101
|
Ischemia-reperfusion Injury in the Transplanted Lung: A Literature Review. Transplant Direct 2021; 7:e652. [PMID: 33437867 PMCID: PMC7793349 DOI: 10.1097/txd.0000000000001104] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 02/07/2023] Open
Abstract
Lung ischemia-reperfusion injury (LIRI) and primary graft dysfunction are leading causes of morbidity and mortality among lung transplant recipients. Although extensive research endeavors have been undertaken, few preventative and therapeutic treatments have emerged for clinical use. Novel strategies are still needed to improve outcomes after lung transplantation. In this review, we discuss the underlying mechanisms of transplanted LIRI, potential modifiable targets, current practices, and areas of ongoing investigation to reduce LIRI and primary graft dysfunction in lung transplant recipients.
Collapse
|
102
|
Miceli V, Bertani A, Chinnici CM, Bulati M, Pampalone M, Amico G, Carcione C, Schmelzer E, Gerlach JC, Conaldi PG. Conditioned Medium from Human Amnion-Derived Mesenchymal Stromal/Stem Cells Attenuating the Effects of Cold Ischemia-Reperfusion Injury in an In Vitro Model Using Human Alveolar Epithelial Cells. Int J Mol Sci 2021; 22:ijms22020510. [PMID: 33419219 PMCID: PMC7825633 DOI: 10.3390/ijms22020510] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/30/2020] [Accepted: 01/03/2021] [Indexed: 02/07/2023] Open
Abstract
The clinical results of lung transplantation (LTx) are still less favorable than other solid organ transplants in both the early and long term. The fragility of the lungs limits the procurement rate and can favor the occurrence of ischemia-reperfusion injury (IRI). Ex vivo lung perfusion (EVLP) with Steen SolutionTM (SS) aims to address problems, and the implementation of EVLP to alleviate the activation of IRI-mediated processes has been achieved using mesenchymal stromal/stem cell (MSC)-based treatments. In this study, we investigated the paracrine effects of human amnion-derived MSCs (hAMSCs) in an in vitro model of lung IRI that includes cold ischemia and normothermic EVLP. We found that SS enriched by a hAMSC-conditioned medium (hAMSC-CM) preserved the viability and delayed the apoptosis of alveolar epithelial cells (A549) through the downregulation of inflammatory factors and the upregulation of antiapoptotic factors. These effects were more evident using the CM of 3D hAMSC cultures, which contained an increased amount of immunosuppressive and growth factors compared to both 2D cultures and encapsulated-hAMSCs. To conclude, we demonstrated an in vitro model of lung IRI and provided evidence that a hAMSC-CM attenuated IRI effects by improving the efficacy of EVLP, leading to strategies for a potential implementation of this technique.
Collapse
Affiliation(s)
- Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (M.B.); (P.G.C.)
- Correspondence: ; Tel.: +39-091-21-92-649
| | - Alessandro Bertani
- Thoracic Surgery and Lung Transplantation Unit, Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione, 90127 Palermo, Italy;
| | - Cinzia Maria Chinnici
- Regenerative Medicine Unit, Fondazione Ri.MED, 90127 Palermo, Italy; (C.M.C.); (M.P.); (G.A.); (C.C.)
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS–ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Matteo Bulati
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (M.B.); (P.G.C.)
| | - Mariangela Pampalone
- Regenerative Medicine Unit, Fondazione Ri.MED, 90127 Palermo, Italy; (C.M.C.); (M.P.); (G.A.); (C.C.)
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS–ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Giandomenico Amico
- Regenerative Medicine Unit, Fondazione Ri.MED, 90127 Palermo, Italy; (C.M.C.); (M.P.); (G.A.); (C.C.)
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS–ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Claudia Carcione
- Regenerative Medicine Unit, Fondazione Ri.MED, 90127 Palermo, Italy; (C.M.C.); (M.P.); (G.A.); (C.C.)
| | - Eva Schmelzer
- Department of Surgery, School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219-3130, USA; (E.S.); (J.C.G.)
| | - Jörg C. Gerlach
- Department of Surgery, School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219-3130, USA; (E.S.); (J.C.G.)
- Department of Bioengineering, School of Medicine, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219-3130, USA
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy; (M.B.); (P.G.C.)
| |
Collapse
|
103
|
Manti S, Parisi GF, Papale M, Mulè E, Aloisio D, Rotolo N, Leonardi S. Looking beyond pulmonary disease in COVID-19: A lesson from patients with cystic fibrosis. Med Hypotheses 2021; 147:110481. [PMID: 33421691 PMCID: PMC7834376 DOI: 10.1016/j.mehy.2020.110481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/11/2020] [Accepted: 12/28/2020] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) caused more than 52.775.271 million confirmed cases, 1.293.106 deaths, globally, and afflicted 208 countries, areas, or territories; and almost three months have passed since the World Health Organisation (WHO) declared COVID-19 as a pandemic. Despite the dramatic and global impact of the Coronavirus, the knowledge about the SARS-CoV-2 infection has been improved remarkably. Herein, we provided the rationale for SARS-CoV-2 infection as endothelial dysfunction rather than respiratory disease. Accordingly, we strongly invited the researchers to look beyond pulmonary injury and shift their attention from respiratory disease to endothelial disorder. This strategy could be particularly relevant to identifying therapeutic weapons stabilizing the endothelium rather than the lungs.
