1
|
Zhuang F, Ning Y, Li C, Luo Y, Gao P, Wang T, Dai C, Xie H, He W, Wu J, Chen C. Single-cell RNA Sequencing of Pig Lung Transplantation Reveals Macrophage Ferroptosis in Lung IschemiaReperfusion Injury. J Heart Lung Transplant 2025:S1053-2498(25)01918-7. [PMID: 40262727 DOI: 10.1016/j.healun.2025.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025] Open
Abstract
Primary graft dysfunction (PGD), which is caused primarily by ischemia-reperfusion injury (IRI), is a major obstacle in lung transplantation. Here, we developed an orthotopic, single-lung transplant pig model to simulate prolonged cold IRI. After 24 hours of cold ischemia and 8 hours of warm reperfusion, the transplanted lung exhibited severe allograft injury. Subsequent single-cell RNA sequencing (scRNA-seq) revealed significant changes in alveolar macrophages after IRI, with prominently enriched ferroptosis pathways. Transmission electron microscopy (TEM) confirmed characteristic ferroptosis changes in lung macrophages, and decreased GPX4 expression in macrophages indicated increased susceptibility to ferroptosis. Overall, our pig orthotopic left lung transplant model effectively simulates IRI after transplantation, which offers a valuable platform for more detailed investigations of early reperfusion injury to pulmonary grafts. Moreover, we preliminarily demonstrated the importance of macrophage ferroptosis in IRI, suggesting that inhibiting macrophage ferroptosis may be a promising therapeutic strategy for lung IRI.
Collapse
Affiliation(s)
- Fenghui Zhuang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Ye Ning
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chongwu Li
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunzhe Luo
- BGI Research, Beijing, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Peigen Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Tao Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chenyang Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenxin He
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China.
| |
Collapse
|
2
|
Wei SL, Zhang XQ, Li J, Zhang RZ, Yang JB, Zhang R, Li YN, Li B. Veno-venous extracorporeal membrane oxygenation exacerbates lung ischaemia-reperfusion injury in a rat model. Eur J Cardiothorac Surg 2025; 67:ezaf106. [PMID: 40163702 DOI: 10.1093/ejcts/ezaf106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/04/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025] Open
Abstract
OBJECTIVES Although lung transplantation has experienced great development in the past decades, the survival rate remains low, and lung ischaemia-reperfusion injury during transplantation is a major cause of primary graft dysfunction, which causes early morbidity and death after lung transplantation. Extracorporeal membrane oxygenation (ECMO) has been increasingly used as intraoperative support during lung transplantation. However, the clinical outcomes of intraoperative ECMO in lung transplantation remain controversial. Here, we established veno-venous ECMO (VV ECMO) in a lung ischaemia-reperfusion rat model to investigate its impact on lung injury. METHODS Eighteen rats were allocated to Sham, ischemia-reperfusion (IR) and IR-ECMO group. Using left pulmonary hilum ischaemia for 1 h, VV ECMO was established during reperfusion for 2 h. Lung tissue, blood sample and bronchoalveolar lavage fluid were collected for further evaluation using haematoxylin and eosin staining, immunohistochemistry, quantitative polymerase chain reaction, bicinchoninic acid assay and enzyme-linked immunosorbent assay. RESULTS VV ECMO aggravates lung ischaemia-reperfusion injury; the pathological injury is more severe in the IR-ECMO group, and biomarkers of lung injury, including soluble receptor for advanced glycation end products and surfactant protein-D, also significantly increased. There are more neutrophil and macrophage infiltrations in the IR-ECMO group as well. We also observed higher expression of inflammatory factors, including interleukin-6, interleukin-1β and tumour necrosis factor-α in the lung tissues and serum. CONCLUSIONS This study found VV ECMO significantly exacerbates lung ischaemia-reperfusion injury and pulmonary inflammatory response in a rat model after lung ischaemia-reperfusion.
Collapse
Affiliation(s)
- Shi-Lin Wei
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Xiao-Qian Zhang
- Department of Cardiac Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jian Li
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Rong-Zhi Zhang
- Department of Anesthesiology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Jian-Bao Yang
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Ran Zhang
- Department of Cardiovascular Medicine, Chinese PLA General Hospital & Chinese PLA Medical School, Beijing, China
| | - Yong-Nan Li
- Department of Cardiac Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Bin Li
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| |
Collapse
|
3
|
Walsh D, Kostyunina DS, Blake A, Boylan J, McLoughlin P. Shear stress-induced restoration of pulmonary microvascular endothelial barrier function following ischemia reperfusion injury requires VEGFR2 signaling. Am J Physiol Lung Cell Mol Physiol 2025; 328:L389-L404. [PMID: 39701597 DOI: 10.1152/ajplung.00200.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
Normal shear stress produced by blood flow is sensed by the vascular endothelium and required for maintenance of the homeostatic functions of the endothelium in systemic conduit and resistance vessels. Many critical illnesses are characterized by periods of abnormally reduced or absent shear stress in the lung (e.g., hemorrhagic shock, embolism, ischemia reperfusion injury, and lung transplantation) and are complicated by pulmonary edema following reperfusion due to microvascular leak. The role of shear stress in regulating the pulmonary microvascular endothelial barrier in the intact vascular bed has not been previously examined. We tested the hypothesis that, in lungs injured by a period of ischemia and reperfusion (IRI), reduced shear stress contributes to increased pulmonary microvascular endothelial barrier permeability and edema formation. Furthermore, we examined the role of vascular endothelial-derived growth factor receptor 2 (VEGFR2) as a mechanosensor mediating the endothelial response to this altered shear stress. Following IRI, we perfused isolated ventilated mouse lungs with a low viscosity solution (LVS) or a higher, physiological viscosity solution (PVS) at constant flow to produce differing endothelial shear stresses in the intact microcirculation. Lungs perfused with LVS developed pulmonary edema due to increased endothelial permeability whereas those perfused with PVS were protected from edema formation by reduced endothelial permeability. This effect of PVS required normal VEGFR2 mechanoreceptor function. These data show for the first time that shear stress has an important role in restoring endothelial barrier function in the intact pulmonary microcirculation following injury and have important implications for the treatment of pulmonary edema in critically ill patients.NEW & NOTEWORTHY Critical illnesses are frequently complicated by noncardiogenic pulmonary edema. Many such illnesses include periods of reduced blood flow, often accompanied by hemodilution, which together reduce endothelial shear stress. We report that in ischemia-reperfusion injury reduced shear stress contributes to increased permeability of the in situ pulmonary microvascular endothelium and worsens alveolar edema. Restoring shear stress toward normal reduces endothelial permeability and edema formation, an effect that requires the normal mechanoreceptor function of VEGFR2.
Collapse
Affiliation(s)
- Don Walsh
- School of Medicine and Conway Institute, University College Dublin, Dublin, Ireland
| | - Daria S Kostyunina
- School of Medicine and Conway Institute, University College Dublin, Dublin, Ireland
| | - Aoife Blake
- School of Medicine and Conway Institute, University College Dublin, Dublin, Ireland
| | - John Boylan
- Department of Anaesthesia and Intensive Care, St. Vincent's University Hospital, Dublin, Ireland
| | - Paul McLoughlin
- School of Medicine and Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
4
|
Yeung JC, Koike T, Wagnetz D, Machuca TN, Bonato R, Liu M, Juvet S, Cypel M, Keshavjee S. Ex vivo delivery of recombinant IL-10 to human donor lungs. JHLT OPEN 2025; 7:100192. [PMID: 40144859 PMCID: PMC11935390 DOI: 10.1016/j.jhlto.2024.100192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Background The immunoregulatory cytokine interleukin-10 (IL-10) has been shown to be a promising therapy for donor lung injuries before transplantation. However, the very short half-life of IL-10 in vivo (∼2 hours) has necessitated the use of gene therapy in almost all animal models of lung transplantation. Because isolation of the donor lung on the ex vivo lung perfusion (EVLP) circuit removes it from the influence of renal and hepatic clearance mechanisms, a much-prolonged half-life of IL-10 is anticipated. Thus, we hypothesized that delivery of recombinant IL-10 (rIL-10) to injured donor lungs isolated on EVLP could be a clinically relevant and a logistically simpler method of employing IL-10 therapy in lung transplantation. Methods Injured human donor lungs clinically rejected for transplantation were split into single lungs and the better of the 2 subjected to 12 hours of EVLP and randomized (n = 5/group) to receive either saline (control), rIL-10 (5 µg in 2-liter perfusate), or rIL-10 (25 µg) aerosolized into the airways. Results Perfusate and intratracheal delivery of rIL-10 did not provide the therapeutic anti-inflammatory action that has been traditionally achieved with gene therapy. It appears that intratracheally delivered rIL-10 moves into the perfusate where it seems to be biologically inactive. Conclusions Gene therapy remains superior as it allows for continued production of IL-10 within the alveoli where it has the potential to continuously act on alveolar macrophages and epithelial cells in a paracrine fashion.
Collapse
Affiliation(s)
- Jonathan C. Yeung
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Terumoto Koike
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Dirk Wagnetz
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Tiago N. Machuca
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Riccardo Bonato
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Stephen Juvet
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Laboratories, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Wong A, Duong A, Wilson G, Yeung J, MacParland S, Han H, Cypel M, Keshavjee S, Liu M. Ischemia-reperfusion responses in human lung transplants at the single-cell resolution. Am J Transplant 2024; 24:2199-2211. [PMID: 39197591 DOI: 10.1016/j.ajt.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/01/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Ischemia-reperfusion is an unavoidable step of organ transplantation. Development of therapeutics for lung injury during transplantation has proved challenging; understanding lung injury from human data at the single-cell resolution is required to accelerate the development of therapeutics. Donor lung biopsies from 6 human lung transplant cases were collected at the end of cold preservation and 2-hour reperfusion and underwent single-cell RNA sequencing. Donor and recipient origin of cells from the reperfusion timepoint were deconvolved. Gene expression profiles were: (1) compared between each donor cell type between timepoints and (2) compared between donor and recipient cells. Inflammatory responses from donor lung macrophages were found after reperfusion with upregulation of multiple cytokines and chemokines, especially IL-1β and IL-1α. Significant inflammatory responses were found in alveolar epithelial cells (featured by CXCL8) and lung endothelial cells (featured by IL-6 upregulation). Different inflammatory responses were noted between donor and recipient monocytes and CD8+ T cells. The inflammatory signals and differences between donor and recipient cells observed provide insight into the cellular and molecular mechanisms of ischemia-reperfusion induced lung injury. Further investigations may lead to the development of novel targeted therapeutics.
Collapse
Affiliation(s)
- Aaron Wong
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Allen Duong
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Gavin Wilson
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Yeung
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sonya MacParland
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hong Han
- Centre for Discovery in Cancer Research and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
6
|
Leroy V, Manual Kollareth DJ, Tu Z, Valisno JAC, Woolet-Stockton M, Saha B, Emtiazjoo AM, Rackauskas M, Moldawer LL, Efron PA, Cai G, Atkinson C, Upchurch GR, Sharma AK. MerTK-dependent efferocytosis by monocytic-MDSCs mediates resolution of ischemia/reperfusion injury after lung transplant. JCI Insight 2024; 9:e179876. [PMID: 39172530 PMCID: PMC11466183 DOI: 10.1172/jci.insight.179876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024] Open
Abstract
Lung transplantation (LTx) outcomes are impeded by ischemia/reperfusion injury (IRI) and subsequent chronic lung allograft dysfunction (CLAD). We examined the undefined role of receptor Mer tyrosine kinase (MerTK) on monocytic myeloid-derived suppressor cells (M-MDSCs) in efferocytosis to facilitate resolution of lung IRI. Single-cell RNA sequencing of lung tissue and bronchoalveolar lavage (BAL) from patients after LTx were analyzed. Murine lung hilar ligation and allogeneic orthotopic LTx models of IRI were used with BALB/c (WT), Cebpb-/- (MDSC-deficient), Mertk-/-, or MerTK-cleavage-resistant mice. A significant downregulation in MerTK-related efferocytosis genes in M-MDSC populations of patients with CLAD was observed compared with healthy individuals. In the murine IRI model, a significant increase in M-MDSCs, MerTK expression, and efferocytosis and attenuation of lung dysfunction was observed in WT mice during injury resolution that was absent in Cebpb-/- and Mertk-/- mice. Adoptive transfer of M-MDSCs in Cebpb-/- mice significantly attenuated lung dysfunction and inflammation. Additionally, in a murine orthotopic LTx model, increases in M-MDSCs were associated with resolution of lung IRI in the transplant recipients. In vitro studies demonstrated the ability of M-MDSCs to efferocytose apoptotic neutrophils in a MerTK-dependent manner. Our results suggest that MerTK-dependent efferocytosis by M-MDSCs can substantially contribute to the resolution of post-LTx IRI.
