, Wang X, Xiong Y, Ma X, Qu L. Effect of sevoflurane pretreatment in relieving liver ischemia/reperfusion-induced pulmonary and hepatic injury. Acta Cir Bras 2019; 34:e201900805. [PMID: 31618405 PMCID: PMC6799973 DOI: 10.1590/s0102-865020190080000005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 07/18/2019] [Indexed: 02/18/2023] Open
Abstract
Purpose To investigate the effect of sevoflurane preconditioning on
ischemia/reperfusion (I/R)-induced pulmonary/hepatic injury Methods Fifty-one Wistar rats were randomly grouped into sham, I/R, and sevoflurane
groups. After reperfusion, the structural change of the lung was measured by
Smith score, the wet and dry weights (W/D) were determined, malondialdehyde
(MDA) myeloperoxidase (MPO) content was determined colorimetrically and by
fluorescence, respectively, and matrix metalloprotein-9 (MMP-9) mRNA was
quantified by RT-PCR. Biopsy and morphological analyses were performed on
liver tissue, activities of aspartate aminotransferase (AST) and alanine
aminotransferase (ALT) were determined, and tumor necrosis factor-alpha
(TNF-α) level was determined. Results The sham group showed no changes in tissue structure. Structural lesions in
the sevoflurane and I/R groups were mild and severe, respectively. Smith
score, W/D, MDA, MPO, and MMP mRNA showed the same trend, and were increased
in the I/R group and recovered in the sevoflurane group, compared with the
sham group (both P<0.05). AST and ALT were significantly increased
compared to the sham group (AST: 655±52.06 vs . 29±9.30
U/L; ALT: 693±75.56 vs . 37±6.71 U/L; P<0.05). In the
sevoflurane group, AST and ALT levels were significantly decreased
(464±47.71 and 516±78.84 U/L; P<0.001). TNF-α presented similar
results. Conclusion The protection of lung and liver by sevoflurane may be mediated by inhibited
leukocyte recruitment and MMP-9 secretion.
Collapse
Affiliation(s)
- Guiping Xu
- Professor, Department of Anesthesia, Xinjiang Uygur Municipal People's Hospital, Urumqi 830001, China. Conception, design, intellectual and scientific content of the study; manuscript writing; critical revision; final approval
| | - Xiaoli Wang
- MD, Department of Anesthesia, Xinjiang Uygur Municipal People's Hospital, Urumqi 830001, China. Acquisition and analysis of data, manuscript writing
| | - Yuxiang Xiong
- MD, Department of Anesthesia, Xinjiang Uygur Municipal People's Hospital, Urumqi 830001, China. Acquisition and analysis of data
| | - Xueping Ma
- MD, Department of Anesthesia, Xinjiang Uygur Municipal People's Hospital, Urumqi 830001, China. Acquisition and analysis of data
| | - Li Qu
- MD, Department of Anesthesia, Xinjiang Uygur Municipal People's Hospital, Urumqi 830001, China. Acquisition and analysis of data
| |
Collapse
|
122
|
Goyal S, Dashey S, Zlocha V, HannaJumma S. The successful use of extra-corporeal membrane oxygenation as rescue therapy for unilateral pulmonary edema following minimally invasive mitral valve surgery. Perfusion 2019; 35:356-359. [DOI: 10.1177/0267659119874696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Unilateral right pulmonary edema has been reported as a potential life-threatening complication after minimally invasive mitral valve surgery. Nearly 2% of these cases in the immediate postoperative period have been reported to require extra-corporeal membrane oxygenation support as a rescue therapy for severe hypoxia. The exact pathophysiology of this condition remains unclear, but has been assumed to be related to ischemia–reperfusion injury and re-expansion pulmonary edema. We present in this report the successful use of extra-corporeal membrane oxygenation to manage two cases of severe hypoxia and multiorgan dysfunction secondary to unilateral right pulmonary edema.
Collapse
Affiliation(s)
- Shraddha Goyal
- Heartlink ECMO Centre, Glenfield Hospital, Leicester, UK
| | - Susan Dashey
- Heartlink ECMO Centre, Glenfield Hospital, Leicester, UK
| | - Viktor Zlocha
- Heartlink ECMO Centre, Glenfield Hospital, Leicester, UK
| | | |
Collapse
|
123
|
Transplant Suitability of Rejected Human Donor Lungs With Prolonged Cold Ischemia Time in Low-Flow Acellular and High-Flow Cellular Ex Vivo Lung Perfusion Systems. Transplantation 2019; 103:1799-1808. [DOI: 10.1097/tp.0000000000002667] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
124
|
Hashimoto K, Yamane M, Sugimoto S, Hirano Y, Kurosaki T, Otani S, Miyoshi K, Ohara T, Okazaki M, Yoshimura T, Oto T, Matsukawa A, Toyooka S. Negative impact of recipient SPRED2 deficiency on transplanted lung in a mouse model. Transpl Immunol 2019; 57:101242. [PMID: 31446154 DOI: 10.1016/j.trim.2019.101242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/10/2019] [Accepted: 08/21/2019] [Indexed: 12/25/2022]
Abstract
Ischemia-reperfusion injury (IRI) after lung transplantation mainly contributes to the development of primary graft dysfunction. The Sprouty-related EVH1-domain-containing (SPRED) protein family inhibits the mitogen activated protein kinase/extracellular-signal-regulated kinase (MAPK/ERK) pathway. Our study was aimed at examining the role of SPRED2 in IRI in mice that received orthotopic lung transplantation. Syngeneic mouse lung transplantation was performed in wild-type C57BL/6 J (WT) mice and Spred2 knockout (Spred2-/-) mice on the C57BL/6 J background from the WT donor. Four hours after reperfusion, blood gas analysis was performed, and lung grafts were sacrificed and analyzed. By using arterial oxygen tension measurements and histological evaluation using Lung Injury Score, we revealed more severe IRI in the grafts transplanted to Spred2-/- recipients, which manifested as exacerbated airway epithelial cell damage, interstitial edema with hemorrhage and neutrophil infiltration. Intragraft ERK1/2 activation and expression levels of proinflammatory cytokines and chemokines in Spred2-/- recipients were higher than those in WT recipients. SPRED2 plays an important role in protecting the lungs from IRI in lung transplantation recipients. We suggest that focused treatments suppressing the activity of the MAPK/ERK pathway in transplantation recipients could be the potential therapeutic option for the prevention of lung IRI.