Collapse
Affiliation(s)
- Sara Manti
- Department of Clinical and Experimental Medicine, Pediatric Respiratory Unit, San Marco Hospital, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy.
| | - Giuseppe Fabio Parisi
- Department of Clinical and Experimental Medicine, Pediatric Respiratory Unit, San Marco Hospital, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Maria Papale
- Department of Clinical and Experimental Medicine, Pediatric Respiratory Unit, San Marco Hospital, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Enza Mulè
- Department of Clinical and Experimental Medicine, Pediatric Respiratory Unit, San Marco Hospital, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Donatella Aloisio
- Department of Clinical and Experimental Medicine, Pediatric Respiratory Unit, San Marco Hospital, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Novella Rotolo
- Department of Clinical and Experimental Medicine, Pediatric Respiratory Unit, San Marco Hospital, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| | - Salvatore Leonardi
- Department of Clinical and Experimental Medicine, Pediatric Respiratory Unit, San Marco Hospital, University of Catania, Via Santa Sofia 78, 95123 Catania, Italy
| |
Collapse
|
104
|
Park Y, Ahn JH, Lee TK, Kim B, Tae HJ, Park JH, Shin MC, Cho JH, Won MH. Therapeutic hypothermia reduces inflammation and oxidative stress in the liver after asphyxial cardiac arrest in rats. Acute Crit Care 2020; 35:286-295. [PMID: 33423440 PMCID: PMC7808856 DOI: 10.4266/acc.2020.00304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/12/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Few studies have evaluated the effects of hypothermia on cardiac arrest (CA)-induced liver damage. This study aimed to investigate the effects of hypothermic therapy on the liver in a rat model of asphyxial cardiac arrest (ACA). METHODS Rats were subjected to 5-minute ACA followed by return of spontaneous circulation (RoSC). Body temperature was controlled at 33°C±0.5°C or 37°C±0.5°C for 4 hours after RoSC in the hypothermia group and normothermia group, respectively. Liver tissues in each group were collected at 6 hours, 12 hours, 1 day, and 2 days after RoSC. To examine hepatic inflammation, mast cells were stained with toluidine blue. Superoxide anion radical production was evaluated using dihydroethidium fluorescence straining and expression of endogenous antioxidants (superoxide dismutase 1 [SOD1] and SOD2) was examined using immunohistochemistry. RESULTS There were significantly more mast cells in the livers of the normothermia group with ACA than in the hypothermia group with ACA. Gradual increase in superoxide anion radical production was found with time in the normothermia group with ACA, but production was significantly suppressed in the hypothermia group with ACA relative to the normothermia group with ACA. SOD1 and SOD2 levels were higher in the hypothermia group with ACA than in the normothermia group with ACA. CONCLUSIONS Experimental hypothermic treatment after ACA significantly inhibited inflammation and superoxide anion radical production in the rat liver, indicating that this treatment enhanced or maintained expression of antioxidants. Our findings suggest that hypothermic therapy after CA can reduce mast cell-mediated inflammation through regulation of oxidative stress and the expression of antioxidants in the liver.
Collapse
Affiliation(s)
- Yoonsoo Park
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Ji Hyeon Ahn
- Department of Physical Therapy, College of Health Science, Youngsan University, Yangsan, Korea.,Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Tae-Kyeong Lee
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Korea
| | - Bora Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Hyun-Jin Tae
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Iksan, Korea
| | - Joon Ha Park
- Department of Anatomy, College of Korean Medicine, Dongguk University, Gyeongju, Korea
| | - Myoung Cheol Shin
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
105
|
Pelleg A. Extracellular adenosine 5'-triphosphate in pulmonary disorders. Biochem Pharmacol 2020; 187:114319. [PMID: 33161021 DOI: 10.1016/j.bcp.2020.114319] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/01/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
Adenosine 5'-triphosphate (ATP) is found in every cell of the human body where it plays a critical role in cellular energetics and metabolism. ATP is released from cells under physiologic and pathophysiologic condition; extracellular ATP is rapidly degraded to adenosine 5'-diphosphate (ADP) and adenosine by ecto-enzymes (mainly, CD39 and CD73). Before its degradation, ATP acts as an autocrine and paracrine agent exerting its effects on targeted cells by activating cell surface receptors named P2 Purinergic receptors. The latter are expressed by different cell types in the lungs, the activation of which is involved in multiple pulmonary disorders. This succinct review summarizes the role of ATP in inflammation processes associated with these disorders including bronchoconstriction, cough, mechanical ventilation-induced lung injury and idiopathic pulmonary fibrosis. All of these disorders still constitute unmet clinical needs. Therefore, the various ATP-signaling pathways in pulmonary inflammation constitute attractive targets for novel drug-candidates that would improve the management of patients with multiple pulmonary diseases.