Collapse
Affiliation(s)
- Victoria Leroy
- Department of Surgery
- Department of Pharmacology and Therapeutics
| | | | - Zhenxiao Tu
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | | | - Biplab Saha
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Amir M. Emtiazjoo
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | | | | | - Guoshuai Cai
- Department of Surgery
- Department of Biostatistics, College of Public Health and Health Professions and College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Carl Atkinson
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | - Ashish K. Sharma
- Department of Surgery
- Department of Pharmacology and Therapeutics
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
7
|
Dai P, He J, Wei Y, Xu M, Zhao J, Zhou X, Tang H. High Dose of Estrogen Protects the Lungs from Ischemia-Reperfusion Injury by Downregulating the Angiotensin II Signaling Pathway. Inflammation 2024; 47:1248-1261. [PMID: 38386131 DOI: 10.1007/s10753-024-01973-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/21/2023] [Accepted: 01/09/2024] [Indexed: 02/23/2024]
Abstract
We explored the sex difference in lung ischemia-reperfusion injury (LIRI) and the role and mechanism of estrogen (E2) and angiotensin II (Ang II) in LIRI. We established a model of LIRI in mice. E2, Ang II, E2 inhibitor (fulvestrant), and angiotensin II receptor blocker (losartan) were grouped for treatment. The lung wet/dry weight ratio, natural killer (NK) cells (by flow cytometry), neutrophils (by flow cytometry), expression of key proteins (by Western blot, immunohistochemistry, ELISA, and immunofluorescence), and expression of related protein mRNA (by qPCR) were detected. The ultrastructure of the alveolar epithelial cells was observed by transmission electron microscopy. We found that E2 and Ang II played an important role in the progression of LIRI. The two signaling pathways showed obvious antagonism, and E2 regulates LIRI in the different sexes by downregulating Ang II, leading to a better prognosis. E2 and losartan reduced the inflammatory cell infiltration in lung tissue and key inflammatory factors in serum while fulvestrant and Ang II had the opposite effect. The protective effect of E2 was related with AKT, p38, COX2, and HIF-1α.
Collapse
Affiliation(s)
- Peng Dai
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jutong He
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yanhong Wei
- Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ming Xu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jinping Zhao
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Xuefeng Zhou
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Hexiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
8
|
Yu J, Fu Y, Gao J, Zhang Q, Zhang N, Zhang Z, Jiang X, Chen C, Wen Z. Cathepsin C from extracellular histone-induced M1 alveolar macrophages promotes NETosis during lung ischemia-reperfusion injury. Redox Biol 2024; 74:103231. [PMID: 38861835 PMCID: PMC11209641 DOI: 10.1016/j.redox.2024.103231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024] Open
Abstract
Primary graft dysfunction (PGD) is a severe form of acute lung injury resulting from lung ischemia/reperfusion injury (I/R) in lung transplantation (LTx), associated with elevated post-transplant morbidity and mortality rates. Neutrophils infiltrating during reperfusion are identified as pivotal contributors to lung I/R injury by releasing excessive neutrophil extracellular traps (NETs) via NETosis. While alveolar macrophages (AMs) are involved in regulating neutrophil chemotaxis and infiltration, their role in NETosis during lung I/R remains inadequately elucidated. Extracellular histones constitute the main structure of NETs and can activate AMs. In this study, we confirmed the significant involvement of extracellular histone-induced M1 phenotype of AMs (M1-AMs) in driving NETosis during lung I/R. Using secretome analysis, public protein databases, and transwell co-culture models of AMs and neutrophils, we identified Cathepsin C (CTSC) derived from AMs as a major mediator in NETosis. Further elucidating the molecular mechanisms, we found that CTSC induced NETosis through a pathway dependent on NADPH oxidase-mediated production of reactive oxygen species (ROS). CTSC could significantly activate p38 MAPK, resulting in the phosphorylation of the NADPH oxidase subunit p47phox, thereby facilitating the trafficking of cytoplasmic subunits to the cell membrane and activating NADPH oxidase. Moreover, CTSC up-regulated and activated its substrate membrane proteinase 3 (mPR3), resulting in an increased release of NETosis-related inflammatory factors. Inhibiting CTSC revealed great potential in mitigating NETosis-related injury during lung I/R. These findings suggests that CTSC from AMs may be a crucial factor in mediating NETosis during lung I/R, and targeting CTSC inhition may represent a novel intervention for PGD in LTx.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Yu Fu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiameng Gao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingqing Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Nan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemei Jiang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
9
|
Nykänen AI, Mariscal A, Duong A, Ali A, Takahagi A, Bai X, Zehong G, Joe B, Takahashi M, Chen M, Gokhale H, Shan H, Hwang DM, Estrada C, Yeung J, Waddell T, Martinu T, Juvet S, Cypel M, Liu M, Davies JE, Keshavjee S. Lung Transplant Immunomodulation with Genetically Engineered Mesenchymal Stromal Cells-Therapeutic Window for Interleukin-10. Cells 2024; 13:859. [PMID: 38786082 PMCID: PMC11119666 DOI: 10.3390/cells13100859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Lung transplantation results are compromised by ischemia-reperfusion injury and alloimmune responses. Ex vivo lung perfusion (EVLP) is used to assess marginal donor lungs before transplantation but is also an excellent platform to apply novel therapeutics. We investigated donor lung immunomodulation using genetically engineered mesenchymal stromal cells with augmented production of human anti-inflammatory hIL-10 (MSCsIL-10). Pig lungs were placed on EVLP for 6 h and randomized to control (n = 7), intravascular delivery of 20 × 106 (n = 5, low dose) or 40 × 106 human MSCs IL-10 (n = 6, high dose). Subsequently, single-lung transplantation was performed, and recipient pigs were monitored for 3 days. hIL-10 secretion was measured during EVLP and after transplantation, and immunological effects were assessed by cytokine profile, T and myeloid cell characterization and mixed lymphocyte reaction. MSCIL-10 therapy rapidly increased hIL-10 during EVLP and resulted in transient hIL-10 elevation after lung transplantation. MSCIL-10 delivery did not affect lung function but was associated with dose-related immunomodulatory effects, with the low dose resulting in a beneficial decrease in apoptosis and lower macrophage activation, but the high MSCIL-10 dose resulting in inflammation and cytotoxic CD8+ T cell activation. MSCIL-10 therapy during EVLP results in a rapid and transient perioperative hIL-10 increase and has a therapeutic window for its immunomodulatory effects.
Collapse
Affiliation(s)
- Antti I. Nykänen
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Andrea Mariscal
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Thoracic Surgery, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Allen Duong
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Aadil Ali
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Akihiro Takahagi
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
| | - Xiaohui Bai
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
| | - Guan Zehong
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
| | - Betty Joe
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
| | - Mamoru Takahashi
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
| | - Manyin Chen
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Hemant Gokhale
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Hongchao Shan
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON M5G 2N2, Canada
| | - David M. Hwang
- Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada;
| | - Catalina Estrada
- Tissue Regeneration Therapeutics, Toronto, ON M5G 1N8, Canada; (C.E.); (J.E.D.)
| | - Jonathan Yeung
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Thoracic Surgery, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Tom Waddell
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Thoracic Surgery, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Tereza Martinu
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Thoracic Surgery, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Stephen Juvet
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Thoracic Surgery, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
- Division of Respirology, Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Marcelo Cypel
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Thoracic Surgery, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Mingyao Liu
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - John E. Davies
- Tissue Regeneration Therapeutics, Toronto, ON M5G 1N8, Canada; (C.E.); (J.E.D.)
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Shaf Keshavjee
- Latner Thoracic Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (A.I.N.); (A.M.); (A.D.); (A.A.); (A.T.); (X.B.); (G.Z.); (B.J.); (M.T.); (M.C.); (H.G.); (H.S.); (J.Y.); (T.W.); (T.M.); (S.J.); (M.C.); (M.L.)
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Thoracic Surgery, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1P5, Canada
- Toronto Lung Transplant Program, Ajmera Transplant Centre, University Health Network, Toronto, ON M5G 2N2, Canada
| |
Collapse
|
10
|
Osorio-Valencia S, Zhou B. Roles of Macrophages and Endothelial Cells and Their Crosstalk in Acute Lung Injury. Biomedicines 2024; 12:632. [PMID: 38540245 PMCID: PMC10968255 DOI: 10.3390/biomedicines12030632] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/08/2024] [Accepted: 03/10/2024] [Indexed: 11/11/2024] Open
Abstract
Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), present life-threatening conditions characterized by inflammation and endothelial injury, leading to increased vascular permeability and lung edema. Key players in the pathogenesis and resolution of ALI are macrophages (Mφs) and endothelial cells (ECs). The crosstalk between these two cell types has emerged as a significant focus for potential therapeutic interventions in ALI. This review provides a brief overview of the roles of Mφs and ECs and their interplay in ALI/ARDS. Moreover, it highlights the significance of investigating perivascular macrophages (PVMs) and immunomodulatory endothelial cells (IMECs) as crucial participants in the Mφ-EC crosstalk. This sheds light on the pathogenesis of ALI and paves the way for innovative treatment approaches.
Collapse
Affiliation(s)
| | - Bisheng Zhou
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| |
Collapse
|
11
|
Li Q, Nie H. Advances in lung ischemia/reperfusion injury: unraveling the role of innate immunity. Inflamm Res 2024; 73:393-405. [PMID: 38265687 DOI: 10.1007/s00011-023-01844-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/03/2023] [Accepted: 12/18/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Lung ischemia/reperfusion injury (LIRI) is a common occurrence in clinical practice and represents a significant complication following pulmonary transplantation and various diseases. At the core of pulmonary ischemia/reperfusion injury lies sterile inflammation, where the innate immune response plays a pivotal role. This review aims to investigate recent advancements in comprehending the role of innate immunity in LIRI. METHODS A computer-based online search was performed using the PubMed database and Web of Science database for published articles concerning lung ischemia/reperfusion injury, cell death, damage-associated molecular pattern molecules (DAMPs), innate immune cells, innate immunity, inflammation. RESULTS During the process of lung ischemia/reperfusion, cellular injury even death can occur. When cells are injured or undergo cell death, endogenous ligands known as DAMPs are released. These molecules can be recognized and bound by pattern recognition receptors (PRRs), leading to the recruitment and activation of innate immune cells. Subsequently, a cascade of inflammatory responses is triggered, ultimately exacerbating pulmonary injury. These steps are complex and interrelated rather than being in a linear relationship. In recent years, significant progress has been made in understanding the immunological mechanisms of LIRI, involving novel types of cell death, the ability of receptors other than PRRs to recognize DAMPs, and a more detailed mechanism of action of innate immune cells in ischemia/reperfusion injury (IRI), laying the groundwork for the development of novel diagnostic and therapeutic approaches. CONCLUSIONS Various immune components of the innate immune system play critical roles in lung injury after ischemia/reperfusion. Preventing cell death and the release of DAMPs, interrupting DAMPs receptor interactions, disrupting intracellular inflammatory signaling pathways, and minimizing immune cell recruitment are essential for lung protection in LIRI.
Collapse
Affiliation(s)
- Qingqing Li
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China
| | - Hanxiang Nie
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang District, Wuhan, 430060, China.
| |
Collapse
|
12
|
Ren J, Zhu F, Sang D, Cong M, Jiang S. The Protective Effect and Mechanism of Mild Hypothermia on Lung Injury after Cardiopulmonary Resuscitation in Pigs. Crit Rev Immunol 2024; 44:51-58. [PMID: 38618728 DOI: 10.1615/critrevimmunol.2024052420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
To explore the protective effect and mechanism of mild hypothermia on lung tissue damage after cardiopulmonary resuscitation in pigs. In this experiment, we electrically stimulated 16 pigs (30 ± 2 kg) for 10 min to cause ventricular fibrillation. The successfully resuscitated animals were randomly divided into two groups, a mild hypothermia group and a control group. We took arterial blood 0.5, 1, 3, and 6 h after ROSC recovery in the two groups of animals for blood gas analysis. We observed the structural changes of lung tissue under an electron microscope and calculate the wet weight/dry weight (W/D) ratio. We quantitatively analyzed the expression differences of representative inflammatory factors [interleukin-6 (IL-6) and tumor necrosis factor-alpha TNF-α)] through the ELISA test. We detected the expression levels of Bax, Bcl-2, and Caspase-3 proteins in lung tissues by Western blot. After 3 h and 6 h of spontaneous circulation was restored, compared with the control group, PaO2/FiO2 decreased significantly (P < 0.05). In addition, the pathological changes, lung W/D and lung MDA of the mild hypothermia group were better than those of the control group. The levels of IL-6 and TNF-α in the lung tissue of the mild hypothermia group were significantly lower than those of the control group (P < 0.05). The content of Caspase-3 and Bax in the mild hypothermia group was significantly lower than that of the control group. Our experiments have shown that mild hypothermia can reduce lung tissue damage after cardiopulmonary resuscitation.