Collapse
Affiliation(s)
- Kohei Hashimoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masaomi Yamane
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Seiichiro Sugimoto
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yutaka Hirano
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takeshi Kurosaki
- Organ Transplant Center, Okayama University Hospital, Okayama, Japan
| | - Shinji Otani
- Organ Transplant Center, Okayama University Hospital, Okayama, Japan
| | - Kentaroh Miyoshi
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshiaki Ohara
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mikio Okazaki
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takahiro Oto
- Organ Transplant Center, Okayama University Hospital, Okayama, Japan
| | - Akihiro Matsukawa
- Department of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
125
|
Mesenchymal stem cell-derived extracellular vesicles improve the molecular phenotype of isolated rat lungs during ischemia/reperfusion injury. J Heart Lung Transplant 2019; 38:1306-1316. [PMID: 31530458 DOI: 10.1016/j.healun.2019.08.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/14/2019] [Accepted: 08/20/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Lung ischemia/reperfusion (IR) injury contributes to the development of severe complications in patients undergoing transplantation. Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) exert beneficial actions comparable to those of MSCs without the risks of the cell-based strategy. This research investigated EV effects during IR injury in isolated rat lungs. METHODS An established model of 180-minutes ex vivo lung perfusion (EVLP) was used. At 60 minutes EVs (n = 5) or saline (n = 5) were administered. Parallel experiments used labeled EVs to determine EV biodistribution (n = 4). Perfusate samples were collected to perform gas analysis and to assess the concentration of nitric oxide (NO), hyaluronan (HA), inflammatory mediators, and leukocytes. Lung biopsies were taken at 180 minutes to evaluate HA, adenosine triphosphate (ATP), gene expression, and histology. RESULTS Compared with untreated lungs, EV-treated organs showed decreased vascular resistance and a rise of perfusate NO metabolites. EVs prevented the reduction in pulmonary ATP caused by IR. Increased medium-high-molecular-weight HA was detected in the perfusate and in the lung tissue of the IR + EV group. Significant differences in cell count on perfusate and tissue samples, together with induction of transcription and synthesis of chemokines, suggested EV-dependent modulation of leukocyte recruitment. EVs upregulated genes involved in the resolution of inflammation and oxidative stress. Biodistribution analysis showed that EVs were retained in the lung tissue and internalized within pulmonary cells. CONCLUSIONS This study shows multiple novel EV influences on pulmonary energetics, tissue integrity, and gene expression during IR. The use of cell-free therapies during EVLP could constitute a valuable strategy for reconditioning and repair of injured lungs before transplantation.
Collapse
|
126
|
Alda-1 Prevents Pulmonary Epithelial Barrier Dysfunction following Severe Hemorrhagic Shock through Clearance of Reactive Aldehydes. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2476252. [PMID: 31467875 PMCID: PMC6699483 DOI: 10.1155/2019/2476252] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 07/09/2019] [Indexed: 01/11/2023]
Abstract
Severe hemorrhagic shock and resuscitation (HS/R) can lead to lung injury, resulting in respiratory insufficiency. We investigated whether treatment with Alda-1, an ALDH2 activator, decreased lung injury induced by severe HS/R in a rat model. Male Sprague-Dawley rats were randomized into three groups, hemorrhagic shock + placebo, hemorrhagic shock + Alda-1, and sham. All animals were heparinized, and then 50% of the total calculated blood volume was collected over 60 minutes. After 40 minutes of hemorrhagic shock, animals were reinfused with the shed blood over 40 minutes and then observed for an additional 2 hours. Concentrations of 4-HNE, TNF-α, IL-6, and ALDH2 activity were detected; lung injury and lung wet-to-dry weight ratios were assessed. Expression of occludin and ZO-1 proteins in lung tissues was also determined. At 2 hours after resuscitation, lung injury was significantly reduced and the wet-to-dry weight ratio was notably decreased in the Alda-1 group compared with placebo (P<0.05). Alda-1 treatment also significantly increased the activity of ALDH2 and decreased the levels of toxic 4-HNE (P<0.05). In the Alda-1 group, IL-6 and TNF-α were dramatically decreased compared with placebo-treated animals (P<0.05). Expression of occludin and ZO-1 proteins was significantly decreased in the placebo group compared with the Alda-1 group (P<0.05). Thus, in a rat model of severe HS/R, treatment with Alda-1 increased the activity of ALDH2, significantly accelerated the clearance of reactive aldehydes, and concomitantly alleviated lung injury through improvement of pulmonary epithelial barrier integrity resulting in decreased alveolar epithelial tissue permeability, lung edema, and diffuse infiltration of inflammatory cells.
Collapse
|
127
|
Affiliation(s)
- Young Chul Yoo
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
128
|
Peng CK, Wu CP, Lin JY, Peng SC, Lee CH, Huang KL, Shen CH. Gas6/Axl signaling attenuates alveolar inflammation in ischemia-reperfusion-induced acute lung injury by up-regulating SOCS3-mediated pathway. PLoS One 2019; 14:e0219788. [PMID: 31318922 PMCID: PMC6638944 DOI: 10.1371/journal.pone.0219788] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 07/01/2019] [Indexed: 01/17/2023] Open
Abstract
Background Axl is a cell surface receptor tyrosine kinase, and activation of the Axl attenuates inflammation induced by various stimuli. Growth arrest-specific 6 (Gas6) has high affinity for Axl receptor. The role of Gas6/Axl signaling in ischemia-reperfusion-induced acute lung injury (IR-ALI) has not been explored previously. We hypothesized that Gas6/Axl signaling regulates IR-induced alveolar inflammation via a pathway mediated by suppressor of cytokine signaling 3 (SOCS3). Methods IR-ALI was induced by producing 30 min of ischemia followed by 90 min of reperfusion in situ in an isolated and perfused rat lung model. The rats were randomly allotted to a control group and IR groups, which were treated with three different doses of Gas6. Mouse alveolar epithelium MLE-12 cells were cultured in control and hypoxia-reoxygenation (HR) conditions with or without Gas6 and Axl inhibitor R428 pretreatment. Results We found that Gas6 attenuated IR-induced lung edema, the production of proinflammatory cytokines in perfusates, and the severity of ALI ex vivo. IR down-regulated SOCS3 expression and up-regulated NF-κB, and Gas6 restored this process. In the model of MLE-12 cells with HR, Gas6 suppressed the activation of TRAF6 and NF-κB by up-regulating SOCS3. Axl expression of alveolar epithelium was suppressed in IR-ALI but Gas6 restored phosphorylation of Axl. The anti-inflammatory effect of Gas6 was antagonized by R428, which highlighted that phosphorylation of Axl mediated the protective role of Gas6 in IR-ALI. Conclusions Gas6 up-regulates phosphorylation of Axl on alveolar epithelium in IR-ALI. The Gas6/Axl signaling activates the SOCS3-mediated pathway and attenuates IR-related inflammation and injury.