Collapse
Affiliation(s)
- Amir Pelleg
- Danmir Therapeutics, LLC, Haverford, PA, USA. http://www.danmirtherapeutics.com
| |
Collapse
|
106
|
Carbon Monoxide-Releasing Molecule-3 Ameliorates Acute Lung Injury in a Model of Hemorrhagic Shock and Resuscitation: Roles of p38MAPK Signaling Pathway. Shock 2020; 55:816-826. [PMID: 33105439 DOI: 10.1097/shk.0000000000001684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE It was reported that carbon monoxide-releasing molecule-3 (CORM-3) administration immediately after hemorrhagic shock and resuscitation (HSR) ameliorates the HSR-induced acute lung injury (ALI); however, the specific mechanism of the protective effects against HSR-induced ALI remains unclear. METHODS To induce hemorrhagic shock, rats were bled to a mean arterial blood pressure of 30 mm Hg for 45 min and then resuscitated with shed blood via the left vein. CORM-3 (4 mg/kg or 8 mg/kg) was respectively administrated after HSR. Twelve hours post-HSR, lung injury was assessed by wet/dry (W/D) ratio, hematoxylin-eosin staining staining, and lung ultrasound; the apoptotic and pyroptotic macrophages were measured by immunofluorescence staining; and the expression of phosphorylated p38 mitogen activated protein kinase (p-p38MAPK) and total p38MAPK was measured by western blotting. SB203580 (5 mg/kg), a special inhibitor of p-p38MAPK, was administrated by abdominal cavity to assess the roles of p38MAPK in HSR-induced ALI. RESULTS Increased B-line score, lung injury score, and W/D ratio indicated the fact of ALI after HSR. Twelve hours post-HSR, CORM-3 administration significantly decreased the B-line score, lung injury score, W/D ratio, apoptotic and pyroptotic macrophages, and the expressions of p-p38MAPK. Further, SB203580 not only reduced HSR-induced ALI, but also enhanced the protective effects of CORM-3 against ALI. CONCLUSION We identified the protective effects of CORM-3 against HSR-induced ALI. The mechanism might be related to the inhibition of p38MAPK signaling pathway in lung macrophages.
Collapse
|
107
|
Xu Y, Li X, Cheng Y, Yang M, Wang R. Inhibition of ACSL4 attenuates ferroptotic damage after pulmonary ischemia-reperfusion. FASEB J 2020; 34:16262-16275. [PMID: 33070393 DOI: 10.1096/fj.202001758r] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 02/05/2023]
Abstract
Lung ischemia-reperfusion (IR) injury is a common clinical pathology associated with high mortality. Ferroptosis, a novel mode of cell death elicited by iron-dependent phospholipid peroxidation, has been implicated in ischemic events. Acyl-CoA synthetase long-chain family member 4 (ACSL4) is one of the main enzymes in pro-ferroptotic lipid metabolism. In this study, the involvement of ferroptotic death in different durations of reperfusion was evaluated by assessing the iron content, malondialdehyde, and glutathione levels, ferroptosis-related protein expression, and mitochondria morphology. The roles of ferroptosis-specific inhibitor, liproxastin-1 (Lip-1), and ACSL4 modulation in a preventive regimen were assessed in vivo and in vitro. The hallmarks of pulmonary function, such as histological lung injury score, wet/dry ratio, and oxygenation index, were evaluated as well. Results showed that lung IR increased the tissue iron content and lipid peroxidation accumulation, along with key protein (GPX4 and ACSL4) expression alteration during reperfusion. Pretreatment with Lip-1 inhibited ferroptosis and ameliorated lung IR-induced injury in animal and cell models. In addition, administering ACSL4 inhibitor rosiglitazone before ischemia diminished the ferroptotic damage in IR-injured lung tissue, consistent with the protective effect of ACSL4 knockdown on lung epithelial cells subjected to hypoxia/reoxygenation. Thus, this study delineated that IR-induced ferroptotic cell death in lung tissue and ACSL4 were correlated with this process. Inhibition of ferroptosis and ACSL4 mitigated the ferroptotic damage in IR-induced lung injury by reducing lipid peroxidation and increasing the glutathione and GPX4 levels.
Collapse
Affiliation(s)
- Yixin Xu
- Department of Anesthesiology, West China Hospital, Sichuan University, The Research Units of West China (2018RU12), Chinese Academy of Medical Sciences, Chengdu, China.,Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xuehan Li
- Department of Anesthesiology, West China Hospital, Sichuan University, The Research Units of West China (2018RU12), Chinese Academy of Medical Sciences, Chengdu, China.,Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Cheng
- Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, China
| | - Mingan Yang
- Division of Biostatistics & Epidemiology, School of Public Health, San Diego State University, San Diego, CA, USA
| | - Rurong Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, The Research Units of West China (2018RU12), Chinese Academy of Medical Sciences, Chengdu, China
| |
Collapse
|
108
|
Yang YY, Lin CJ, Wang CC, Chen CM, Kao WJ, Chen YH. Consecutive Hypoxia Decreases Expression of NOTCH3, HEY1, CC10, and FOXJ1 via NKX2-1 Downregulation and Intermittent Hypoxia-Reoxygenation Increases Expression of BMP4, NOTCH1, MKI67, OCT4, and MUC5AC via HIF1A Upregulation in Human Bronchial Epithelial Cells. Front Cell Dev Biol 2020; 8:572276. [PMID: 33015064 PMCID: PMC7500169 DOI: 10.3389/fcell.2020.572276] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 08/17/2020] [Indexed: 01/11/2023] Open
Abstract
Previous studies have shown that the experimental models of hypoxia-reoxygenation (H/R) mimics the physiological conditions of ischemia-reperfusion and induce oxidative stress and injury in various types of organs, tissues, and cells, both in vivo and in vitro, including human lung adenocarcinoma epithelial cells. Nonetheless, it had not been reported whether H/R affected proliferation, apoptosis, and expression of stem/progenitor cell markers in the bronchial epithelial cells. In this study, we investigated differential effects of consecutive hypoxia and intermittent 24/24-h cycles of H/R on human bronchial epithelial (HBE) cells derived from the same-race and age-matched healthy subjects (i.e., NHBE) and subjects with chronic obstructive pulmonary disease (COPD) (i.e., DHBE). To analyze gene/protein expression during differentiation, both the NHBE and DHBE cells at the 2nd passage were cultured at the air-liquid interface (ALI) in the differentiation medium under normoxia for 3 days, followed by either culturing under hypoxia (1% O2) for consecutively 9 days and then returning to normoxia for another 9 days, or culturing under 24/24-h cycles of H/R (i.e., 24 h of 1% O2 followed by 24 h of 21% O2, repetitively) for 18 days in total, so that all differentiating HBE cells were exposed to hypoxia for a total of 9 days. In both the normal and diseased HBE cells, intermittent H/R significantly increased HIF1A, BMP4, NOTCH1, MKI67, OCT4, and MUC5AC expression, while consecutive hypoxia significantly decreased NKX2-1, NOTCH3, HEY1, CC10, and FOXJ1 expression. Inhibition of HIF1A or NKX2-1 expression by siRNA transfection respectively decreased BMP4/NOTCH1/MKI67/OCT4/MUC5AC and NOTCH3/HEY1/CC10/FOXJ1 expression in the HBE cells cultured under intermittent H/R to the same levels under normoxia. Overexpression of NKX2-1 via cDNA transfection caused more than 2.8-fold increases in NOTCH3, HEY1, and FOXJ1 mRNA levels in the HBE cells cultured under consecutive hypoxia compared to the levels under normoxia. Taken together, our results show for the first time that consecutive hypoxia decreased expression of the co-regulated gene module NOTCH3/HEY1/CC10 and the ciliogenesis-inducing transcription factor gene FOXJ1 via NKX2-1 mRNA downregulation, while intermittent H/R increased expression of the co-regulated gene module BMP4/NOTCH1/MKI67/OCT4 and the predominant airway mucin gene MUC5AC via HIF1A mRNA upregulation.