Collapse
Affiliation(s)
- Jinlin Ren
- Department of Emergency, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250000, Shandong, China
| | - Fangfang Zhu
- General Practice, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250000, Shandong, China
| | - Dongdong Sang
- Department of Emergency, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250000, Shandong, China
| | - Mulin Cong
- Department of Emergency, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250000, Shandong, China
| | - Shujuan Jiang
- Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University
| |
Collapse
|
13
|
Kuppusamy M, Ta HQ, Davenport HN, Bazaz A, Kulshrestha A, Daneva Z, Chen YL, Carrott PW, Laubach VE, Sonkusare SK. Purinergic P2Y2 receptor-induced activation of endothelial TRPV4 channels mediates lung ischemia-reperfusion injury. Sci Signal 2023; 16:eadg1553. [PMID: 37874885 PMCID: PMC10683978 DOI: 10.1126/scisignal.adg1553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 10/02/2023] [Indexed: 10/26/2023]
Abstract
Lung ischemia-reperfusion injury (IRI), characterized by inflammation, vascular permeability, and lung edema, is the major cause of primary graft dysfunction after lung transplantation. Here, we investigated the cellular mechanisms underlying lung IR-induced activation of endothelial TRPV4 channels, which play a central role in lung edema and dysfunction after IR. In a left lung hilar-ligation model of IRI in mice, we found that lung IRI increased the efflux of ATP through pannexin 1 (Panx1) channels at the endothelial cell (EC) membrane. Elevated extracellular ATP activated Ca2+ influx through endothelial TRPV4 channels downstream of purinergic P2Y2 receptor (P2Y2R) signaling. P2Y2R-dependent activation of TRPV4 channels was also observed in human and mouse pulmonary microvascular endothelium in ex vivo and in vitro models of IR. Endothelium-specific deletion of P2Y2R, TRPV4, or Panx1 in mice substantially prevented lung IRI-induced activation of endothelial TRPV4 channels and lung edema, inflammation, and dysfunction. These results identify endothelial P2Y2R as a mediator of the pathological sequelae of IRI in the lung and show that disruption of the endothelial Panx1-P2Y2R-TRPV4 signaling pathway could be a promising therapeutic strategy for preventing lung IRI after transplantation.
Collapse
Affiliation(s)
- Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Huy Q. Ta
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Hannah N. Davenport
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Abhishek Bazaz
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Astha Kulshrestha
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Zdravka Daneva
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Yen-Lin Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Philip W. Carrott
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Victor E. Laubach
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Swapnil K. Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
14
|
Chacon-Alberty L, Fernandez R, Jindra P, King M, Rosas I, Hochman-Mendez C, Loor G. Primary Graft Dysfunction in Lung Transplantation: A Review of Mechanisms and Future Applications. Transplantation 2023; 107:1687-1697. [PMID: 36650643 DOI: 10.1097/tp.0000000000004503] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Lung allograft recipients have worse survival than all other solid organ transplant recipients, largely because of primary graft dysfunction (PGD), a major form of acute lung injury affecting a third of lung recipients within the first 72 h after transplant. PGD is the clinical manifestation of ischemia-reperfusion injury and represents the predominate cause of early morbidity and mortality. Despite PGD's impact on lung transplant outcomes, no targeted therapies are currently available; hence, care remains supportive and largely ineffective. This review focuses on molecular and innate immune mechanisms of ischemia-reperfusion injury leading to PGD. We also discuss novel research aimed at discovering biomarkers that could better predict PGD and potential targeted interventions that may improve outcomes in lung transplantation.
Collapse
Affiliation(s)
| | - Ramiro Fernandez
- Division of Cardiothoracic Transplantation and Mechanical Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Peter Jindra
- Division of Cardiothoracic Transplantation and Mechanical Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Madelyn King
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, TX
| | - Ivan Rosas
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | | | - Gabriel Loor
- Division of Cardiothoracic Transplantation and Mechanical Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
- Cardiothoracic Surgery Professional Staff, The Texas Heart Institute, Houston, TX
| |
Collapse
|
15
|
Britt RD, Ruwanpathirana A, Ford ML, Lewis BW. Macrophages Orchestrate Airway Inflammation, Remodeling, and Resolution in Asthma. Int J Mol Sci 2023; 24:10451. [PMID: 37445635 PMCID: PMC10341920 DOI: 10.3390/ijms241310451] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Asthma is a heterogenous chronic inflammatory lung disease with endotypes that manifest different immune system profiles, severity, and responses to current therapies. Regardless of endotype, asthma features increased immune cell infiltration, inflammatory cytokine release, and airway remodeling. Lung macrophages are also heterogenous in that there are separate subsets and, depending on the environment, different effector functions. Lung macrophages are important in recruitment of immune cells such as eosinophils, neutrophils, and monocytes that enhance allergic inflammation and initiate T helper cell responses. Persistent lung remodeling including mucus hypersecretion, increased airway smooth muscle mass, and airway fibrosis contributes to progressive lung function decline that is insensitive to current asthma treatments. Macrophages secrete inflammatory mediators that induce airway inflammation and remodeling. Additionally, lung macrophages are instrumental in protecting against pathogens and play a critical role in resolution of inflammation and return to homeostasis. This review summarizes current literature detailing the roles and existing knowledge gaps for macrophages as key inflammatory orchestrators in asthma pathogenesis. We also raise the idea that modulating inflammatory responses in lung macrophages is important for alleviating asthma.
Collapse
Affiliation(s)
- Rodney D Britt
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Anushka Ruwanpathirana
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215, USA
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH 43205, USA
| | - Maria L Ford
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215, USA
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH 43205, USA
| | - Brandon W Lewis
- Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43215, USA
| |
Collapse
|
16
|
Kuppusamy M, Ta HQ, Davenport HN, Bazaz A, Kulshrestha A, Daneva Z, Chen YL, Carrott PW, Laubach VE, Sonkusare SK. Purinergic P2Y2 Receptor-Induced Activation of Endothelial TRPV4 Channels Mediates Lung Ischemia-Reperfusion Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.29.542520. [PMID: 37397979 PMCID: PMC10312453 DOI: 10.1101/2023.05.29.542520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Lung ischemia-reperfusion injury (IRI), characterized by inflammation, vascular permeability, and lung edema, is the major cause of primary graft dysfunction after lung transplantation. We recently reported that endothelial cell (EC) TRPV4 channels play a central role in lung edema and dysfunction after IR. However, the cellular mechanisms for lung IR-induced activation of endothelial TRPV4 channels are unknown. In a left-lung hilar ligation model of IRI in mice, we found that lung IR increases the efflux of extracellular ATP (eATP) through pannexin 1 (Panx1) channels at the EC membrane. Elevated eATP activated elementary Ca2+ influx signals through endothelial TRPV4 channels through purinergic P2Y2 receptor (P2Y2R) signaling. P2Y2R-dependent activation of TRPV4 channels was also observed in human and mouse pulmonary microvascular endothelium in ex vivo and in vitro surrogate models of lung IR. Endothelium-specific deletion of P2Y2R, TRPV4, and Panx1 in mice had substantial protective effects against lung IR-induced activation of endothelial TRPV4 channels, lung edema, inflammation, and dysfunction. These results identify endothelial P2Y2R as a novel mediator of lung edema, inflammation, and dysfunction after IR, and show that disruption of endothelial Panx1-P2Y2R-TRPV4 signaling pathway could represent a promising therapeutic strategy for preventing lung IRI after transplantation.
Collapse
Affiliation(s)
- Maniselvan Kuppusamy
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Huy Q. Ta
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Hannah N. Davenport
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Abhishek Bazaz
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Astha Kulshrestha
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Zdravka Daneva
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Yen-Lin Chen
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
| | - Philip W. Carrott
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Victor E. Laubach
- Department of Surgery, University of Virginia, Charlottesville, VA 22908
| | - Swapnil K. Sonkusare
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
17
|
Billah M, Naz A, Noor R, Bhindi R, Khachigian LM. Early Growth Response-1: Friend or Foe in the Heart? Heart Lung Circ 2023; 32:e23-e35. [PMID: 37024319 DOI: 10.1016/j.hlc.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 04/07/2023]
Abstract
Cardiovascular disease is a major cause of mortality and morbidity worldwide. Early growth response-1 (Egr-1) plays a critical regulatory role in a range of experimental models of cardiovascular diseases. Egr-1 is an immediate-early gene and is upregulated by various stimuli including shear stress, oxygen deprivation, oxidative stress and nutrient deprivation. However, recent research suggests a new, underexplored cardioprotective side of Egr-1. The main purpose of this review is to explore and summarise the dual nature of Egr-1 in cardiovascular pathobiology.
Collapse
Affiliation(s)
- Muntasir Billah
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, Sydney, NSW, Australia; Sydney Medical School Northern, The University of Sydney, Sydney, NSW, Australia.
| | - Adiba Naz
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Rashed Noor
- School of Environmental and Life Sciences, Independent University Bangladesh, Dhaka, Bangladesh
| | - Ravinay Bhindi
- Department of Cardiology, Kolling Institute of Medical Research, Northern Sydney Local Health District, Sydney, NSW, Australia; Sydney Medical School Northern, The University of Sydney, Sydney, NSW, Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
18
|
Yamanashi K, Ohsumi A, Oda H, Tanaka S, Yamada Y, Nakajima D, Date H. Reduction of donor mononuclear phagocytes with clodronate-liposome during ex vivo lung perfusion attenuates ischemia-reperfusion injury. J Thorac Cardiovasc Surg 2023; 165:e181-e203. [PMID: 36404143 DOI: 10.1016/j.jtcvs.2022.10.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Clodronate-liposome is used for depleting mononuclear phagocytes associated with ischemia-reperfusion injury. We hypothesized that administration of clodronate-liposome into the perfusate during ex vivo lung perfusion could reduce mononuclear phagocytes and attenuate ischemia-reperfusion injury. METHODS First, the number of mononuclear phagocytes in flushed grafts (minimum cold ischemic time, 6-hour cold ischemic time, 15-hour cold ischemic time, and 18-hour cold ischemic time; n = 6 each) was determined using flow cytometry. Second, grafts (15-hour cold ischemic time) were allocated to control or clodronate (n = 5 each). In the clodronate group, clodronate-liposome is administered into the perfusate. After 4 hours of ex vivo lung perfusion, the number of mononuclear phagocytes in the perfusate and lung tissues was measured. Third, grafts (15-hour cold ischemic time) were allocated to control or clodronate (n = 6 each). After 4 hours of ex vivo lung perfusion, the left lungs were transplanted and reperfused for 2 hours. Lung function was evaluated, and samples were analyzed. RESULTS First, mononuclear phagocytes remain in flushed grafts after prolonged cold ischemia. Second, the number of mononuclear phagocytes in lung tissues after ex vivo lung perfusion was significantly reduced in the clodronate group (P = .008). Third, lung compliance and vascular resistance during ex vivo lung perfusion were significantly improved in the clodronate group (P < .001 for both). Blood oxygenation and pulmonary edema were significantly improved in the clodronate group after 2 hours of reperfusion (P = .015 and P = .026, respectively). Histological findings showed reduced lung injury in the clodronate group (P = .013). CONCLUSIONS Administration of clodronate-liposome into the perfusate during ex vivo lung perfusion resulted in a significant reduction of mononuclear phagocytes in donor lungs, leading to attenuation of ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Keiji Yamanashi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihiro Ohsumi
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Hiromi Oda
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satona Tanaka
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshito Yamada
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Daisuke Nakajima
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
19
|
Zhu W, Zhang Y, Wang Y. Immunotherapy strategies and prospects for acute lung injury: Focus on immune cells and cytokines. Front Pharmacol 2022; 13:1103309. [PMID: 36618910 PMCID: PMC9815466 DOI: 10.3389/fphar.2022.1103309] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a disastrous condition, which can be caused by a wide range of diseases, such as pneumonia, sepsis, traumas, and the most recent, COVID-19. Even though we have gained an improved understanding of acute lung injury/acute respiratory distress syndrome pathogenesis and treatment mechanism, there is still no effective treatment for acute lung injury/acute respiratory distress syndrome, which is partly responsible for the unacceptable mortality rate. In the pathogenesis of acute lung injury, the inflammatory storm is the main pathological feature. More and more evidences show that immune cells and cytokines secreted by immune cells play an irreplaceable role in the pathogenesis of acute lung injury. Therefore, here we mainly reviewed the role of various immune cells in acute lung injury from the perspective of immunotherapy, and elaborated the crosstalk of immune cells and cytokines, aiming to provide novel ideas and targets for the treatment of acute lung injury.