Collapse
Affiliation(s)
- Chung-Kan Peng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Pyng Wu
- Department of Critical Care Medicine, Landseed Hospital, Taoyuan, Taiwan
| | - Jr-Yu Lin
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Chi Peng
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Hsing Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Hao Shen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
129
|
de Oliveira THC, Souza DG, Teixeira MM, Amaral FA. Tissue Dependent Role of PTX3 During Ischemia-Reperfusion Injury. Front Immunol 2019; 10:1461. [PMID: 31354697 PMCID: PMC6635462 DOI: 10.3389/fimmu.2019.01461] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 06/10/2019] [Indexed: 01/06/2023] Open
Abstract
Reperfusion of an ischemic tissue is the treatment of choice for several diseases, including myocardial infarction and stroke. However, reperfusion of an ischemic tissue causes injury, known as Ischemia and Reperfusion Injury (IRI), that limits the benefit of blood flow restoration. IRI also occurs during solid organ transplantation. During IRI, there is activation of the innate immune system, especially neutrophils, which contributes to the degree of injury. It has been shown that PTX3 can regulate multiple aspects of innate immunity and tissue inflammation during sterile injury, as observed during IRI. In humans, levels of PTX3 increase in blood and elevated levels associate with extent of IRI. In mice, there is also enhanced expression of PTX3 in tissues and plasma after IRI. In general, absence of PTX3, as seen in PTX3-deficient mice, results in worse outcome after IRI. On the contrary, increased expression of PTX3, as seen in PTX3 transgenic mice and after PTX3 administration, is associated with better outcome after IRI. The exception is the gut where PTX3 seems to have a clear deleterious role. Here, we discuss mechanisms by which PTX3 contributes to IRI and the potential of taming this system for the treatment of injuries associated with reperfusion of solid organs.
Collapse
Affiliation(s)
| | - Danielle G Souza
- Host-Microorganism Interaction Laboratory, Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Flávio Almeida Amaral
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
130
|
Gong J, Ju YN, Wang XT, Zhu JL, Jin ZH, Gao W. Ac2-26 ameliorates lung ischemia-reperfusion injury via the eNOS pathway. Biomed Pharmacother 2019; 117:109194. [PMID: 31387174 DOI: 10.1016/j.biopha.2019.109194] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/19/2019] [Accepted: 06/28/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Lung ischemia-reperfusion injury (LIRI) is a major complication after lung transplantation. Annexin A1 (AnxA1) ameliorates inflammation in various injured organs. This study aimed to determine the effects and mechanism of AnxA1 on LIRI after lung transplantation. METHODS Thirty-two rats were randomized into sham, saline, Ac2-26 and Ac2-26/L groups. Rats in the saline, Ac2-26 and Ac2-26/L groups underwent left lung transplantation and received saline, Ac2-26, and Ac2-26/L-NIO, respectively. After 24 h of reperfusion, serum and transplanted lung tissues were examined. RESULTS The partial pressure of oxygen (PaO2) was increased in the Ac2-26 group compared to that in the saline group but was decreased by L-NIO treatment. In the Ac2-26 group, the wet-to-dry (W/D) weight ratios, total protein concentrations, proinflammatory factors and inducible nitric oxide synthase levels were notably decreased, but the concentrations of anti-inflammatory factors and endothelial nitric oxide synthase levels were significantly increased. Ac2-26 attenuated histological injury and cell apoptosis, and this improvement was reversed by L-NIO. CONCLUSIONS Ac2-26 reduced LIRI and improved alveoli-capillary permeability by inhibiting oxygen stress, inflammation and apoptosis. The protective effect of Ac2-26 on LIRI largely depended on the endothelial nitric oxide synthase pathway.
Collapse
Affiliation(s)
- Jing Gong
- Anesthesiology Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150000, China.
| | - Ying-Nan Ju
- Department of ICU, The Tumor Hospital of Harbin Medical University, 150 Haping Road, Harbin 150081, China.
| | - Xue-Ting Wang
- Anesthesiology Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150000, China.
| | - Jing-Li Zhu
- Anesthesiology Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150000, China.
| | - Zhe-Hao Jin
- Anesthesiology Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150000, China.
| | - Wei Gao
- Anesthesiology Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin 150000, China.
| |
Collapse
|
131
|
Endothelial Glycocalyx Shedding Predicts Donor Organ Acceptability and Is Associated With Primary Graft Dysfunction in Lung Transplant Recipients. Transplantation 2019; 103:1277-1285. [DOI: 10.1097/tp.0000000000002539] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
132
|
Protective Effect of Thymosin β4 against Abdominal Aortic Ischemia-Reperfusion-Induced Acute Lung Injury in Rats. ACTA ACUST UNITED AC 2019; 55:medicina55050187. [PMID: 31121838 PMCID: PMC6572620 DOI: 10.3390/medicina55050187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/25/2019] [Accepted: 05/17/2019] [Indexed: 12/29/2022]
Abstract
Background and objectives: Ischemia-reperfusion (IR) caused by infrarenal abdominal aorta cross-clamping is an important factor in the development of ischemia-reperfusion injury in various distant organs. Materials and Methods: We investigated potential antioxidant/anti-inflammatory effects of thymosin beta 4 (Tβ4) in a rat model of abdominal aortic surgery-induced IR. Tβ4 (10 mg/kg, intravenous (i.v.)) was administered to rats with IR (90-min ischemia, 180-min reperfusion) at two different periods. One group received Tβ4 1 h before ischemia, and the other received 15 min before the reperfusion period. Results: Results were compared to control and non-Tβ4-treated rats with IR. Serum, bronchoalveolar lavage fluid and lung tissue levels of oxidant parameters were higher, while antioxidant levels were lower in the IR group compared to control. IR also increased inflammatory cytokine levels. Tβ4 reverted these parameters in both Tβ4-treated groups compared to the untreated IR group. Conclusions: Since there is no statistical difference between the prescribed results of both Tβ4-treated groups, our study demonstrates that Tβ4 reduced lung oxidative stress and inflammation following IR and prevented lung tissue injury regardless of timing of administration.