Collapse
Affiliation(s)
- Yung-Yu Yang
- Department of General Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chao-Ju Lin
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Chin Wang
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan.,Section of Respiratory Therapy, Rueifang Miner Hospital, New Taipei City, Taiwan
| | - Chieh-Min Chen
- Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Wen-Jen Kao
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Hui Chen
- Graduate Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
109
|
Han D, Sun J, Fan D, Zhang C, Du S, Zhang W. Simvastatin ameliorates oxygen glucose deprivation/reoxygenation-induced pulmonary endothelial barrier dysfunction by restoring cell-cell junctions and actin cytoskeleton dynamics via the PI3K/Akt signaling pathway. Am J Transl Res 2020; 12:5586-5596. [PMID: 33042440 PMCID: PMC7540106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 08/01/2020] [Indexed: 06/11/2023]
Abstract
Endothelial barrier dysfunction is a critical pathophysiological process of pulmonary ischemia/reperfusion (I/R) injury in patients scheduled for cardiopulmonary bypass. Impaired actin cytoskeleton dynamics and cell-cell junctions are the main causes of endothelial dysfunction. Statins have protective effects on I/R-induced lung injury; however, the mechanism is unclear. We explored the therapeutic potential of simvastatin (SV) in endothelial cells subjected to oxygen-glucose deprivation/reoxygenation (OGD/R). SV pretreatment promoted the barrier function of human pulmonary microvascular endothelial cells (HPMECs) subjected to OGD/R. LY294002 was used to evaluate the role of the PI3K/Akt pathway in regulating the barrier function of HPMECs subjected to OGD/R. LY294002 suppressed the barrier function of HPMECs. SV restored the endothelial barrier function by rescuing endothelial cell migration and permeability, which are involved in the regulation of cytoskeleton dynamics and intercellular junction expression via the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Dong Han
- Department of Cardiovascular Surgery, The Central Hospital of NanyangNanyang, Henan, PR China
| | - Junjun Sun
- Department of Cardiovascular Surgery, The Central Hospital of NanyangNanyang, Henan, PR China
| | - Dikun Fan
- Department of Cardiovascular Surgery, The Central Hospital of NanyangNanyang, Henan, PR China
| | - Chao Zhang
- Department of Cardiovascular Surgery, The Central Hospital of NanyangNanyang, Henan, PR China
| | - Shoufeng Du
- Department of Cardiovascular Surgery, The Central Hospital of NanyangNanyang, Henan, PR China
| | - Wang Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, Zhejiang, PR China
| |
Collapse
|
110
|
Creatine Supply Attenuates Ischemia-Reperfusion Injury in Lung Transplantation in Rats. Nutrients 2020; 12:nu12092765. [PMID: 32927837 PMCID: PMC7551831 DOI: 10.3390/nu12092765] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/01/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) is one of the factors limiting the success of lung transplantation (LTx). IRI increases death risk after transplantation through innate immune system activation and inflammation induction. Some studies have shown that creatine (Cr) protects tissues from ischemic damage by its antioxidant action. We evaluated the effects of Cr supplementation on IRI after unilateral LTx in rats. Sixty-four rats were divided into four groups: water + 90 min of ischemia; Cr + 90 min of ischemia; water + 180 min of ischemia; and Cr + 180 min of ischemia. Donor animals received oral Cr supplementation (0.5 g/kg/day) or vehicle (water) for five days prior to LTx. The left lung was exposed to cold ischemia for 90 or 180 min, followed by reperfusion for 2 h. We evaluated the ventilatory mechanics and inflammatory responses of the graft. Cr-treated animals showed a significant decrease in exhaled nitric oxide levels and inflammatory cells in blood, bronchoalveolar lavage fluid and lung tissue. Moreover, edema, cell proliferation and apoptosis in lung parenchyma were reduced in Cr groups. Finally, TLR-4, IL-6 and CINC-1 levels were lower in Cr-treated animals. We concluded that Cr caused a significant decrease in the majority of inflammation parameters evaluated and had a protective effect on the IRI after LTx in rats.