Collapse
Affiliation(s)
- Wenfang Zhu
- Department of Respiratory Medicine, Anhui Chest Hospital, Hefei, China
| | - Yiwen Zhang
- Department of Respiratory Medicine, Anhui Chest Hospital, Hefei, China,*Correspondence: Yiwen Zhang, ; Yinghong Wang,
| | - Yinghong Wang
- Department of Pharmacy, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China,*Correspondence: Yiwen Zhang, ; Yinghong Wang,
| |
Collapse
|
20
|
Leroy V, Cai J, Tu Z, McQuiston A, Sharma S, Emtiazjoo A, Atkinson C, Upchurch GR, Sharma AK. Resolution of post-lung transplant ischemia-reperfusion injury is modulated via Resolvin D1-FPR2 and Maresin 1-LGR6 signaling. J Heart Lung Transplant 2022; 42:562-574. [PMID: 36628837 DOI: 10.1016/j.healun.2022.12.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Dysregulation of inflammation-resolution pathways leads to postlung transplant (LTx) ischemia-reperfusion (IR) injury and allograft dysfunction. Our hypothesis is that combined treatment with specialized pro-resolving lipid mediators, that is, Resolvin D1 (RvD1) and Maresin-1 (MaR1), enhances inflammation-resolution of lung IR injury. METHODS Expression of RvD1 and MaR1 was analyzed in bronchoalveolar lavage (BAL) fluid of patients on days 0, 1, and 7 post-LTx. Lung IR injury was evaluated in C57BL/6 (WT), FPR2-/-, and LGR6 siRNA treated mice using a hilar-ligation model with or without administration with RvD1 and/or MaR1. A donation after circulatory death and murine orthotopic lung transplantation model was used to evaluate the protection by RvD1 and MaR1 against lung IR injury. In vitro studies analyzed alveolar macrophages and type II epithelial cell activation after treatment with RvD1 or MaR1. RESULTS RvD1 and MaR1 expressions in BAL from post-LTx patients was significantly increased on day 7 compared to days 0 and 1. Concomitant RvD1 and MaR1 treatment significantly mitigated early pulmonary inflammation and lung IR injury in WT mice, which was regulated via FPR2 and LGR6 receptors. In the murine orthotopic donation after cardiac death LTx model, RvD1 and MaR1 treatments significantly attenuated lung IR injury and increased PaO2 levels compared to saline-treated controls. Mechanistically, RvD1/FPR2 signaling on alveolar macrophages attenuated HMGB1 and TNF-α secretion and upregulated uptake of macrophage-dependent apoptotic neutrophils (efferocytosis), whereas MaR1/LGR6 signaling mitigated CXCL1 secretion by epithelial cells. CONCLUSIONS Bioactive proresolving lipid mediator-dependent signaling that is, RvD1/FPR2 and MaR1/LGR6- offers a novel therapeutic strategy in post-LTx injury.
Collapse
Affiliation(s)
- Victoria Leroy
- Department of Surgery, University of Florida, Gainesville, Florida
| | - Jun Cai
- Department of Surgery, University of Florida, Gainesville, Florida
| | - Zhenxiao Tu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Florida, Gainesville, Florida
| | - Alexander McQuiston
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Florida, Gainesville, Florida
| | - Simrun Sharma
- Department of Surgery, University of Florida, Gainesville, Florida
| | - Amir Emtiazjoo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Florida, Gainesville, Florida
| | - Carl Atkinson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Florida, Gainesville, Florida
| | | | - Ashish K Sharma
- Department of Surgery, University of Florida, Gainesville, Florida; Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Florida, Gainesville, Florida.
| |
Collapse
|
21
|
Konlaan Y, Asamoah Sakyi S, Kumi Asare K, Amoah Barnie P, Opoku S, Nakotey GK, Victor Nuvor S, Amoani B. Evaluating immunohaematological profile among COVID-19 active infection and recovered patients in Ghana. PLoS One 2022; 17:e0273969. [PMID: 36094915 PMCID: PMC9467340 DOI: 10.1371/journal.pone.0273969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 08/18/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction The rapid spread of COVID-19 has been a global public health problem and it is yet to be put under control. Active COVID-19 is associated with unrestrained secretion of pro-inflammatory cytokines and imbalances in haematological profile including anaemia, leukocytosis and thrombocytopaenia. However, the haematological profile and immune status following recovery from COVID-19 has not been recognized. We evaluated the immunohaematological profile among COVID-19 patients with active infection, recovered cases and unexposed healthy individuals in the Ashanti region of Ghana. Methodology A total of 95 adult participants, consisting of 35 positive, 30 recovered and 30 unexposed COVID-19 negative individuals confirmed by RT-PCR were recruited for the study. All the patients had the complete blood count performed using the haematological analyzer Sysmex XN-1500. Their plasma cytokine levels of interleukin (IL)-1β, IL-6, IL-10, IL-17, tumour necrosis factor-alpha (TNF-α) and interferon gamma (IFN-γ) were analysed using ELISA. Statistical analyses were performed on R statistical software. Result The Patients with COVID-19 active infection had significantly higher levels of IL10 (181±6.14 pg/mL vs 155.00±14.32 pg/mL vs 158.80±11.70 pg/mL, p = 0.038), WBC count (5.5±0.4 x109 /L vs 4.5±0.6 x109 /L vs 3.8±0.5, p < 0.0001) and percentage basophil (1.8±0.1% vs 0.8±0.3% vs 0.7±0.2%, p = 0.0040) but significantly lower levels of IFN-γ (110.10±9.52 pg/mL vs 142.80±5.46 pg/mL vs 140.80±6.39 pg/mL, p = 0.021), haematocrit (24.1±3.7% vs 38.3± 3.0% vs 38.5±2.2%, p < 0.0001), haemoglobin concentration (9.4±0.1g/dl vs 12.5± 5.0g/dl vs 12.7±0.8, p < 0.0001) and MPV (9.8±0.2fL vs 11.1±0.5fL vs 11.6±0.3fL, p < 0.0001) compared to recovered and unexposed controls respectively. There were significant association between IL-1β & neutrophils (r = 0.42, p<0.05), IL-10 & WBC (r = 0.39, p<0.05), IL-10 & Basophils (r = -0.51, p<0.01), IL-17 & Neutrophil (r = 0.39, p<0.05) in the active COVID-19 cases. Conclusion COVID-19 active infection is associated with increased IL-10 and WBC with a concomitant decrease in IFN-γ and haemoglobin concentration. However, recovery from the disease is associated with immune recovery with appareantly normal haematological profile.
Collapse
Affiliation(s)
- Yatik Konlaan
- Department of Microbiology and Immunology, School of Medical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Samuel Asamoah Sakyi
- Department of Molecular Medicine, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Kwame Kumi Asare
- Department of Biomedical Sciences, College of Health and Allied Sciences, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Prince Amoah Barnie
- Department of Forensic Science, School of Biological Sciences, College of Agriculture and Natural Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Stephen Opoku
- Department of Molecular Medicine, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Gideon Kwesi Nakotey
- Department of Biomedical Sciences, College of Health and Allied Sciences, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Samuel Victor Nuvor
- Department of Microbiology and Immunology, School of Medical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Benjamin Amoani
- Department of Biomedical Sciences, College of Health and Allied Sciences, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
22
|
Nykänen AI, Mariscal A, Duong A, Estrada C, Ali A, Hough O, Sage A, Chao BT, Chen M, Gokhale H, Shan H, Bai X, Zehong G, Yeung J, Waddell T, Martinu T, Juvet S, Cypel M, Liu M, Davies JE, Keshavjee S. Engineered mesenchymal stromal cell therapy during human lung ex vivo lung perfusion is compromised by acidic lung microenvironment. Mol Ther Methods Clin Dev 2021; 23:184-197. [PMID: 34703841 PMCID: PMC8516994 DOI: 10.1016/j.omtm.2021.05.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 05/07/2021] [Indexed: 11/29/2022]
Abstract
Ex vivo lung perfusion (EVLP) is an excellent platform to apply novel therapeutics, such as gene and cell therapies, before lung transplantation. We investigated the concept of human donor lung engineering during EVLP by combining gene and cell therapies. Premodified cryopreserved mesenchymal stromal cells with augmented anti-inflammatory interleukin-10 production (MSCIL-10) were administered during EVLP to human lungs that had various degrees of underlying lung injury. Cryopreserved MSCIL-10 had excellent viability, and they immediately and efficiently elevated perfusate and lung tissue IL-10 levels during EVLP. However, MSCIL-10 function was compromised by the poor metabolic conditions present in the most damaged lungs. Similarly, exposing cultured MSCIL-10 to poor metabolic, and especially acidic, conditions decreased their IL-10 production. In conclusion, we found that "off-the-shelf" MSCIL-10 therapy of human lungs during EVLP is safe and feasible, and results in rapid IL-10 elevation, and that the acidic target-tissue microenvironment may compromise the efficacy of cell-based therapies.
Collapse
Affiliation(s)
- Antti I Nykänen
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Andrea Mariscal
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Allen Duong
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Catalina Estrada
- Tissue Regeneration Therapeutics, 790 Bay Street, Toronto, ON M5G 1N8, Canada
| | - Aadil Ali
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Olivia Hough
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Andrew Sage
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Bonnie T Chao
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Manyin Chen
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Hemant Gokhale
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Hongchao Shan
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Xiaohui Bai
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Guan Zehong
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Jonathan Yeung
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Tom Waddell
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Tereza Martinu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Stephen Juvet
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - John E Davies
- Institute of Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON M5S 3G9, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network and University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
- Institute of Biomedical Engineering, University of Toronto, 164 College St, Toronto, ON M5S 3G9, Canada
| |
Collapse
|
23
|
Abdelghany L, Zhang X, Kawabata T, Goto S, El-Mahdy N, Jingu K, Li TS. Nicaraven prevents the fast growth of inflamed tumors by an anti-inflammatory mechanism. Med Oncol 2021; 39:7. [PMID: 34761342 DOI: 10.1007/s12032-021-01602-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/21/2021] [Indexed: 11/25/2022]
Abstract
Inflammatory microenvironment is known to accelerate the progression of malignant tumors. We investigated the possible anti-inflammatory effect of nicaraven on slowing tumor growth. Tumor-bearing mice randomly received nicaraven injection (50 mg/kg daily, i.p, n = 8) or placebo treatment (n = 8) for 10 days, and then sacrificed for evaluations. Nicaraven administration effectively inhibited the fast growth of tumor, as a large tumor (> 1.0 g) developed finally in three of the eight mice received placebo treatment. Cytokines/chemokines array indicated that nicaraven reduced the levels of CXCL10 and SDF-1 in the tumor as well as the levels of IL-2 and MIP-2 in serum. Immunofluorescence staining showed that nicaraven significantly reduced the recruitment of macrophages and neutrophils in the tumor. Interestingly, western blot indicated that the expression of CD86, CD206, and NIMP-R14 was especially enhanced in the three large-size tumors, suggesting the potential role of nicaraven in preventing the hyper-inflammatory tumor microenvironment. Moreover, the expression of PARP-1 was downregulated, but the expression of phospho-p38 MAPK, phospho-MKK-3/6, and phospho-MSK-1 was upregulated in the large-size tumors, suggesting the involvement of p38 MAPK pathway in the anti-inflammatory effect of nicaraven. Taken together, our study suggests that nicaraven may effectively prevent the fast growth of inflamed tumors by an anti-inflammatory mechanism.
Collapse
Affiliation(s)
- Lina Abdelghany
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Xu Zhang
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Tsuyoshi Kawabata
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Shinji Goto
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Nageh El-Mahdy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Keiichi Jingu
- Department of Radiation Oncology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan. .,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| |
Collapse
|
24
|
Selo MA, Sake JA, Kim KJ, Ehrhardt C. In vitro and ex vivo models in inhalation biopharmaceutical research - advances, challenges and future perspectives. Adv Drug Deliv Rev 2021; 177:113862. [PMID: 34256080 DOI: 10.1016/j.addr.2021.113862] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/11/2022]
Abstract
Oral inhalation results in pulmonary drug targeting and thereby reduces systemic side effects, making it the preferred means of drug delivery for the treatment of respiratory disorders such as asthma, chronic obstructive pulmonary disease or cystic fibrosis. In addition, the high alveolar surface area, relatively low enzymatic activity and rich blood supply of the distal airspaces offer a promising pathway to the systemic circulation. This is particularly advantageous when a rapid onset of pharmacological action is desired or when the drug is suffering from stability issues or poor biopharmaceutical performance following oral administration. Several cell and tissue-based in vitro and ex vivo models have been developed over the years, with the intention to realistically mimic pulmonary biological barriers. It is the aim of this review to critically discuss the available models regarding their advantages and limitations and to elaborate further which biopharmaceutical questions can and cannot be answered using the existing models.