Collapse
|
133
|
Wang ML, Wei CH, Wang WD, Wang JS, Zhang J, Wang JJ. Melatonin attenuates lung ischaemia-reperfusion injury via inhibition of oxidative stress and inflammation. Interact Cardiovasc Thorac Surg 2019; 26:761-767. [PMID: 29346581 DOI: 10.1093/icvts/ivx440] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/22/2017] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES Lung ischaemia-reperfusion injury is a complex pathophysiological process due to the production of reactive oxygen species and the generation of inflammatory reaction. We investigated the protective effects and the corresponding mechanism of melatonin (MT), a potent free-radical scavenger, on lung injury induced by ischaemia-reperfusion in a mouse model. METHODS Adult male C57BL/6J mice (n = 30) were randomly and equally allocated into 5 groups: sham controls, IR, IR + 10 mg/kg MT, IR + 20 mg/kg MT and IR + 30 mg/kg MT. Lung ischaemia-reperfusion injury was induced by thoracotomy followed by clamping of the left hilum for 1 h and subsequent reperfusion for 2 h. RESULTS Histological scoring analysis showed that lung parenchymal damage was ameliorated in the melatonin pretreatment groups when compared with the IR group, with the IR + 20 mg/kg MT group showing the strongest effect among the melatonin pretreatment groups. Wet-to-dry weight ratio, detection of malondialdehyde, protein expressions of inflammatory factors (tumour necrosis factor-α, interleukin-1β, NF-κB and IKK-γ) and apoptotic factors (cleaved caspase-3 and Bax/Bcl-2), as well as TUNEL assay showed changes similar to those of the lung injury scores in all groups. In contrast, the examination of superoxide dismutase showed a pattern contrary to that of the lung injury score in all groups. In addition, immunohistochemistry staining showed that the expressions of the antioxidants glutathione peroxidase and glutathione reductase were increased in the melatonin pretreatment groups. CONCLUSIONS This study demonstrated that melatonin pretreatment attenuated lung ischaemia-reperfusion injury via inhibition of oxidative stress, inflammation and apoptosis.
Collapse
Affiliation(s)
- Ming-Liang Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun-Hua Wei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Dong Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Shun Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Jun Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
134
|
Yang S, Tian J, Zhang F, Liu A, Xie B, Chen Q. The protective effects of heat shock protein 22 in lung ischemia-reperfusion injury mice. Biochem Biophys Res Commun 2019; 512:698-704. [PMID: 30922561 DOI: 10.1016/j.bbrc.2019.03.048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 03/08/2019] [Indexed: 10/27/2022]
Abstract
Lung ischemia-reperfusion injury (LIRI) often results in respiratory insufficiency after pulmonary embolism, lung transplantation, etc. To investigate the role of HSP22 in LIRI mice, ischemia-reperfusion injury was established in the left lung of an HSP22 overexpression transgenic mouse. Twelve HSP22 transgenic (TG) mice and twelve wild-type (WT) mice were randomly divided into 2 groups: the sham-operated group (SO: TG-SO, WT-SO) and the ischemia-reperfusion group (I/R: TG-I/R, WT-I/R), respectively. We tested the PaO2, W/D ratio, and MDA level; observed morphology changes; and calculated the index of alveolar damage. HSP22 expression was examined in lung tissues of TG and WT C57BL mice by immunohistochemistry. TUNEL assay was performed to measure apoptosis. We found that HSP22 was significantly overexpressed in TG mice. There was no difference in PaO2 among the four groups. In the I/R group, the W/D ratio, MDA and index of alveolar damage were higher than those in the SO group. Moreover, compared with WT-I/R group, the W/D ratio, MDA and index of alveolar damage in the TG-I/R group were significantly decreased. Apoptosis in the I/R groups was increased compared to that in the SO groups, while apoptosis in the TG-I/R groups was decreased compared to that in the WT-I/R groups. Our results showed that HSP22 TG mice and the LIRI model were successfully established. In addition, HSP22 overexpression has protective effects on LIRI by inhibiting lipid peroxidation and apoptosis.
Collapse
Affiliation(s)
- Shasha Yang
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Jie Tian
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Fufeng Zhang
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Aibin Liu
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Bin Xie
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China
| | - Qiong Chen
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
135
|
Liu WC, Chen SB, Liu S, Ling X, Xu QR, Yu BT, Tang J. Inhibition of mitochondrial autophagy protects donor lungs for lung transplantation against ischaemia-reperfusion injury in rats via the mTOR pathway. J Cell Mol Med 2019; 23:3190-3201. [PMID: 30887674 PMCID: PMC6484325 DOI: 10.1111/jcmm.14177] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/05/2018] [Accepted: 01/02/2019] [Indexed: 12/12/2022] Open
Abstract
Impaired mitochondrial function is a key factor attributing to lung ischaemia‐reperfusion (IR) injury, which contributes to major post‐transplant complications. Thus, the current study was performed to investigate the role of mitochondrial autophagy in lung I/R injury and the involvement of the mTOR pathway. We established rat models of orthotopic left lung transplantation to investigate the role of mitochondrial autophagy in I/R injury following lung transplantation. Next, we treated the donor lungs with 3‐MA and Rapamycin to evaluate mitochondrial autophagy, lung function and cell apoptosis with different time intervals of cold ischaemia preservation and reperfusion. In addition, mitochondrial autophagy, and cell proliferation and apoptosis of pulmonary microvascular endothelial cells (PMVECs) exposed to hypoxia‐reoxygenation (H/R) were monitored after 3‐MA administration or Rapamycin treatment. The cell apoptosis could be inhibited by mitochondrial autophagy at the beginning of lung ischaemia, but was rendered out of control when mitochondrial autophagy reached normal levels. After I/R of donor lung, the mitochondrial autophagy was increased until 6 hours after reperfusion and then gradually decreased. The elevation of mitochondrial autophagy was accompanied by promoted apoptosis, aggravated lung injury and deteriorated lung function. Moreover, the suppression of mitochondrial autophagy by 3‐MA inhibited cell apoptosis of donor lung to alleviate I/R‐induced lung injury as well as inhibited H/R‐induced PMVEC apoptosis, and enhanced its proliferation. Finally, mTOR pathway participated in I/R‐ and H/R‐mediated mitochondrial autophagy in regulation of cell apoptosis. Inhibition of I/R‐induced mitochondrial autophagy alleviated lung injury via the mTOR pathway, suggesting a potential therapeutic strategy for lung I/R injury.