Collapse
|
111
|
Shi X, Liu Z, Li J. Protective effects of dexmedetomidine on hypoxia/reoxygenation injury in cardiomyocytes by regulating the CHOP signaling pathway. Mol Med Rep 2020; 22:3307-3315. [PMID: 32945482 PMCID: PMC7453597 DOI: 10.3892/mmr.2020.11442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/02/2020] [Indexed: 12/20/2022] Open
Abstract
Hypoxia/reoxygenation (H/R) injury in myocardial cells occurs frequently during cardiac surgery and affects the prognosis of patients. The present study aimed to investigate the protective effects of dexmedetomidine (Dex) on H/R injury and its association with the C/EBP-homologous protein (CHOP) signaling pathway. An H/R model was constructed in H9C2 cells to investigate the effects of Dex on H/R injury. Cell viability, apoptosis and lactate dehydrogenase (LDH) levels were determined by MTT, flow cytometry and 2,4-dinitrophenylhydrazine colorimetric assays, respectively. The expression levels of inflammatory factors were measured by reverse transcription-quantitative PCR (RT-qPCR), and CHOP and glucose-regulated protein-78 (Grp78) expression levels were detected by RT-qPCR and western blotting. CHOP was overexpressed or knocked down to detect the cell viability, apoptosis, LDH level and the expression levels of inflammatory factors and Grp78. The results demonstrated that in the H/R group, cell viability was lower and apoptosis was higher, and that higher levels of LDH and inflammatory factors were present compared with those in the Dex+H/R group. Silencing of CHOP significantly reversed the H/R-reduced cell viability, high apoptotic rate and LDH levels, as well as the elevated expression levels of inflammatory factors and Grp78 caused by H/R injury, whereas the overexpression of CHOP inhibited cell viability and promoted apoptosis, elevated LDH level and expression of inflammatory factors and Grp78 compared with the negative control. Additionally, pretreatment with Dex significantly alleviated the H/R injury; thus, Dex may protect H9C2 cells against H/R induced cell injury, possibly by suppressing the CHOP signaling pathway.
Collapse
Affiliation(s)
- Xiaoqiao Shi
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhiwen Liu
- Department of Anesthesiology, The Second Affiliated Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Junwei Li
- Department of Anesthesiology, The Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410005, P.R. China
| |
Collapse
|
112
|
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
|
113
|
Jin Z, Hana Z, Alam A, Rajalingam S, Abayalingam M, Wang Z, Ma D. Review 1: Lung transplant-from donor selection to graft preparation. J Anesth 2020; 34:561-574. [PMID: 32476043 PMCID: PMC7261511 DOI: 10.1007/s00540-020-02800-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 05/17/2020] [Indexed: 12/16/2022]
Abstract
For various end-stage lung diseases, lung transplantation remains one of the only viable treatment options. While the demand for lung transplantation has steadily risen over the last few decades, the availability of donor grafts is limited, which have resulted in progressively longer waiting lists. In the early years of lung transplantation, only the 'ideal' donor grafts are considered for transplantation. Due to the donor shortages, there is ongoing discussion about the safe use of 'suboptimal' grafts to expand the donor pool. In this review, we will discuss the considerations around donor selection, donor-recipient matching, graft preparation and graft optimisation.
Collapse
Affiliation(s)
- Zhaosheng Jin
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Zac Hana
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Azeem Alam
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Shamala Rajalingam
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Mayavan Abayalingam
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK
| | - Zhiping Wang
- Department of Anesthesiology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Daqing Ma
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, SW10 9NH, UK.
| |
Collapse
|
114
|
Zhao N, Li CC, Di B, Xu LL. Recent advances in the NEK7-licensed NLRP3 inflammasome activation: Mechanisms, role in diseases and related inhibitors. J Autoimmun 2020; 113:102515. [PMID: 32703754 DOI: 10.1016/j.jaut.2020.102515] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022]
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor containing pyrin domain 3 (NLRP3) inflammasome is a high-molecular-weight complex mediated by the activation of pattern-recognition receptors (PRRs) seed in innate immunity. Once NLRP3 is activated, the following recruitment of the adapter apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD) (ASC) and procaspase-1 would be initiated. Cleavage of procaspase-1 into active caspase-1 then leads to the maturation of the precursor forms of interleukin (IL)-1β and IL-18 into biologically active IL-1β and IL-18. The activation of NLRP3 inflammasome is thought to be tightly associated with a regulator never in mitosis A (NIMA)-related kinase 7 (NEK7), apart from other signaling events such as K+ efflux and reactive oxygen species (ROS). Plus, the NLRP3 inflammasome has been linked to various metabolic disorders, chronic inflammation and other diseases. In this review, we firstly describe the cellular/molecular mechanisms of the NEK7-licensed NLRP3 inflammasome activation. Then we detail the potential inhibitors that can selectively and effectively modulate either the NEK7-NLRP3 complex itself or the related molecular/cellular events. Finally, we describe some inhibitors as promising therapeutic strategies for diverse diseases driven by NLRP3 inflammasome.