Collapse
|
25
|
Patel PM, Connolly MR, Coe TM, Calhoun A, Pollok F, Markmann JF, Burdorf L, Azimzadeh A, Madsen JC, Pierson RN. Minimizing Ischemia Reperfusion Injury in Xenotransplantation. Front Immunol 2021; 12:681504. [PMID: 34566955 PMCID: PMC8458821 DOI: 10.3389/fimmu.2021.681504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/12/2021] [Indexed: 12/21/2022] Open
Abstract
The recent dramatic advances in preventing "initial xenograft dysfunction" in pig-to-non-human primate heart transplantation achieved by minimizing ischemia suggests that ischemia reperfusion injury (IRI) plays an important role in cardiac xenotransplantation. Here we review the molecular, cellular, and immune mechanisms that characterize IRI and associated "primary graft dysfunction" in allotransplantation and consider how they correspond with "xeno-associated" injury mechanisms. Based on this analysis, we describe potential genetic modifications as well as novel technical strategies that may minimize IRI for heart and other organ xenografts and which could facilitate safe and effective clinical xenotransplantation.
Collapse
Affiliation(s)
- Parth M. Patel
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Margaret R. Connolly
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Taylor M. Coe
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Anthony Calhoun
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Franziska Pollok
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Anesthesiology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - James F. Markmann
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Transplantation, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Lars Burdorf
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Agnes Azimzadeh
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Joren C. Madsen
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Richard N. Pierson
- Department of Surgery, Center for Transplantation Sciences, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Department of Surgery, Division of Cardiac Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
26
|
Li L, Qu M, Yang L, Liu J, Wang Q, Zhong P, Zeng Y, Wang T, Xiao H, Liu D, Huang X, Wang J, Zhou J. Effects of Ultrashort Wave Therapy on Inflammation and Macrophage Polarization after Acute Lung Injury in Rats. Bioelectromagnetics 2021; 42:464-472. [PMID: 34130351 DOI: 10.1002/bem.22353] [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: 05/26/2020] [Revised: 03/25/2021] [Accepted: 05/27/2021] [Indexed: 02/05/2023]
Abstract
Acute lung injury (ALI) features dysregulated pulmonary inflammation. Ultrashort waves (USWs) exert anti-inflammatory effects but no studies have evaluated their activity in ALI. Herein, we used an in vivo lipopolysaccharide (LPS)-induced ALI model to investigate whether the anti-inflammatory activity of USWs is mediated by altering the polarization of M1 to M2 macrophages. Twenty-four male Sprague-Dawley rats were randomly divided into control, untreated ALI, and ALI treated with USW groups (n = 8 in each group). ALI was induced by intratracheal LPS instillation. Rats in the USW group were treated for 15 min at 0, 4, and 8 h after a single LPS intratracheal instillation. Histopathologic examination, wet/dry lung weight ratio, enzyme-linked immunosorbent assay, quantitative real-time polymerase chain reaction, and western blot analyses were performed to evaluate the degree of lung injury and to determine macrophage phenotypes. Histopathologic examination disclosed attenuation of ALI, with reduced alveolar hemorrhage and neutrophilic infiltration in the USW group. Serum levels of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were significantly decreased after USW therapy. Moreover, the messenger RNA (mRNA) expressions of TNF-α and IL-1β were significantly decreased in the USW group, whereas the mRNA expression of Arginase 1 (Arg1) and the protein expression of mannose receptor significantly increased in comparison with the untreated ALI group. We conclude that USW therapy may attenuate inflammation in LPS-induced ALI through the modulation of macrophage polarization. © 2021 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Lan Li
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Mengjian Qu
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Lu Yang
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Jing Liu
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Qian Wang
- Department of Rehabilitation, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Peirui Zhong
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Yahua Zeng
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Ting Wang
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Hao Xiao
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Danni Liu
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Xiarong Huang
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Jinling Wang
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| | - Jun Zhou
- Rehabilitation Medicine Center, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Department of Rehabilitation, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China.,Rehabilitation Laboratory, The First Affiliated Hospital of University of South China, Hengyang, People's Republic of China
| |
Collapse
|
27
|
Lung Transplantation, Pulmonary Endothelial Inflammation, and Ex-Situ Lung Perfusion: A Review. Cells 2021; 10:cells10061417. [PMID: 34200413 PMCID: PMC8229792 DOI: 10.3390/cells10061417] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/31/2022] Open
Abstract
Lung transplantation (LTx) is the gold standard treatment for end-stage lung disease; however, waitlist mortality remains high due to a shortage of suitable donor lungs. Organ quality can be compromised by lung ischemic reperfusion injury (LIRI). LIRI causes pulmonary endothelial inflammation and may lead to primary graft dysfunction (PGD). PGD is a significant cause of morbidity and mortality post-LTx. Research into preservation strategies that decrease the risk of LIRI and PGD is needed, and ex-situ lung perfusion (ESLP) is the foremost technological advancement in this field. This review addresses three major topics in the field of LTx: first, we review the clinical manifestation of LIRI post-LTx; second, we discuss the pathophysiology of LIRI that leads to pulmonary endothelial inflammation and PGD; and third, we present the role of ESLP as a therapeutic vehicle to mitigate this physiologic insult, increase the rates of donor organ utilization, and improve patient outcomes.
Collapse
|
28
|
Natalini JG, Diamond JM. Primary Graft Dysfunction. Semin Respir Crit Care Med 2021; 42:368-379. [PMID: 34030200 DOI: 10.1055/s-0041-1728794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2022]
Abstract
Primary graft dysfunction (PGD) is a form of acute lung injury after transplantation characterized by hypoxemia and the development of alveolar infiltrates on chest radiograph that occurs within 72 hours of reperfusion. PGD is among the most common early complications following lung transplantation and significantly contributes to increased short-term morbidity and mortality. In addition, severe PGD has been associated with higher 90-day and 1-year mortality rates compared with absent or less severe PGD and is a significant risk factor for the subsequent development of chronic lung allograft dysfunction. The International Society for Heart and Lung Transplantation released updated consensus guidelines in 2017, defining grade 3 PGD, the most severe form, by the presence of alveolar infiltrates and a ratio of PaO2:FiO2 less than 200. Multiple donor-related, recipient-related, and perioperative risk factors for PGD have been identified, many of which are potentially modifiable. Consistently identified risk factors include donor tobacco and alcohol use; increased recipient body mass index; recipient history of pulmonary hypertension, sarcoidosis, or pulmonary fibrosis; single lung transplantation; and use of cardiopulmonary bypass, among others. Several cellular pathways have been implicated in the pathogenesis of PGD, thus presenting several possible therapeutic targets for preventing and treating PGD. Notably, use of ex vivo lung perfusion (EVLP) has become more widespread and offers a potential platform to safely investigate novel PGD treatments while expanding the lung donor pool. Even in the presence of significantly prolonged ischemic times, EVLP has not been associated with an increased risk for PGD.
Collapse
Affiliation(s)
- Jake G Natalini
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua M Diamond
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Ma JQ, Li LS, Li YH, Hu YZ. PLCε1 mediates one-lung ventilation injury by regulating the p38/RhoA/NFκB activation loop. Mol Immunol 2021; 133:135-145. [PMID: 33662817 DOI: 10.1016/j.molimm.2021.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 01/28/2021] [Accepted: 02/14/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Phospholipase C epsilon-1 (PLCε1) might be a novel and potential target in treating inflammatory conditions. In the present study, we aimed to clarify whether PLCε1 is involved in lung injury caused by one-lung ventilation (OLV) and to elucidate the potential molecular mechanism of PLCε1-mediated signaling pathway on OLV induced inflammatory response and injury. METHODS Male Sprague-Dawley (SD) rats were divided into wide-type (PLCε1-WT) group and PLCε1-KO group, and were treated with OLV for 0.5 h, 1 h, and 2 h respectively. Observation of lung tissue injury in rats was performed by Hematoxylin and eosin (HE) staining and Wet/dry (W/D) radios. In addition, pulmonary microvascular endothelial cells (PMVECs) transfected with PLCε1-si RNA, were stimulated by lipopolysaccharide (LPS). To explore the possible roles of PLCε1 in the OLV induced inflammatory injury and the involved pathway underlying, the lung tissue and bronchoalveolar lavage fluids (BALF) of OLV rats, as well as the PMVECs were prepared for further analysis. Enzyme-linked immunoassay (ELISA) was used to detect the expression of pro-inflammatory factors. The activities of related pathway proteins (NF-κB, phospho-p38, p38, phospho-ERK1/2, ERK1/2, RhoA and ROCK) were also detected by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. RESULTS Compared to the PLCε1-WT rats, PLCε1-KOrats exhibited marked alleviation of lung inflammation as shown by great reduction in lung wet/dry weight ratios, decreases in the expressions of pro-inflammatory mediators, and declines in the number of neutrophils and the protein concentration in bronchoalveolar lavage fluid (BALF). Moreover, the increased expressions of RhoA and NF-κB p65 mRNA induced by OLV were significantly inhibited in PLCε1-KO rats. In LPS treated PMVECs, PLCε1-si RNA transfection ones also showed the decrease expression of proinflammatory mediators, reduction in p38 phosphorylation levels and downregulation of RhoA/ROCK signaling activation. Co-cultured with PLCε1-si RNA and BTRB796 (p38 inhibitors) in LPS-stimulated PMVECs resulted in a significant reduction in RhoA and NF-κB activity. In addition, treatment with either ROCK inhibitor (Y-27632) or dominant negative mutant of RhoA (RhoT19 N) significantly reduced the expression of NF-κB in PLCε1-si RNA treated PMVECs. CONCLUSION The results indicated that PLCε1 played an important role in the inflammatory response induced by OLV. Moreover, through promoting p38/RhoA/ROCK activation loop, PLCε1 promoted NF-κB activation and thereby increased the expressions of inflammatory mediators, which induced the PMVECs inflammation and subsequent injury. The results of this study provide a potential therapeutic target for the reduction of inflammatory response in patients with OLV.
Collapse
Affiliation(s)
- Jia-Qin Ma
- Experimental Center of Medical Function, Kunming Medical University, No 1168 West Chunrong Rd, Kunming,650500, China
| | - Li-Sha Li
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No 157 Jinbi Rd, Kunming, 650032, China
| | - Yan-Hua Li
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No 157 Jinbi Rd, Kunming, 650032, China
| | - Yu-Zhen Hu
- Department of Anesthesiology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No 157 Jinbi Rd, Kunming, 650032, China
| |
Collapse
|
30
|
Lin Y, Lu Q, Chen C, Wang B, Guo L, Xie J, Chen C, Huang L, Dong L. A synthetic chalcone derivative, compound 39, alleviates lipopolysaccharide-induced acute lung injury in mice. Eur J Pharmacol 2021; 891:173730. [PMID: 33188742 DOI: 10.1016/j.ejphar.2020.173730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 11/06/2020] [Accepted: 11/06/2020] [Indexed: 11/22/2022]
Abstract
Our research group has previously synthesized various chalcone analogues. Of these analogues, compound 39 has been shown to exhibit potent antioxidative activities but its anti-inflammatory and anti-apoptosis effects remain unclear. Thus, the present study investigated the in vivo and in vitro effects and mechanisms of compound 39 in lipopolysaccharide (LPS)-induced acute lung injury (ALI). To induce ALI, the mice received LPS via a tracheal instillation 6 h after intragastric administration of compound 39 or vehicle. Histological changes, the lung wet/dry weight ratio, and the amounts of protein and inflammatory cells in the broncho-alveolar lavage (BAL) fluid were assessed after 24 h. Additionally, to determine its underlying mechanisms, Western blot and immunofluorescence analyses were used. Moreover, the in vitro effects of compound 39 were also investigated. In the in vivo experiment, compound 39 markedly alleviated histopathological alterations, lung edema, and protein leakage, and exhibited potent anti-inflammatory effects. In the in vitro experiments, compound 39 dose-dependently reduced the levels of pro-inflammatory cytokines and reactive oxygen species. The results suggested that the anti-inflammatory effects of compound 39 were due to suppression of the mitogen-activated protein kinase (MAPK)/nuclear factor κB (NF-κB) pathway. Compound 39 also enhanced the protein levels of Bcl-2 and reduced the protein levels of Bax and cleaved caspase-3. The present study confirmed the anti-inflammatory, oxy-radical prohibitive, and anti-apoptosis activities of compound 39 against LPS-induced tissue and cell damage, and revealed the mechanisms underlying those processes.