Collapse
Affiliation(s)
- Wei-Cheng Liu
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Shi-Biao Chen
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Sheng Liu
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Xiang Ling
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Qi-Rong Xu
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Ben-Tong Yu
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Jian Tang
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| |
Collapse
|
136
|
Activation mechanisms and multifaceted effects of mast cells in ischemia reperfusion injury. Exp Cell Res 2019; 376:227-235. [DOI: 10.1016/j.yexcr.2019.01.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/20/2019] [Accepted: 01/31/2019] [Indexed: 12/16/2022]
|
137
|
The Amide Local Anesthetic Ropivacaine Attenuates Acute Rejection After Allogeneic Mouse Lung Transplantation. Lung 2019; 197:217-226. [DOI: 10.1007/s00408-019-00197-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 01/27/2019] [Indexed: 02/07/2023]
|
138
|
Abstract
Lung transplantation is a life-saving operation for patients with advanced lung disease. Pulmonary allografts eventually fail because of infection, thromboembolism, malignancy, airway complications, and chronic rejection, otherwise known as chronic lung allograft dysfunction (CLAD). Emerging evidence suggests that a highly-compromised airway circulation contributes to the evolution of airway complications and CLAD. There are two significant causes of poor perfusion and airway hypoxia in lung transplantation: an abnormal bronchial circulation which causes airway complications and microvascular rejection which induces CLAD. At the time of transplantation, the bronchial artery circulation, a natural component of the airway circulatory anatomy, is not surgically connected, and bronchi distal to the anastomosis become hypoxic. Subsequently, the bronchial anastomosis is left to heal under ischemic conditions. Still later, the extant microvessels in transplant bronchi are subjected to alloimmune insults that can further negatively impact pulmonary function. This review describes how airway tissue hypoxia evolves in lung transplantation, why depriving oxygenation in the bronchi and more distal bronchioles contributes to disease pathology and what therapeutic interventions are currently emerging to address these vascular injuries. Improving anastomotic vascular healing at the time of transplantation and preventing microvascular loss during acute rejection episodes are two steps that could limit airway hypoxia and improve patient outcomes.
Collapse
Affiliation(s)
- Shravani Pasnupneti
- VA Palo Alto Health Care System/Stanford University, 3801 Miranda Ave., Palo Alto CA 94304 USA
| | - Mark R. Nicolls
- VA Palo Alto Health Care System/Stanford University, 3801 Miranda Ave., Palo Alto CA 94304 USA
| |
Collapse
|
139
|
Garutti I, Gonzalez-Moraga F, Sanchez-Pedrosa G, Casanova J, Martin-Piñeiro B, Rancan L, Simón C, Vara E. The effect of anesthetic preconditioning with sevoflurane on intracellular signal-transduction pathways and apoptosis, in a lung autotransplant experimental model. BRAZILIAN JOURNAL OF ANESTHESIOLOGY (ENGLISH EDITION) 2019. [PMID: 30459087 PMCID: PMC9391783 DOI: 10.1016/j.bjane.2018.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background Anesthetic pre-conditioning attenuates inflammatory response during ischemia-reperfusion lung injury. The molecular mechanisms to explain it are not fully understood. The aim of our investigation was to analyze the molecular mechanism that explain the anti-inflammatory effects of anesthetic pre-conditioning with sevoflurane focusing on its effects on MAPKs (mitogen-activated protein kinases), NF-κB (nuclear factor kappa beta) pathways, and apoptosis in an experimental lung autotransplant model. Methods Twenty large white pigs undergoing pneumonectomy plus lung autotransplant were divided into two 10-member groups on the basis of the anesthetic received (propofol or sevoflurane). Anesthetic pre-conditioning group received sevoflurane 3% after anesthesia induction and it stopped when one-lung ventilation get started. Control group did not receive sevoflurane in any moment during the whole study period. Intracellular signal-transduction pathways (MAPK family), transcription factor (NF-κB), and apoptosis (caspases 3 and 9) were analyzed during experiment. Results Pigs that received anesthetic pre-conditioning with sevoflurane have shown significant lower values of MAPK-p38, MAPK-P-p38, JNK (c-Jun N-terminal kinases), NF-κB p50 intranuclear, and caspases (p < 0.05) than pigs anesthetized with intravenous propofol. Conclusions Lung protection of anesthetic pre-conditioning with sevoflurane during experimental lung autotransplant is, at least, partially associated with MAPKs and NF κB pathways attenuation, and antiapoptotic effects.
Collapse
Affiliation(s)
- Ignacio Garutti
- Hospital General Universitario Gregorio Marañon, Departamento de Anestesiologia, Madri, Espanha; Universidad Complutense de Madrid, Departamento de Farmacologia, Madri, Espanha.
| | | | | | - Javier Casanova
- Hospital General Universitario Gregorio Marañon, Departamento de Anestesiologia, Madri, Espanha
| | | | - Lisa Rancan
- Universidad Complutense de Madrid, Facultad de Medicina, Departamento de Bioquímica, Madri, Espanha
| | - Carlos Simón
- Hospital General Universitario Gregorio Marañon, Departamento de Cirugía Torácica, Madri, Espanha; Universidad Complutense de Madrid, Departamento de Cirugía, Madri, Espanha
| | - Elena Vara
- Universidad Complutense de Madrid, Facultad de Medicina, Departamento de Bioquímica, Madri, Espanha
| |
Collapse
|
140
|
Abreu MDM, Almeida FMD, Santos KBD, Assis EACPD, Hamada RKF, Jatene FB, Pêgo-Fernandes PM, Pazetti R. Does methylene blue attenuate inflammation in nonischemic lungs after lung transplantation? J Bras Pneumol 2018; 44:378-382. [PMID: 30517338 PMCID: PMC6467587 DOI: 10.1590/s1806-37562017000000172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 12/07/2017] [Indexed: 11/22/2022] Open
Abstract
Objective: To evaluate whether methylene blue (MB) could minimize the effects of ischemia-reperfusion injury in the nonischemic lung on a lung transplantation rodent model. Methods: Forty female Sprague-Dawley rats were divided into 20 donors and 20 recipients. The 20 recipient rats were divided into two groups (n = 10) according to the treatment (0.9% saline vs. 1% MB solutions). All animals underwent unilateral lung transplantation. Recipients received 2 mL of saline or MB intraperitoneally prior to transplantation. After 2 h of reperfusion, the animals were euthanized and histopathological and immunohistochemical analyses were performed in the nonischemic lung. Results: There was a significant decrease in inflammation-neutrophil count and intercellular adhesion molecule-1 (ICAM-1) expression in lung parenchyma were higher in the saline group in comparison with the MB group-and in apoptosis-caspase-3 expression was higher in the saline group and Bcl-2 expression was higher in MB group. Conclusions: MB is an effective drug for the protection of nonischemic lungs against inflammation and apoptosis following unilateral lung transplantation in rats.