Collapse
Affiliation(s)
- Ni Zhao
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Cui-Cui Li
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Bin Di
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China.
| | - Li-Li Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
115
|
|
116
|
Zhou Y, Zhang L, Guan J, Yin X. Improvement of lung ischemia-reperfusion injury by inhibition of microRNA-155 via reductions in neuroinflammation and oxidative stress of vagal afferent nerve. Pulm Circ 2020; 10:2045894020922125. [PMID: 32547733 PMCID: PMC7273343 DOI: 10.1177/2045894020922125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/25/2020] [Indexed: 11/25/2022] Open
Abstract
Lung ischemia–reperfusion injury (LIRI) is a common clinical concern. As the injury
occurs, the pulmonary afferent nerves play a key role in regulating respiratory functions
under pathophysiological conditions. The present study was to examine the effects of
inhibiting microRNA-155 on the levels of proinflammatory cytokines and products of
oxidative stress in the pulmonary vagal afferent nerves and the commissural nucleus of the
solitary tract (cNTS) after LIRI. A rat model of LIRI was used. ELISA method was employed
to examine proinflammatory cytokines, namely, IL-1β, IL-6 and TNF-α; and key biomarkers of
oxidative stress, 8-isoprostaglandin F2α (8-iso PGF2α) and 8-hydroxy-2′-deoxyguanosine
(8-OHdG). In results, in the process of LIRI, the levels of microRNA-155 were amplified in
the vagal afferent nerves and cNTS, and this was accompanied with increases of IL-1β, IL-6
and TNF-α; and 8-iso PGF2α and 8-OHdG. Application of microRNA-155 inhibitor, but not its
scramble, attenuated the elevation of proinflammatory cytokines and amplification of 8-iso
PGF2α and 8-OHdG in those nerve tissues. In conclusion, we observed the abnormalities in
the pulmonary afferent pathways at the levels of the peripheral nerves and brainstem,
which is likely to affect respiratory functions as LIRI occurs. Our data suggest that
blocking microRNA-155 signal pathways plays a beneficial role in regulating LIRI via
inhibiting responses of neuroinflammation and oxidative stress signal pathways to
LIRI.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Lianjie Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jingjing Guan
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xin Yin
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
117
|
Regadenoson to the rescue: On the road toward prevention of primary graft dysfunction after lung transplantation. J Heart Lung Transplant 2020; 39:571-572. [PMID: 32303398 DOI: 10.1016/j.healun.2020.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/20/2020] [Indexed: 11/21/2022] Open
|
118
|
Liu G, Chen X, Wang Q, Yuan L. NEK7: a potential therapy target for NLRP3-related diseases. Biosci Trends 2020; 14:74-82. [PMID: 32295992 DOI: 10.5582/bst.2020.01029] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
NLRP3 inflammasome plays an essential role in innate immunity, yet the activation mechanism of NLRP3 inflammasome is not clear. In human or animal models, inappropriate NLRP3 inflammasome activation is implicated in many NLRP3-related diseases, such as tumors, inflammatory diseases and autoimmune diseases. Until now, a great number of inhibitors have been used to disturb the related signaling pathways, such as IL-1β blockade, IL-18 blockade and caspase-1 inhibitors. Unfortunately, most of these inhibitors just disturb the signaling pathways after the activation of NLRP3 inflammasome. Inhibitors that directly regulate NLRP3 to abolish the inflammation response may be more effective. NEK7 is a multifunctional kinase affecting centrosome duplication, mitochondrial regulation, intracellular protein transport, DNA repair and mitotic spindle assembly. Researchers have made significant observations on the regulation of gene transcription or protein expression of the NLRP3 inflammasome signaling pathway by NEK7. Those signaling pathways include ROS signaling, potassium efflux, lysosomal destabilization, and NF-κB signaling. Furthermore, NEK7 has been proved to be involved in many NLRP3-related diseases in humans or in animal models. Inhibitors focused on NEK7 may regulate NLRP3 to abolish the inflammation response and NEK7 may be a potential therapeutic target for NLRP3-related diseases.
Collapse
Affiliation(s)
- Ganglei Liu
- Department of Geriatrics Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xueliang Chen
- Department of Geriatrics Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qianqian Wang
- Department of Oncology, The Affiliated Zhuzhou Hospital of Xiangya Medical College, Central South University, Zhuzhou, Hunan, China
| | - Lianwen Yuan
- Department of Geriatrics Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
119
|
Dorrello NV, Vunjak-Novakovic G. Bioengineering of Pulmonary Epithelium With Preservation of the Vascular Niche. Front Bioeng Biotechnol 2020; 8:269. [PMID: 32351946 PMCID: PMC7174601 DOI: 10.3389/fbioe.2020.00269] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/16/2020] [Indexed: 12/20/2022] Open
Abstract
The shortage of transplantable donor organs directly affects patients with end-stage lung disease, for which transplantation remains the only definitive treatment. With the current acceptance rate of donor lungs of only 20%, rescuing even one half of the rejected donor lungs would increase the number of transplantable lungs threefold, to 60%. We review recent advances in lung bioengineering that have potential to repair the epithelial and vascular compartments of the lung. Our focus is on the long-term support and recovery of the lung ex vivo, and the replacement of defective epithelium with healthy therapeutic cells. To this end, we first review the roles of the lung epithelium and vasculature, with focus on the alveolar-capillary membrane, and then discuss the available and emerging technologies for ex vivo bioengineering of the lung by decellularization and recellularization. While there have been many meritorious advances in these technologies for recovering marginal quality lungs to the levels needed to meet the standards for transplantation – many challenges remain, motivating further studies of the extended ex vivo support and interventions in the lung. We propose that the repair of injured epithelium with preservation of quiescent vasculature will be critical for the immediate blood supply to the lung and the lung survival and function following transplantation.