Collapse
Affiliation(s)
- Yuting Lin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qingdi Lu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chaolei Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Beibei Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lisha Guo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingwen Xie
- Department of Pharmacy, Pharmacy School, Wenzhou Medical University, Wenzhou, China
| | - Chengshui Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lili Huang
- Department of Pharmacy, Ningbo Medical Centre Lihuili Hospital, Ningbo, China.
| | - Li Dong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
31
|
Ricardo-da-Silva FY, Armstrong-Jr R, Vidal-dos-Santos M, Correia CDJ, Coutinho e Silva RDS, da Anunciação LF, Moreira LFP, Leuvenink HGD, Breithaupt-Faloppa AC. Long-term lung inflammation is reduced by estradiol treatment in brain dead female rats. Clinics (Sao Paulo) 2021; 76:e3042. [PMID: 34406272 PMCID: PMC8341046 DOI: 10.6061/clinics/2021/e3042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/28/2021] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVES Lung transplantation is limited by the systemic repercussions of brain death (BD). Studies have shown the potential protective role of 17β-estradiol on the lungs. Here, we aimed to investigate the effect of estradiol on the long-lasting lung inflammatory state to understand a possible therapeutic application in lung donors with BD. METHODS Female Wistar rats were separated into 3 groups: BD, subjected to brain death (6h); E2-T0, treated with 17β-estradiol (50 μg/mL, 2 mL/h) immediately after brain death; and E2-T3, treated with 17β-estradiol (50 μg/ml, 2 ml/h) after 3h of BD. Complement system activity and macrophage presence were analyzed. TNF-α, IL-1β, IL-10, and IL-6 gene expression (RT-PCR) and levels in 24h lung culture medium were quantified. Finally, analysis of caspase-3 gene and protein expression in the lung was performed. RESULTS Estradiol reduced complement C3 protein and gene expression. The presence of lung macrophages was not modified by estradiol, but the release of inflammatory mediators was reduced and TNF-α and IL-1β gene expression were reduced in the E2-T3 group. In addition, caspase-3 protein expression was reduced by estradiol in the same group. CONCLUSIONS Brain death-induced lung inflammation in females is modulated by estradiol treatment. Study data suggest that estradiol can control the inflammatory response by modulating the release of mediators after brain death in the long term. These results strengthen the idea of estradiol as a therapy for donor lungs and improving transplant outcomes.
Collapse
Affiliation(s)
- Fernanda Yamamoto Ricardo-da-Silva
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Roberto Armstrong-Jr
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Marina Vidal-dos-Santos
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Cristiano de Jesus Correia
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Raphael dos Santos Coutinho e Silva
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Lucas Ferreira da Anunciação
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Luiz Felipe Pinho Moreira
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | | - Ana Cristina Breithaupt-Faloppa
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Corresponding author. E-mail:
| |
Collapse
|
32
|
Fatty acid nitroalkenes inhibit the inflammatory response to bleomycin-mediated lung injury. Toxicol Appl Pharmacol 2020; 407:115236. [PMID: 32931793 DOI: 10.1016/j.taap.2020.115236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/03/2020] [Accepted: 09/08/2020] [Indexed: 12/29/2022]
Abstract
Fatty acid nitroalkenes are reversibly-reactive electrophiles, endogenously detectable at nM concentrations, displaying anti-inflammatory actions. Nitroalkenes like 9- or 10-nitro-octadec-9-enoic acid (e.g. nitro-oleic acid, OA-NO2) pleiotropically suppress cardiovascular inflammatory responses, with pulmonary responses less well defined. C57BL/6 J male mice were intratracheally administered bleomycin (3 U/kg, ITB), to induce pulmonary inflammation and acute injury, or saline and were treated with 50 μL OA-NO2 (50 μg) or vehicle in the same instillation and 72 h post-exposure to assess anti-inflammatory properties. Bronchoalveolar lavage (BAL) and lung tissue were collected 7d later. ITB mice lost body weight, with OA-NO2 mitigating this loss (-2.3 ± 0.94 vs -0.4 ± 0.83 g). Histology revealed ITB induced cellular infiltration, proteinaceous debris deposition, and tissue injury, all significantly reduced by OA-NO2. Flow cytometry analysis of BAL demonstrated loss of Siglec F+/F4/80+/CD45+ alveolar macrophages with ITB (89 ± 3.5 vs 30 ± 3.7%). Analysis of CD11b/CD11c expressing cells showed ITB-induced non-resident macrophage infiltration (4 ± 2.3 vs 43 ± 2.4%) was decreased by OA-NO2 (24 ± 2.4%). Additionally, OA-NO2 attenuated increases in mature, activated interstitial macrophages (23 ± 4.8 vs. 43 ± 5.4%) in lung tissue digests. Flow analysis of CD31-/CD45-/Sca-1+ mesenchymal cells revealed ITB increased CD44+ populations (1 ± 0.4 vs 4 ± 0.4MFI), significantly reduced by OA-NO2 (3 ± 0.4MFI). Single cell analysis of mesenchymal cells by western blotting showed profibrotic ZEB1 protein expression induced by ITB. Lung digest CD45+ cells revealed ITB increased HMGB1+ cells, with OA-NO2 suppressing this response. Inhibition of HMGB1 expression correlated with increased basal phospholipid production and SP-B expression in the lung lining. These findings indicate OA-NO2 inhibits ITB-induced pro-inflammatory responses by modulating resident cell function.
Collapse
|
33
|
Almeida FM, Battochio AS, Napoli JP, Alves KA, Balbin GS, Oliveira-Junior M, Moriya HT, Pego-Fernandes PM, Vieira RP, Pazetti R. Creatine Supply Attenuates Ischemia-Reperfusion Injury in Lung Transplantation in Rats. Nutrients 2020; 12:2765. [PMID: 32927837 PMCID: PMC7551831 DOI: 10.3390/nu12092765] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
Affiliation(s)
- Francine M. Almeida
- Instituto do Coraçao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-060, Brazil; (F.M.A.); (A.S.B.); (P.M.P.-F.)
| | - Angela S. Battochio
- Instituto do Coraçao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-060, Brazil; (F.M.A.); (A.S.B.); (P.M.P.-F.)
| | - João P. Napoli
- Laboratorio de Pesquisa em Cirurgia Toracica-LIM61, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-060, Brazil; (J.P.N.); (K.A.A.); (G.S.B.)
| | - Katiusa A. Alves
- Laboratorio de Pesquisa em Cirurgia Toracica-LIM61, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-060, Brazil; (J.P.N.); (K.A.A.); (G.S.B.)
| | - Grace S. Balbin
- Laboratorio de Pesquisa em Cirurgia Toracica-LIM61, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-060, Brazil; (J.P.N.); (K.A.A.); (G.S.B.)
| | - Manoel Oliveira-Junior
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), Sao Jose dos Campos 04372-020, Brazil; (M.O.-J.); (R.P.V.)
| | - Henrique T. Moriya
- Biomedical Engineering Laboratory-LEB, University of Sao Paulo, Sao Paulo 05508-060, Brazil;
| | - Paulo M. Pego-Fernandes
- Instituto do Coraçao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-060, Brazil; (F.M.A.); (A.S.B.); (P.M.P.-F.)
| | - Rodolfo P. Vieira
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), Sao Jose dos Campos 04372-020, Brazil; (M.O.-J.); (R.P.V.)
- Post-Graduation Program in Bioengineering, Universidade Brasil, Sao Paulo 05403-000, Brazil
- Post-Graduation Program in Sciences of Human Movement and Rehabilitation, Federal University of Sao Paulo (UNIFESP), Santos 04021-001, Brazil
| | - Rogerio Pazetti
- Laboratorio de Pesquisa em Cirurgia Toracica-LIM61, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05508-060, Brazil; (J.P.N.); (K.A.A.); (G.S.B.)
| |
Collapse
|
34
|
Frye CC, Bery AI, Kreisel D, Kulkarni HS. Sterile inflammation in thoracic transplantation. Cell Mol Life Sci 2020; 78:581-601. [PMID: 32803398 DOI: 10.1007/s00018-020-03615-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/20/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
The life-saving benefits of organ transplantation can be thwarted by allograft dysfunction due to both infectious and sterile inflammation post-surgery. Sterile inflammation can occur after necrotic cell death due to the release of endogenous ligands [such as damage-associated molecular patterns (DAMPs) and alarmins], which perpetuate inflammation and ongoing cellular injury via various signaling cascades. Ischemia-reperfusion injury (IRI) is a significant contributor to sterile inflammation after organ transplantation and is associated with detrimental short- and long-term outcomes. While the vicious cycle of sterile inflammation and cellular injury is remarkably consistent amongst different organs and even species, we have begun understanding its mechanistic basis only over the last few decades. This understanding has resulted in the developments of novel, yet non-specific therapies for mitigating IRI-induced graft damage, albeit with moderate results. Thus, further understanding of the mechanisms underlying sterile inflammation after transplantation is critical for identifying personalized therapies to prevent or interrupt this vicious cycle and mitigating allograft dysfunction. In this review, we identify common and distinct pathways of post-transplant sterile inflammation across both heart and lung transplantation that can potentially be targeted.
Collapse
Affiliation(s)
- C Corbin Frye
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Amit I Bery
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA.
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA
| |
Collapse
|
35
|
Kopecky BJ, Frye C, Terada Y, Balsara KR, Kreisel D, Lavine KJ. Role of donor macrophages after heart and lung transplantation. Am J Transplant 2020; 20:1225-1235. [PMID: 31850651 PMCID: PMC7202685 DOI: 10.1111/ajt.15751] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/06/2019] [Accepted: 12/08/2019] [Indexed: 01/25/2023]
Abstract
Since the 1960s, heart and lung transplantation has remained the optimal therapy for patients with end-stage disease, extending and improving quality of life for thousands of individuals annually. Expanding donor organ availability and immunologic compatibility is a priority to help meet the clinical demand for organ transplant. While effective, current immunosuppression is imperfect as it lacks specificity and imposes unintended adverse effects such as opportunistic infections and malignancy that limit the health and longevity of transplant recipients. In this review, we focus on donor macrophages as a new target to achieve allograft tolerance. Donor organ-directed therapies have the potential to improve allograft survival while minimizing patient harm related to global suppression of recipient immune responses. Topics highlighted include the role of ontogenically distinct donor macrophage populations in ischemia-reperfusion injury and rejection, including their interaction with allograft-infiltrating recipient immune cells and potential therapeutic approaches. Ultimately, a better understanding of how donor intrinsic immunity influences allograft acceptance and survival will provide new opportunities to improve the outcomes of transplant recipients.
Collapse
Affiliation(s)
| | - Christian Frye
- Department of Surgery, Washington University, Saint Louis, Missouri
| | - Yuriko Terada
- Department of Surgery, Washington University, Saint Louis, Missouri
| | - Keki R. Balsara
- Department of Surgery, Vanderbilt University, Nashville, Tennessee
| | - Daniel Kreisel
- Department of Surgery, Washington University, Saint Louis, Missouri
- Department of Pathology and Immunology, Washington University, Saint Louis, Missouri
| | - Kory J. Lavine
- Department of Medicine, Washington University, Saint Louis, Missouri
- Department of Pathology and Immunology, Washington University, Saint Louis, Missouri
- Department of Developmental Biology, Washington University, Saint Louis, Missouri
| |
Collapse
|
36
|
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: 1.8] [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
|
37
|
Liu H, Zhou K, Liao L, Zhang T, Yang M, Sun C. Lipoxin A4 receptor agonist BML-111 induces autophagy in alveolar macrophages and protects from acute lung injury by activating MAPK signaling. Respir Res 2018; 19:243. [PMID: 30518355 PMCID: PMC6282312 DOI: 10.1186/s12931-018-0937-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022] Open
Abstract
Background Acute lung injury (ALI) is a life-threatening lung disease where alveolar macrophages (AMs) play a central role both in the early phase to initiate inflammatory responses and in the late phase to promote tissue repair. In this study, we examined whether BML-111, a lipoxin A4 receptor agonist, could alter the phenotypes of AM and thus present prophylactic benefits for ALI. Methods In vitro, isolated AMs were treated with lipopolysaccharide (LPS) to induce ALI. In response to BML-111 pre-treatment, apoptosis and autophagy of AMs were examined by flow cytometry, and by measuring biomarkers for each process. The potential involvement of MAPK1 and mTOR signaling pathway was analyzed. In vivo, an LPS-induced septic ALI model was established in rats and the preventative significance of BML-111 was assessed. On the cellular and molecular levels, the pro-inflammatory cytokines TNF-α and IL-6 from bronchoalveolar lavage were measured by ELISA, and the autophagy in AMs examined using Western blot. Results BML-111 inhibited apoptosis and induced autophagy of AMs in response to ALI inducer, LPS. The enhancement of autophagy was mediated through the suppression of MAPK1 and MAPK8 signaling, but independent of mTOR signaling. In vivo, BML-111 pre-treatment significantly alleviated LPS-induced ALI, which was associated with the reduction of apoptosis, the dampened production of pro-inflammatory cytokines in the lung tissue, as well as the increase of autophagy of AMs. Conclusions This study reveals the prophylactic significance of BML-111 in ALI and the underlying mechanism: by targeting the MAPK signaling but not mTOR pathway, BML-111 stimulates autophagy in AMs, attenuates the LPS-induced cell apoptosis, and promotes the resolution of ALI.