Collapse
Affiliation(s)
- Marcus da Matta Abreu
- . Departamento de Cardiopneumologia, Disciplina de Cirurgia Torácica, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Francine Maria de Almeida
- . Departamento de Cardiopneumologia, Disciplina de Cirurgia Torácica, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Kelli Borges Dos Santos
- . Núcleo de Pesquisa em Transplante, Faculdade de Enfermagem, Universidade Federal de Juiz de Fora, Juiz de Fora (MG) Brasil
| | | | | | - Fabio Biscegli Jatene
- . Departamento de Cardiopneumologia, Disciplina de Cirurgia Torácica, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Paulo Manuel Pêgo-Fernandes
- . Departamento de Cardiopneumologia, Disciplina de Cirurgia Torácica, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| | - Rogerio Pazetti
- . Departamento de Cardiopneumologia, Disciplina de Cirurgia Torácica, Instituto do Coração, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo (SP) Brasil
| |
Collapse
|
141
|
Steinmeyer J, Becker S, Avsar M, Salman J, Höffler K, Haverich A, Warnecke G, Mühlfeld C, Ochs M, Schnapper-Isl A. Cellular and acellular ex vivo lung perfusion preserve functional lung ultrastructure in a large animal model: a stereological study. Respir Res 2018; 19:238. [PMID: 30509256 PMCID: PMC6278069 DOI: 10.1186/s12931-018-0942-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 11/19/2018] [Indexed: 01/07/2023] Open
Abstract
Background Ex vivo lung perfusion (EVLP) is used by an increasing number of transplant centres. It is still controversial whether an acellular or cellular (erythrocyte enriched) perfusate is preferable. The aim of this paper was to evaluate whether acellular (aEVLP) or cellular EVLP (cEVLP) preserves functional lung ultrastructure better and to generate a hypothesis regarding possible underlying mechanisms. Methods Lungs of 20 pigs were assigned to 4 groups: control, ischaemia (24 h), aEVLP and cEVLP (both EVLP groups: 24 h ischaemia + 12 h EVLP). After experimental procedures, whole lungs were perfusion fixed, samples for light and electron microscopic stereology were taken, and ventilation, diffusion and perfusion related parameters were estimated. Results Lung structure was well preserved in all groups. Lungs had less atelectasis and higher air content after EVLP. No significant group differences were found in alveolar septum composition or blood-air barrier thickness. Small amounts of intraalveolar oedema were detected in both EVLP groups but significantly more in aEVLP than in cEVLP. Conclusions Both EVLP protocols supported lungs well for up to 12 h and could largely prevent ischaemia ex vivo reperfusion associated lung injury. In both EVLP groups, oedema volume remained below the level of functional relevance. The group difference in oedema formation was possibly due to inferior septal perfusion in aEVLP. Electronic supplementary material The online version of this article (10.1186/s12931-018-0942-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jasmin Steinmeyer
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Simon Becker
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover, Germany.,Department of Anesthesiology, Intensive Care, Palliative Care and Pain Medicine, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Murat Avsar
- Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, Germany
| | - Jawad Salman
- Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, Germany
| | - Klaus Höffler
- Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, Germany
| | - Axel Haverich
- REBIRTH Cluster of Excellence, Hannover, Germany.,Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Gregor Warnecke
- REBIRTH Cluster of Excellence, Hannover, Germany.,Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Matthias Ochs
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Anke Schnapper-Isl
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany. .,REBIRTH Cluster of Excellence, Hannover, Germany.
| |
Collapse
|
142
|
Schiavon M, Zampieri D, Marulli G, Verderi E, Rebusso A, Comacchio GM, Nicotra S, Loy M, Lunardi F, Feltracco P, Calabrese F, Cozzi E, Rea F. Pushing the limits of reconditioning: extended normothermic lung perfusion in an extended criteria donor. J Thorac Dis 2018; 10:E796-E801. [PMID: 30746256 DOI: 10.21037/jtd.2018.10.125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Marco Schiavon
- Thoracic Surgery Division, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| | - Davide Zampieri
- Thoracic Surgery Division, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| | - Giuseppe Marulli
- Thoracic Surgery Division, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| | - Enrico Verderi
- Thoracic Surgery Division, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| | - Alessandro Rebusso
- Thoracic Surgery Division, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| | - Giovanni Maria Comacchio
- Thoracic Surgery Division, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| | - Samuele Nicotra
- Thoracic Surgery Division, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| | - Monica Loy
- Thoracic Surgery Division, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| | - Francesca Lunardi
- Pathology Division, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| | - Paolo Feltracco
- Institute of Anesthesiology and Intensive Care, Department of Medicine, Padova University Hospital, Padova, Italy
| | - Fiorella Calabrese
- Pathology Division, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| | - Emanuele Cozzi
- Transplant Immunology Unit, Department of Cardiac, Thoracic and Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| | - Federico Rea
- Thoracic Surgery Division, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padova University Hospital, Padova, Italy
| |
Collapse
|
143
|
Wei J, Wang P, Li Y, Dou Q, Lin J, Tao W, Lin J, Fu X, Huang Z, Zhang W. Inhibition of RHO Kinase by Fasudil Attenuates Ischemic Lung Injury After Cardiac Arrest in Rats. Shock 2018; 50:706-713. [DOI: 10.1097/shk.0000000000001097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
144
|
|
145
|
Garutti I, Gonzalez-Moraga F, Sanchez-Pedrosa G, Casanova J, Martin-Piñeiro B, Rancan L, Simón C, Vara E. [The effect of anesthetic preconditioning with sevoflurane on intracellular signal-transduction pathways and apoptosis, in a lung autotransplant experimental model]. Rev Bras Anestesiol 2018; 69:48-57. [PMID: 30459087 DOI: 10.1016/j.bjan.2018.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 06/29/2018] [Accepted: 07/13/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Anesthetic pre-conditioning attenuates inflammatory response during ischemia-reperfusion lung injury. The molecular mechanisms to explain it are not fully understood. The aim of our investigation was to analyze the molecular mechanism that explain the anti-inflammatory effects of anesthetic pre-conditioning with sevoflurane focusing on its effects on MAPKs (mitogen-activated protein kinases), NF-κB (nuclear factor kappa beta) pathways, and apoptosis in an experimental lung autotransplant model. METHODS Twenty large white pigs undergoing pneumonectomy plus lung autotransplant were divided into two 10-member groups on the basis of the anesthetic received (propofol or sevoflurane). Anesthetic pre-conditioning group received sevoflurane 3% after anesthesia induction and it stopped when one-lung ventilation get started. Control group did not receive sevoflurane in any moment during the whole study period. Intracellular signal-transduction pathways (MAPK family), transcription factor (NF-κB), and apoptosis (caspases 3 and 9) were analyzed during experiment. RESULTS Pigs that received anesthetic pre-conditioning with sevoflurane have shown significant lower values of MAPK-p38, MAPK-P-p38, JNK (c-Jun N-terminal kinases), NF-κB p50 intranuclear, and caspases (p<0.05) than pigs anesthetized with intravenous propofol. CONCLUSIONS Lung protection of anesthetic pre-conditioning with sevoflurane during experimental lung autotransplant is, at least, partially associated with MAPKs and NF κB pathways attenuation, and antiapoptotic effects.