Collapse
Affiliation(s)
- N Valerio Dorrello
- Department of Pediatrics, Columbia University, New York, NY, United States
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY, United States.,Department of Medicine, Columbia University, New York, NY, United States
| |
Collapse
|
120
|
Ye C, Qi W, Dai S, Zou G, Liu W, Yu B, Tang J. microRNA-223 promotes autophagy to aggravate lung ischemia-reperfusion injury by inhibiting the expression of transcription factor HIF2α. Am J Physiol Lung Cell Mol Physiol 2020; 319:L1-L10. [PMID: 32267722 DOI: 10.1152/ajplung.00009.2020] [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: 01/14/2023] Open
Abstract
Lung ischemia-reperfusion (I/R) injury severely endangers human health, and recent studies have suggested that certain microRNAs (miRNAs) play important roles in this pathological phenomenon. The current study aimed to ascertain the ability of miR-223 to influence lung I/R injury by targeting hypoxia-inducible factor-2α (HIF2α). First, mouse models of lung I/R injury were established: during surgical procedures, pulmonary arteries and veins and unilateral pulmonary portal vessels were blocked and resuming bilateral pulmonary ventilation, followed by restoration of bipulmonary ventilation. In addition, a lung I/R injury cell model was constructed by exposure to hypoxic reoxygenation (H/R) in mouse pulmonary microvascular endothelial cells (PMVECs). Expression of miR-223, HIF2α and β-catenin in tissues or cells was determined by RT-qPCR and Western blot analysis. Correlation between miR-223 and HIF2α was analyzed by dual luciferase reporter gene assay. Further, lung tissue injury and mouse PMVEC apoptosis was evaluated by HE, TUNEL staining and flow cytometry. Autophagosomes in cells were detected by light chain3 immunofluorescence assay. miR-223 was expressed at a high level while HIF2α/β-catenin was downregulated in tissues and cells with lung I/R injury. Further, miR-223 targeted and repressed HIF2α expression to downregulate β-catenin expression. The miR-223/HIF2α/β-catenin axis aggravated H/R injury in mouse PMVECs and lung I/R injury in mice by enhancing autophagy. Taken together, miR-223 inhibits HIF2α to repress β-catenin, thus contributing to autophagy to complicate lung I/R injury. These findings provide a promising therapeutic target for treating lung I/R injury.
Collapse
Affiliation(s)
- Chunlin Ye
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, China
| | - Wanghong Qi
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, China
| | - Shaohua Dai
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, China
| | - Guowen Zou
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, China
| | - Weicheng Liu
- Department of Anesthesiology, the First Affiliated Hospital of Nanchang University
| | - Bentong Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, China
| | - Jian Tang
- Thoracic Surgery, the First Affiliated Hospital of Nanchang University, China
| |
Collapse
|
121
|
Jin Y, Sun M, Lv X, Wang X, Jiang G, Chen C, Wen Z. Extracellular histones play a pathogenic role in primary graft dysfunction after human lung transplantation. RSC Adv 2020; 10:12485-12491. [PMID: 35497627 PMCID: PMC9051052 DOI: 10.1039/d0ra00127a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/22/2020] [Indexed: 11/21/2022] Open
Abstract
Primary graft dysfunction (PGD) causes early mortality and late graft failure after lung transplantation. The mechanisms of PGD are not fully understood but ischemia/reperfusion (I/R) injury may be involved. Extracellular histones have recently been identified as major contributors to I/R injury. Hence, we investigated whether extracellular histones are associated with PGD after lung transplantation. In total, 65 lung transplant patients were enrolled into this study. Blood samples were collected from patients before and serially after transplantation (24 h, 48 h, and 72 h) and measured for extracellular histones, myeloperoxidase (MPO), lactate dehydrogenase (LDH), and multiple cytokines. Besides, the patients' sera were cultured with human pulmonary artery endothelial cells (HPAEC) and human monocyte cell line (THP1) cells, respectively, and cellular viability and cytokine production were determined. Heparin or anti-histone antibody were used to study the effects of histone-neutralized interventions. The results showed that extracellular histones increased markedly after lung transplantation, peaked by 24 h and tended to decrease thereafter, but still retained high levels up to 72 h. Extracellular histones were more abundant in patients with PGD (n = 8) than patients without PGD (n = 57) and linearly correlated with MPO, LDH, and most detected cytokines. Ex vivo studies showed that the patients' sera collected within 24 h after transplantation were very damaging to HPAEC cells and promoted cytokine production in cultured THP1 cells, which could be largely prevented by heparin or anti-histone antibodies. These data suggested a pathogenic role for extracellular histones in PGD after lung transplantation. Targeting extracellular histones may serve as a preventive and therapeutic strategy for PGD following lung transplantation.
Collapse
Affiliation(s)
- Yang Jin
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| | - Meng Sun
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| | - Xingan Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine Pittsburgh PA 15213 USA
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine Shanghai 200433 China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine Shanghai 200433 China
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine Zhengmin Road 507 Shanghai 200433 China
| |
Collapse
|
122
|
Yeudall S, Leitinger N, Laubach VE. Extracellular nucleotide signaling in solid organ transplantation. Am J Transplant 2020; 20:633-640. [PMID: 31605463 PMCID: PMC7042041 DOI: 10.1111/ajt.15651] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/12/2019] [Accepted: 09/25/2019] [Indexed: 01/25/2023]
Abstract
The role of extracellular purine nucleotides, including adenosine triphosphate (ATP) and adenosine, as modulators of posttransplantation outcome and ischemia-reperfusion injury is becoming increasingly evident. Upon pathological release of ATP, binding and activation of P2 purinergic surface receptors promote tissue injury and inflammation, while the expression and activation of P1 receptors for adenosine have been shown to attenuate inflammation and limit ischemia-induced damage, which are central to the viability and long-term success of allografts. Here we review the current state of the transplant field with respect to the role of extracellular nucleotide signaling, with a focus on the sources and functions of extracellular ATP. The connection between ischemia reperfusion, purinergic signaling, and graft preservation, as well as the role of ATP and adenosine as driving factors in the promotion and suppression of posttransplant inflammation and allograft rejection, are discussed. We also examine novel therapeutic approaches that take advantage of the ischemia-reperfusion-responsive and immunomodulatory roles for purinergic signaling with the goal of enhancing graft viability, attenuating posttransplant inflammation, and minimizing complications including rejection, graft failure, and associated comorbidities.