Collapse
Affiliation(s)
- Huaizheng Liu
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China
| | - Kefu Zhou
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China
| | - Liangkan Liao
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China
| | - Tianyi Zhang
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China
| | - Mingshi Yang
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China
| | - Chuanzheng Sun
- Emergency and Intensive Care Center, The Third Xiangya Hospital of Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan Province, PR, China.
| |
Collapse
|
38
|
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: 11] [Impact Index Per Article: 1.6] [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
|
39
|
Sharma AK, Charles EJ, Zhao Y, Narahari AK, Baderdinni PK, Good ME, Lorenz UM, Kron IL, Bayliss DA, Ravichandran KS, Isakson BE, Laubach VE. Pannexin-1 channels on endothelial cells mediate vascular inflammation during lung ischemia-reperfusion injury. Am J Physiol Lung Cell Mol Physiol 2018; 315:L301-L312. [PMID: 29745255 PMCID: PMC6139659 DOI: 10.1152/ajplung.00004.2018] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/17/2018] [Accepted: 05/02/2018] [Indexed: 12/31/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury (IRI), which involves inflammation, vascular permeability, and edema, remains a major challenge after lung transplantation. Pannexin-1 (Panx1) channels modulate cellular ATP release during inflammation. This study tests the hypothesis that endothelial Panx1 is a key mediator of vascular inflammation and edema after I/R and that IRI can be blocked by Panx1 antagonism. A murine hilar ligation model of IRI was used whereby left lungs underwent 1 h of ischemia and 2 h of reperfusion. Treatment of wild-type mice with Panx1 inhibitors (carbenoxolone or probenecid) significantly attenuated I/R-induced pulmonary dysfunction, edema, cytokine production, and neutrophil infiltration versus vehicle-treated mice. In addition, VE-Cad-CreERT2+/Panx1fl/fl mice (tamoxifen-inducible deletion of Panx1 in vascular endothelium) treated with tamoxifen were significantly protected from IRI (reduced dysfunction, endothelial permeability, edema, proinflammatory cytokines, and neutrophil infiltration) versus vehicle-treated mice. Furthermore, extracellular ATP levels in bronchoalveolar lavage fluid is Panx1-mediated after I/R as it was markedly attenuated by Panx1 antagonism in wild-type mice and by endothelial-specific Panx1 deficiency. Panx1 gene expression in lungs after I/R was also significantly elevated compared with sham. In vitro experiments demonstrated that TNF-α and/or hypoxia-reoxygenation induced ATP release from lung microvascular endothelial cells, which was attenuated by Panx1 inhibitors. This study is the first, to our knowledge, to demonstrate that endothelial Panx1 plays a key role in mediating vascular permeability, inflammation, edema, leukocyte infiltration, and lung dysfunction after I/R. Pharmacological antagonism of Panx1 activity may be a novel therapeutic strategy to prevent IRI and primary graft dysfunction after lung transplantation.
Collapse
Affiliation(s)
- Ashish K Sharma
- Department of Surgery, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Eric J Charles
- Department of Surgery, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Yunge Zhao
- Department of Surgery, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Adishesh K Narahari
- Department of Pharmacology, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Pranav K Baderdinni
- Department of Pharmacology, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Miranda E Good
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Ulrike M Lorenz
- Department of Microbiology, Immunology, and Cancer, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Irving L Kron
- Department of Surgery, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Douglas A Bayliss
- Department of Pharmacology, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Kodi S Ravichandran
- Department of Microbiology, Immunology, and Cancer, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine , Charlottesville, Virginia
| | - Victor E Laubach
- Department of Surgery, University of Virginia School of Medicine , Charlottesville, Virginia
| |
Collapse
|
40
|
Fan T, Huang Z, Wang W, Zhang B, Xu Y, Mao Z, Chen L, Hu H, Geng Q. Proteasome inhibition promotes autophagy and protects from endoplasmic reticulum stress in rat alveolar macrophages exposed to hypoxia-reoxygenation injury. J Cell Physiol 2018; 233:6748-6758. [PMID: 29741768 DOI: 10.1002/jcp.26516] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 01/30/2018] [Indexed: 02/06/2023]
Abstract
Alveolar macrophages play vital roles in acute lung injury, and macrophage response to hypoxia play relevant roles to disease mechanisms. There is growing evidence that cell death pathways play crucial roles in physiological and pathological settings and that the ubiquitin-proteasome system is involved in the regulation of these processes. However, the functional role of proteasome in alveolar macrophages exposed to hypoxia-reoxygenation (H/R) injury is unknown. We aimed to investigate the function of proteasome on alveolar macrophages exposed to H/R and the underlying mechanisms. NR8383 cells were pretreated with proteasome activator sulforaphane (SFN) or inhibitor MG-132 for 1 hr, and then submitted to 2/6 hr, 4/6 hr, and 6/6 hr H/R treatment. Cell viability was assessed with MTT assay. Autophagy was monitored using electron transmission microscope and flow cytometry and western blotting. The endoplasmic reticulum (ER) stress and unfolded protein response (UPR) pathways were equally analyzed by western blotting. Cell apoptosis was detected by immunohistochemistry, caspase3/7 activity, and western blotting. The viability of NR8383 cells exposed to H/R was affected by proteasome activity and proteasome inhibition significantly inhibited cell death. Treatment with MG-132 led to autophagy activation and induced the survival of NR8383 cells exposed to H/R. Pretreatment with SFN significantly decreased cell autophagy and induced cell death. ER stress was activated in H/R-treated NR8383 cells, and SFN further promoted ER stress whereas proteasome inhibition led to contrary results. Proteasome inhibtion hindered cell apoptosis as demonstrated by decreased caspase-3/7 activity, immunolabelling, and western blot results. Proteasome inhibition might be a promising approach for treating H/R injury-related lung diseases.
Collapse
Affiliation(s)
- Tao Fan
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan
| | - Zhixin Huang
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan
| | - Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan
| | - Boyou Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan
| | - Yao Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan
| | - Zhangfan Mao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan
| | - Lei Chen
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan
| | - Hao Hu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan
| |
Collapse
|
41
|
Abstract
The goal of the present study was to investigate the role of M1 macrophages in acute lung injury (ALI). To address this, we used lipopolysaccharide (LPS)-treated wild-type and CD11b-DTR mice, and examined their M1 macrophage levels, and the extent of their inflammation and pulmonary injuries. In addition, we evaluated pulmonary function by measuring the expressions of SP-A and SP-B in infiltrated M1 macrophages. Finally, we co-cultured the mouse type II-like alveolar epithelial cells (AT-II) and mouse pulmonary microvascular endothelial cells (PMECs) with M1 macrophages in the presence of TNF-α or H2O2 and assessed them for viability and apoptosis. After LPS treatment, we observed that the number of pulmonary M1/M2 macrophages and the serum levels of interleukin-1β (IL-1β), tumor necrosis factor α (TNF-α), and reactive oxygen species (ROS) significantly increased. Furthermore, the increase in cytokines was accompanied with the initiation of lung injury indicated by the decreased levels of SP-A and SP-B. In macrophage-depleted CD11b-DTR mice, ALI was attenuated, serum levels of IL-1β, TNF-α and ROS were reduced, and lung levels of monocyte chemoattractant protein-1 (MCP-1) and macrophage inflammatory protein-2 (MIP-2) were decreased. After administering TNF-α and H2O2, the proapoptotic effect of M1 macrophages on AT-II or PMECs significantly increased, the cell viabilities significantly decreased, and apoptosis significantly increased. Our results suggest that M1 macrophages are recruited to the lungs where they significantly contribute to an increase in TNF-α and ROS production, thus initiating ALI.
Collapse
|
42
|
Lamb CA, Kirby JA. Donor intravascular monocyte trafficking: a potential therapeutic target for primary graft dysfunction following lung transplantation? Thorax 2018; 73:303-304. [PMID: 29386299 DOI: 10.1136/thoraxjnl-2017-210274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Christopher Andrew Lamb
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.,Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - John Andrew Kirby
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
43
|
Uehara H, Minami K, Quante M, Nian Y, Heinbokel T, Azuma H, El Khal A, Tullius SG. Recall features and allorecognition in innate immunity. Transpl Int 2018; 31:6-13. [PMID: 28926127 PMCID: PMC7781186 DOI: 10.1111/tri.13073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/20/2017] [Accepted: 09/15/2017] [Indexed: 11/28/2022]
Abstract
Alloimmunity traditionally distinguishes short-lived, rapid and nonspecific innate immune responses from adaptive immune responses that are characterized by a highly specific response initiated in a delayed fashion. Key players of innate immunity such as natural killer (NK) cells and macrophages present the first-line defence of immunity. The concept of unspecific responses in innate immunity has recently been challenged. The discovery of pattern recognition receptors (PRRs) has demonstrated that innate immune cells respond in a semi-specific fashion through the recognition of pathogen-associated molecular patterns (PAMPs) representing conserved molecular structures shared by large groups of microorganisms. Although immunological memory has generally been considered as exclusive to adaptive immunity, recent studies have demonstrated that innate immune cells have the potential to acquire memory. Here, we discuss allospecific features of innate immunity and their relevance in transplantation.
Collapse
Affiliation(s)
- Hirofumi Uehara
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Urology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Koichiro Minami
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Urology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Markus Quante
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Yeqi Nian
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Timm Heinbokel
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Haruhito Azuma
- Department of Urology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Abdala El Khal
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Stefan G. Tullius
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
44
|
Schnapper A, Christmann A, Knudsen L, Rahmanian P, Choi YH, Zeriouh M, Karavidic S, Neef K, Sterner-Kock A, Guschlbauer M, Hofmaier F, Maul AC, Wittwer T, Wahlers T, Mühlfeld C, Ochs M. Stereological assessment of the blood-air barrier and the surfactant system after mesenchymal stem cell pretreatment in a porcine non-heart-beating donor model for lung transplantation. J Anat 2017; 232:283-295. [PMID: 29193065 DOI: 10.1111/joa.12747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2017] [Indexed: 01/09/2023] Open
Abstract
More frequent utilization of non-heart-beating donor (NHBD) organs for lung transplantation has the potential to relieve the shortage of donor organs. In particular with respect to uncontrolled NHBD, concerns exist regarding the risk of ischaemia/reperfusion (IR) injury-related graft damage or dysfunction. Due to their immunomodulating and tissue-remodelling properties, bone-marrow-derived mesenchymal stem cells (MSCs) have been suspected of playing a beneficial role regarding short- and long-term survival and function of the allograft. Thus, MSC administration might represent a promising pretreatment strategy for NHBD organs. To study the initial effects of warm ischaemia and MSC application, a large animal lung transplantation model was generated, and the structural organ composition of the transplanted lungs was analysed stereologically with particular respect to the blood-gas barrier and the surfactant system. In this study, porcine lungs (n = 5/group) were analysed. Group 1 was the sham-operated control group. In pigs of groups 2-4, cardiac arrest was induced, followed by a period of 3 h of ventilated ischaemia at room temperature. In groups 3 and 4, 50 × 106 MSCs were administered intravascularly via the pulmonary artery and endobronchially, respectively, during the last 10 min of ischaemia. The left lungs were transplanted, followed by a reperfusion period of 4 h. Then, lungs were perfusion-fixed and processed for light and electron microscopy. Samples were analysed stereologically for IR injury-related structural parameters, including volume densities and absolute volumes of parenchyma components, alveolar septum components, intra-alveolar oedema, and the intracellular and intra-alveolar surfactant pool. Additionally, the volume-weighted mean volume of lamellar bodies (lbs) and their profile size distribution were determined. Three hours of ventilated warm ischaemia was tolerated without eliciting histological or ultrastructural signs of IR injury, as revealed by qualitative and quantitative assessment. However, warm ischaemia influenced the surfactant system. The volume-weighted mean volume of lbs was reduced significantly (P = 0.024) in groups subjected to ischaemia (group medians of groups 2-4: 0.180-0.373 μm³) compared with the sham control group (median 0.814 μm³). This was due to a lower number of large lb profiles (size classes 5-15). In contrast, the intra-alveolar surfactant system was not altered significantly. No significant differences were encountered comparing ischaemia alone (group 2) or ischaemia plus application of MSCs (groups 3 and 4) in this short-term model.