Collapse
Affiliation(s)
- Ignacio Garutti
- Hospital General Universitario Gregorio Marañon, Departamento de Anestesiologia, Madri, Espanha; Universidad Complutense de Madrid, Departamento de Farmacologia, Madri, Espanha.
| | | | | | - Javier Casanova
- Hospital General Universitario Gregorio Marañon, Departamento de Anestesiologia, Madri, Espanha
| | | | - Lisa Rancan
- Universidad Complutense de Madrid, Facultad de Medicina, Departamento de Bioquímica, Madri, Espanha
| | - Carlos Simón
- Hospital General Universitario Gregorio Marañon, Departamento de Cirugía Torácica, Madri, Espanha; Universidad Complutense de Madrid, Departamento de Cirugía, Madri, Espanha
| | - Elena Vara
- Universidad Complutense de Madrid, Facultad de Medicina, Departamento de Bioquímica, Madri, Espanha
| |
Collapse
|
146
|
Kawasaki T, Chen W, Htwe YM, Tatsumi K, Dudek SM. DPP4 inhibition by sitagliptin attenuates LPS-induced lung injury in mice. Am J Physiol Lung Cell Mol Physiol 2018; 315:L834-L845. [PMID: 30188745 DOI: 10.1152/ajplung.00031.2018] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe clinical condition marked by acute respiratory failure and dysregulated inflammation. Pulmonary vascular endothelial cells (PVECs) function as an important pro-inflammatory source in ARDS, suggesting that modulation of inflammatory events at the endothelial level may have a therapeutic benefit. Dipeptidyl peptidase-4 (DPP4) inhibitors, widely used for the treatment of diabetes mellitus, have been reported to have possible anti-inflammatory effects. However, the potential anti-inflammatory effects of DPP4 inhibition on PVEC function and ARDS pathophysiology are unknown. Therefore, we evaluated the effects of sitagliptin, a DPP4 inhibitor in wide clinical use, on LPS-induced lung injury in mice and in human lung ECs in vitro. In vivo, sitagliptin reduced serum DPP4 activity, bronchoalveolar lavage protein concentration, cell number, and proinflammatory cytokine levels after LPS and alleviated histological findings of lung injury. LPS decreased the expression levels of CD26/DPP4 on pulmonary epithelial cells and PVECs isolated from mouse lungs, and the effect was partially reversed by sitagliptin. In vitro, human lung microvascular ECs (HLMVECs) expressed higher levels of CD26/DPP4 than human pulmonary arterial ECs. LPS induced the release of TNFα, IL-6, and IL-8 by HLMVECs that were inhibited by sitagliptin. LPS promoted the proliferation of HLMVECs, and sitagliptin suppressed this response. However, sitagliptin failed to reverse LPS-induced permeability in cultured ECs or lung epithelial cells in vitro. In summary, sitagliptin attenuates LPS-induced lung injury in mice and exerts anti-inflammatory effects on HLMVECs. These novel observations indicate DPP4 inhibitors may have potential as therapeutic drugs for ARDS.
Collapse
Affiliation(s)
- Takeshi Kawasaki
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois.,Department of Respirology, Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Weiguo Chen
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Yu Maw Htwe
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Steven M Dudek
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| |
Collapse
|
147
|
Shock Wave Therapy Enhances Mitochondrial Delivery into Target Cells and Protects against Acute Respiratory Distress Syndrome. Mediators Inflamm 2018; 2018:5425346. [PMID: 30420790 PMCID: PMC6215567 DOI: 10.1155/2018/5425346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/05/2018] [Indexed: 01/11/2023] Open
Abstract
This study tested the hypothesis that shock wave therapy (SW) enhances mitochondrial uptake into the lung epithelial and parenchymal cells to attenuate lung injury from acute respiratory distress syndrome (ARDS). ARDS was induced in rats through continuous inhalation of 100% oxygen for 48 h, while SW entailed application 0.15 mJ/mm2 for 200 impulses at 6 Hz per left/right lung field. In vitro and ex vivo studies showed that SW enhances mitochondrial uptake into lung epithelial and parenchyma cells (all p < 0.001). Flow cytometry demonstrated that albumin levels and numbers of inflammatory cells (Ly6G+/CD14+/CD68+/CD11b/c+) in bronchoalveolar lavage fluid were the highest in untreated ARDS, were progressively reduced across SW, Mito, and SW + Mito (all p < 0.0001), and were the lowest in sham controls. The same profile was also seen for fibrosis/collagen deposition, levels of biomarkers of oxidative stress (NOX-1/NOX-2/oxidized protein), inflammation (MMP-9/TNF-α/NF-κB/IL-1β/ICAM-1), apoptosis (cleaved caspase 3/PARP), fibrosis (Smad3/TGF-β), mitochondrial damage (cytosolic cytochrome c) (all p < 0.0001), and DNA damage (γ-H2AX+), and numbers of parenchymal inflammatory cells (CD11+/CD14+/CD40L+/F4/80+) (p < 0.0001). These results suggest that SW-assisted Mito therapy effectively protects the lung parenchyma from ARDS-induced injury.
Collapse
|
148
|
Maltesen RG, Buggeskov KB, Andersen CB, Plovsing R, Wimmer R, Ravn HB, Rasmussen BS. Lung Protection Strategies during Cardiopulmonary Bypass Affect the Composition of Bronchoalveolar Fluid and Lung Tissue in Cardiac Surgery Patients. Metabolites 2018; 8:metabo8040054. [PMID: 30241409 PMCID: PMC6316472 DOI: 10.3390/metabo8040054] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/31/2018] [Accepted: 09/19/2018] [Indexed: 11/18/2022] Open
Abstract
Pulmonary dysfunction is among the most frequent complications to cardiac surgeries. Exposure of blood to the cardiopulmonary bypass (CPB) circuit with subsequent lung ischemia-reperfusion leads to the production of inflammatory mediators and increases in microvascular permeability. The study aimed to elucidate histological, cellular, and metabolite changes following two lung protective regimens during CPB with Histidine-Tryptophan-Ketoglutarate (HTK) enriched or warm oxygenated blood pulmonary perfusion compared to standard regimen with no pulmonary perfusion. A total of 90 patients undergoing CPB were randomized to receiving HTK, oxygenated blood or standard regimen. Of these, bronchoalveolar lavage fluid (BALF) and lung tissue biopsies were obtained before and after CPB from 47 and 25 patients, respectively. Histopathological scores, BALF cell counts and metabolite screening were assessed. Multivariate and univariate analyses were performed. Profound histological, cellular, and metabolic changes were identified in all patients after CPB. Histological and cellular changes were similar in the three groups; however, some metabolite profiles were different in the HTK patients. While all patients presented an increase in inflammatory cells, metabolic acidosis, protease activity and oxidative stress, HTK patients seemed to be protected against severe acidosis, excessive fatty acid oxidation, and inflammation during ischemia-reperfusion. Additional studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Raluca G Maltesen
- Department of Anesthesia and Intensive Care Medicine, Aalborg University Hospital, 9000 Aalborg, Denmark.