Collapse
Affiliation(s)
- Anke Schnapper
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH (From Regenerative Biology to Reconstructive Therapy), Cluster of Excellence, Hannover, Germany
| | - Astrid Christmann
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH (From Regenerative Biology to Reconstructive Therapy), Cluster of Excellence, Hannover, Germany
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH (From Regenerative Biology to Reconstructive Therapy), Cluster of Excellence, Hannover, Germany
| | - Parwis Rahmanian
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Yeong-Hoon Choi
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany.,Center of Molecular Medicine, University of Cologne, Cologne, Germany
| | - Mohamed Zeriouh
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Samira Karavidic
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Klaus Neef
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany.,Center of Molecular Medicine, University of Cologne, Cologne, Germany
| | - Anja Sterner-Kock
- Center for Experimental Medicine, University of Cologne, Cologne, Germany
| | - Maria Guschlbauer
- Center for Experimental Medicine, University of Cologne, Cologne, Germany.,Decentral Animal Facility, University of Cologne, Cologne, Germany
| | - Florian Hofmaier
- Center for Experimental Medicine, University of Cologne, Cologne, Germany
| | - Alexandra C Maul
- Center for Experimental Medicine, University of Cologne, Cologne, Germany
| | - Thorsten Wittwer
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany.,Center of Molecular Medicine, University of Cologne, Cologne, Germany
| | - Thorsten Wahlers
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany.,Center of Molecular Medicine, University of Cologne, Cologne, Germany
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany.,REBIRTH (From Regenerative Biology to Reconstructive Therapy), 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 (From Regenerative Biology to Reconstructive Therapy), 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
| |
Collapse
|
45
|
Abstract
Primary graft dysfunction is a form of acute injury after lung transplantation that is associated with significant short- and long-term morbidity and mortality. Multiple mechanisms contribute to the pathogenesis of primary graft dysfunction, including ischemia reperfusion injury, epithelial cell death, endothelial cell dysfunction, innate immune activation, oxidative stress, and release of inflammatory cytokines and chemokines. This article reviews the epidemiology, pathogenesis, risk factors, prevention, and treatment of primary graft dysfunction.
Collapse
Affiliation(s)
- Mary K Porteous
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, 423 Guardian Drive, Philadelphia, PA 19104, USA.
| | - James C Lee
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
46
|
Role of monocytes and macrophages in regulating immune response following lung transplantation. Curr Opin Organ Transplant 2017; 21:239-45. [PMID: 26977996 DOI: 10.1097/mot.0000000000000313] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Advances in the field of monocyte and macrophage biology have dramatically changed our understanding of their role during homeostasis and inflammation. Here we review the role of these important innate immune effectors in the lung during inflammatory challenges including lung transplantation. RECENT FINDINGS Neutrophil extravasation into lung tissue and the alveolar space have been shown to be pathogenic during acute lung injury as well as primary graft dysfunction following lung transplantation. Recent advances in lung immunology have demonstrated the remarkable plasticity of both monocytes and macrophages and demonstrated their importance as mediators of neutrophil recruitment and transendothelial migration during inflammation. SUMMARY Monocytes and macrophages are emerging as key players in mediating both the pathogen response and sterile lung inflammation, including that arising from barotrauma and ischemia-reperfusion injury. Ongoing studies will establish the mechanisms by which these monocytes and macrophages initiate a variety of immune response that lay the fundamental basis of injury response in the lung.
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Lungs are extremely susceptible to injury, and despite advances in surgical management and immunosuppression, outcomes for lung transplantation are the worst of any solid organ transplant. The success of lung transplantation is limited by high rates of primary graft dysfunction because of ischemia-reperfusion injury characterized by robust inflammation, alveolar damage, and vascular permeability. This review will summarize major mechanisms of lung ischemia-reperfusion injury with a focus on the most recent findings in this area. RECENT FINDINGS Over the past 18 months, numerous studies have described strategies to limit lung ischemia-reperfusion injury in experimental settings, which often reveal mechanistic insight. Many of these strategies involved the use of various antioxidants, anti-inflammatory agents, mesenchymal stem cells, and ventilation with gaseous molecules. Further advancements have been achieved in understanding mechanisms of innate immune cell activation, neutrophil infiltration, endothelial barrier dysfunction, and oxidative stress responses. SUMMARY Methods for prevention of primary graft dysfunction after lung transplant are urgently needed, and understanding mechanisms of ischemia-reperfusion injury is critical for the development of novel and effective therapeutic approaches. In doing so, both acute and chronic outcomes of lung transplant recipients will be significantly improved.
Collapse
|
48
|
Maresin 1 Ameliorates Lung Ischemia/Reperfusion Injury by Suppressing Oxidative Stress via Activation of the Nrf-2-Mediated HO-1 Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9634803. [PMID: 28751936 PMCID: PMC5511669 DOI: 10.1155/2017/9634803] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/08/2017] [Indexed: 12/13/2022]
Abstract
Lung ischemia/reperfusion (I/R) injury occurs in various clinical conditions and heavily damaged lung function. Oxidative stress reaction and antioxidant enzymes play a pivotal role in the etiopathogenesis of lung I/R injury. In the current study, we investigated the impact of Maresin 1 on lung I/R injury and explored the possible mechanism involved in this process. MaR 1 ameliorated I/R-induced lung injury score, wet/dry weight ratio, myeloperoxidase, tumor necrosis factor, bronchoalveolar lavage fluid (BALF) leukocyte count, BALF neutrophil ratio, and pulmonary permeability index levels in lung tissue. MaR 1 significantly reduced ROS, methane dicarboxylic aldehyde, and 15-F2t-isoprostane generation and restored antioxidative enzyme (superoxide dismutase, glutathione peroxidase, and catalase) activities. Administration of MaR 1 improved the expression of nuclear Nrf-2 and cytosolic HO-1 in I/R-treated lung tissue. Furthermore, we also found that the protective effects of MaR 1 on lung tissue injury and oxidative stress were reversed by HO-1 activity inhibitor, Znpp-IX. Nrf-2 transcription factor inhibitor, brusatol, significantly decreased MaR 1-induced nuclear Nrf-2 and cytosolic HO-1 expression. In conclusion, these results indicate that MaR 1 protects against lung I/R injury through suppressing oxidative stress. The mechanism is partially explained by activation of the Nrf-2-mediated HO-1 signaling pathway.
Collapse
|
49
|
Intratracheal Administration of Small Interfering RNA Targeting Fas Reduces Lung Ischemia-Reperfusion Injury. Crit Care Med 2017; 44:e604-13. [PMID: 26963318 DOI: 10.1097/ccm.0000000000001601] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Lung ischemia-reperfusion injury is the main cause of primary graft dysfunction after lung transplantation and results in increased morbidity and mortality. Fas-mediated apoptosis is one of the pathologic mechanisms involved in the development of ischemia-reperfusion injury. We hypothesized that the inhibition of Fas gene expression in lungs by intratracheal administration of small interfering RNA could reduce lung ischemia-reperfusion injury in an ex vivo model reproducing the procedural sequence of lung transplantation. DESIGN Prospective, randomized, controlled experimental study. SETTING University research laboratory. SUBJECTS C57/BL6 mice weighing 28-30 g. INTERVENTIONS Ischemia-reperfusion injury was induced in lungs isolated from mice, 48 hours after treatment with intratracheal small interfering RNA targeting Fas, control small interfering RNA, or vehicle. Isolated lungs were exposed to 6 hours of cold ischemia (4°C), followed by 2 hours of warm (37°C) reperfusion with a solution containing 10% of fresh whole blood and mechanical ventilation with constant low driving pressure. MEASUREMENTS AND MAIN RESULTS Fas gene expression was significantly silenced at the level of messenger RNA and protein after ischemia-reperfusion in lungs treated with small interfering RNA targeting Fas compared with lungs treated with control small interfering RNA or vehicle. Silencing of Fas gene expression resulted in reduced edema formation (bronchoalveolar lavage protein concentration and lung histology) and improvement in lung compliance. These effects were associated with a significant reduction of pulmonary cell apoptosis of lungs treated with small interfering RNA targeting Fas, which did not affect cytokine release and neutrophil infiltration. CONCLUSIONS Fas expression silencing in the lung by small interfering RNA is effective against ischemia-reperfusion injury. This approach represents a potential innovative strategy of organ preservation before lung transplantation.
Collapse
|
50
|
Tatham KC, O'Dea KP, Romano R, Donaldson HE, Wakabayashi K, Patel BV, Thakuria L, Simon AR, Sarathchandra P, Marczin N, Takata M. Intravascular donor monocytes play a central role in lung transplant ischaemia-reperfusion injury. Thorax 2017; 73:350-360. [PMID: 28389600 PMCID: PMC5870457 DOI: 10.1136/thoraxjnl-2016-208977] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 02/28/2017] [Accepted: 03/10/2017] [Indexed: 12/22/2022]
Abstract
Rationale Primary graft dysfunction in lung transplant recipients derives from the initial, largely leukocyte-dependent, ischaemia-reperfusion injury. Intravascular lung-marginated monocytes have been shown to play key roles in experimental acute lung injury, but their contribution to lung ischaemia-reperfusion injury post transplantation is unknown. Objective To define the role of donor intravascular monocytes in lung transplant-related acute lung injury and primary graft dysfunction. Methods Isolated perfused C57BL/6 murine lungs were subjected to warm ischaemia (2 hours) and reperfusion (2 hours) under normoxic conditions. Monocyte retention, activation phenotype and the effects of their depletion by intravenous clodronate-liposome treatment on lung inflammation and injury were determined. In human donor lung transplant samples, the presence and activation phenotype of monocytic cells (low side scatter, 27E10+, CD14+, HLA-DR+, CCR2+) were evaluated by flow cytometry and compared with post-implantation lung function. Results In mouse lungs following ischaemia-reperfusion, substantial numbers of lung-marginated monocytes remained within the pulmonary microvasculature, with reduced L-selectin and increased CD86 expression indicating their activation. Monocyte depletion resulted in reductions in lung wet:dry ratios, bronchoalveolar lavage fluid protein, and perfusate levels of RAGE, MIP-2 and KC, while monocyte repletion resulted in a partial restoration of the injury. In human lungs, correlations were observed between pre-implantation donor monocyte numbers/their CD86 and TREM-1 expression and post-implantation lung dysfunction at 48 and 72 hours. Conclusions These results indicate that lung-marginated intravascular monocytes are retained as a ‘passenger’ leukocyte population during lung transplantation, and play a key role in the development of transplant-associated ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- Kate Colette Tatham
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Kieran Patrick O'Dea
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Rosalba Romano
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.,Departments of Anaesthesia and Cardiothoracic Transplantation, Harefield Hospital, Royal Brompton and Harefield NHS Foundation Trust, Harefield, Middlesex, UK
| | - Hannah Elizabeth Donaldson
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Kenji Wakabayashi
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Brijesh Vipin Patel
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - Louit Thakuria
- Departments of Anaesthesia and Cardiothoracic Transplantation, Harefield Hospital, Royal Brompton and Harefield NHS Foundation Trust, Harefield, Middlesex, UK
| | - Andre Rudiger Simon
- Departments of Anaesthesia and Cardiothoracic Transplantation, Harefield Hospital, Royal Brompton and Harefield NHS Foundation Trust, Harefield, Middlesex, UK
| | - Padmini Sarathchandra
- Faculty of Medicine, National Heart & Lung Institute, Imperial College, Heart Science Centre, Harefield Hospital, Harefield, Middlesex, UK
| | | | - Nandor Marczin
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK.,Departments of Anaesthesia and Cardiothoracic Transplantation, Harefield Hospital, Royal Brompton and Harefield NHS Foundation Trust, Harefield, Middlesex, UK
| | - Masao Takata
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| |
Collapse
|