| | - Katrine B Buggeskov
- Department of Cardiothoracic Anesthesia, Heart Centre, Rigshospitalet, 2100 Copenhagen, Denmark.
| | - Claus B Andersen
- Department of Forensic Medicine, University of Copenhagen, 2100 Copenhagen, Denmark.
| | - Ronni Plovsing
- Department of Intensive Care, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark.
- Department of Anesthesiology, Hvidovre Hospital, University of Copenhagen, 2650 Hvidovre, Denmark.
| | - Reinhard Wimmer
- Department of Chemistry and Bioscience, Aalborg University, 9220 Aalborg, Denmark.
| | - Hanne B Ravn
- Department of Cardiothoracic Anesthesia, Heart Centre, Rigshospitalet, 2100 Copenhagen, Denmark.
| | - Bodil S Rasmussen
- Department of Anesthesia and Intensive Care Medicine, Aalborg University Hospital, 9000 Aalborg, Denmark.
- Department of Clinical Medicine, School of Medicine and Health, Aalborg University, 9220 Aalborg, Denmark.
| |
Collapse
|
149
|
Shen CH, Lin JY, Chang YL, Wu SY, Peng CK, Wu CP, Huang KL. Inhibition of NKCC1 Modulates Alveolar Fluid Clearance and Inflammation in Ischemia-Reperfusion Lung Injury via TRAF6-Mediated Pathways. Front Immunol 2018; 9:2049. [PMID: 30271405 PMCID: PMC6146090 DOI: 10.3389/fimmu.2018.02049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 08/20/2018] [Indexed: 12/15/2022] Open
Abstract
Background: The expression of Na-K-2Cl cotransporter 1 (NKCC1) in the alveolar epithelium is responsible for fluid homeostasis in acute lung injury (ALI). Increasing evidence suggests that NKCC1 is associated with inflammation in ALI. We hypothesized that inhibiting NKCC1 would attenuate ALI after ischemia-reperfusion (IR) by modulating pathways that are mediated by tumor necrosis-associated factor 6 (TRAF6). Methods: IR-ALI was induced by producing 30 min of ischemia followed by 90 min of reperfusion in situ in an isolated and perfused rat lung model. The rats were randomly allotted into four groups comprising two control groups and two IR groups with and without bumetanide. Alveolar fluid clearance (AFC) was measured for each group. Mouse alveolar MLE-12 cells were cultured in control and hypoxia-reoxygenation (HR) conditions with or without bumetanide. Flow cytometry and transwell monolayer permeability assay were carried out for each group. Results: Bumetanide attenuated the activation of p-NKCC1 and lung edema after IR. In the HR model, bumetanide decreased the cellular volume and increased the transwell permeability. In contrast, bumetanide increased the expression of epithelial sodium channel (ENaC) via p38 mitogen-activated protein kinase (p38 MAPK), which attenuated the reduction of AFC after IR. Bumetanide also modulated lung inflammation via nuclear factor-κB (NF-κB). TRAF6, which is upstream of p38 MAPK and NF-κB, was attenuated by bumetanide after IR and HR. Conclusions: Inhibition of NKCC1 by bumetanide reciprocally modulated epithelial p38 MAPK and NF-κB via TRAF6 in IR-ALI. This interaction attenuated the reduction of AFC via upregulating ENaC expression and reduced lung inflammation.
Collapse
Affiliation(s)
- Chih-Hao Shen
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Jr-Yu Lin
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Chung-Kan Peng
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan
| | - Chin-Pyng Wu
- Department of Critical Care Medicine, Landseed Hospital, Taoyuan, Taiwan
| | - Kun-Lun Huang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan.,Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
150
|
Penehyclidine hydrochloride preconditioning provides pulmonary and systemic protection in a rat model of lung ischaemia reperfusion injury. Eur J Pharmacol 2018; 839:1-11. [PMID: 30201378 DOI: 10.1016/j.ejphar.2018.09.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 11/23/2022]
Abstract
Penehyclidine hydrochloride (PHC) is a new anticholinergic agent that provides protective effects in experimental models of heart and brain ischaemia as well as reperfusion (I/R) injury. In this study, we tested the hypothesis that PHC can alleviate lung ischaemia-reperfusion injury and improve pulmonary and systemic function in rats. PHC was administered intravenously at various doses (d= 0.1, 0.3, 1, 3 mg/kg) to I/R rats. We used six indicators, including lung function, histologic examination, pulmonary oedema, oxidative stress, inflammatory responses, and apoptosis staining to quantify the pulmonary and systemic protective effects of PHC. Haematoxylin and eosin staining was used for pulmonary histologic examination. The expression of Toll-like receptor (TLR) 4, phospho-inhibitor of NF-κB (p-IκB) and nuclear factor-kappa B (NF-κB) was analysed using western blotting. ELISA was conducted to detect inflammatory mediators. Oxidative stress markers as well as myeloperoxidase (MPO) were determined using an assay kit. PHC preconditioning (with concentrations ranging from 0.3 mg/kg to 3 mg/kg 30 min before the onset of I/R) significantly reduced lung histopathological changes, down regulated TLR4, p-IκB and NF-κB expression, and decreased inflammatory mediators as well as the total number of leukocytes and neutrophils in bronchoalveolar lavage (BAL) fluid and plasma. The lung tissue contents of reactive oxygen species (ROS), malondialdehyde (MDA), and MPO as well as pulmonary oedema formation decreased, while SOD (superoxide dismutase) activity was significantly upregulated. PHC preconditioning (with concentrations ranging from 1 mg/kg to 3 mg/kg) significantly improved the lung function and attenuated the apoptotic rate. The probable mechanism for this finding is the inhibition of proinflammatory mediators via the suppression of reactive oxygen species production and the TLR4/NF-κB signalling pathway.
Collapse
|