Collapse
Affiliation(s)
- Scott Yeudall
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia,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
|
123
|
Commentary: Ischemia reperfusion-Looking ahead. J Thorac Cardiovasc Surg 2020; 161:e124-e125. [PMID: 32107028 DOI: 10.1016/j.jtcvs.2019.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 11/24/2022]
|
124
|
Shade BC, Dudley S, McCabe T, Gray-DeAngelis K. Use of pulmonoplegia and delivery system during recipient surgery in lung transplantation. Perfusion 2020; 35:587-590. [PMID: 32081092 DOI: 10.1177/0267659120906078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lung transplantation in the United States has steadily grown over the last decade. Major attention has been with the understanding of lung ischemia-reperfusion injury and how it relates to primary graft dysfunction. In 2015, our institution implemented the use of a pulmonoplegia solution during recipient surgery of lung transplantation. A unique circuit utilizing the heart lung machine is used to deliver the pulmonoplegia solution. This system is considered to be a key contributing factor to the success of our lung transplant program.
Collapse
Affiliation(s)
- Brandon C Shade
- Department of Perfusion Services, Heart and Vascular Center, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Steven Dudley
- Department of Perfusion Services, Heart and Vascular Center, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Tara McCabe
- Department of Perfusion Services, Heart and Vascular Center, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Kathryn Gray-DeAngelis
- Department of Perfusion Services, Heart and Vascular Center, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
125
|
Wang F, Wang F, Li F, Wang D, Li H, He X, Zhang J. Methane attenuates lung ischemia-reperfusion injury via regulating PI3K-AKT-NFκB signaling pathway. J Recept Signal Transduct Res 2020; 40:209-217. [PMID: 32079441 DOI: 10.1080/10799893.2020.1727925] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective: This study aims to investigate the protective effects and possible mechanism of methane-rich saline (MS) on lung ischemia-reperfusion injury (LIRI) in rats.Methods: MS (2 ml/kg and 20 ml/kg) was injected intraperitoneally in rats after LIRI. Lung injury was assayed by Hematoxylin-eosin (HE) staining and wet-to-dry weight (W/D). The cells in the bronchoalveolar lavage fluid (BALF) and blood were counted. Oxidative stress was examined by the level of malondialdehyde (MDA) and superoxide dismutase (SOD). Inflammatory factors including tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-10 (IL-10) were determined by ELISA. Lung tissue apoptosis was detected by TUNEL staining and western blotting of Bcl-2, Bax, and caspase-3. The expressions of IкBα, p38, PI3K, AKT, and NF-κB were analyzed with Western blotting.Results: MS effectively decreased the lung W/D ratio as well as the lung pathological damage and reduced the localized infiltration of inflammatory cells. Methane suppressed the expression of the PI3K-AKT-NFκB signaling pathway during the lung IR injury, which inhibited the activation of NF-kB and decreased the level of inflammatory cytokines, such as TNF-α, IL-1β, and IL-10. Moreover, we found that MS treatment relieved reactive oxygen species (ROS) damage by downregulating MDA and upregulating SOD. MS treatment also regulated apoptosis-related proteins, such as Bcl-2, Bax, and caspase-3.Conclusions: MS could repair LIRI and reduce the release of oxidative stress, inflammatory cytokines, and cell apoptosis via the PI3K-AKT-NFκB signaling pathway, which may provide a novel and promising strategy for the treatment of LIRI.
Collapse
Affiliation(s)
- Fang Wang
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Feidi Wang
- Hou Zonglian Medical Experimental Class, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fengtao Li
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dong Wang
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haopeng Li
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xijing He
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jingyao Zhang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
126
|
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
|
127
|
Yang C, Yang W, He Z, He H, Yang X, Lu Y, Li H. Kaempferol Improves Lung Ischemia-Reperfusion Injury via Antiinflammation and Antioxidative Stress Regulated by SIRT1/HMGB1/NF-κB Axis. Front Pharmacol 2020; 10:1635. [PMID: 32116668 PMCID: PMC7025570 DOI: 10.3389/fphar.2019.01635] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/16/2019] [Indexed: 12/24/2022] Open
Abstract
Trauma, organ transplantation, and thromboembolism are the main causes of lung ischemia-reperfusion injury (LIRI), and new therapies and drugs are urgent to relieve LIRI. In preliminary experiment, authors found that kaempferol could improve LIRI in rats, and the current study further explored its possible mechanism. The model of rat LIRI was established and appropriate research methods were implemented. Results shown that kaempferol could significantly improve LIRI, inhibit release of inflammatory factors including interleukin (IL) 6 and tumor necrosis factor (TNF) α in bronchoalveolar lavage fluid, and reduce oxidative stress reaction. Western blotting was used to detect protein expression levels and found that kaempferol could up-regulate the protein expressions of phosphorylated (p-) p65 and p65, and down-regulate the protein expression of sirtuin (SIRT) 1. Immunofluorescence was used to localize the expression of high mobility group box (HMGB) 1 and found its higher expression in outside of nucleus. However, the above effects of kaempferol on LIRI markedly attenuated by EX 527, a selective inhibitor of SIRT 1. Taken together, we first reported the protective effect of kaempferol on rat LIRI and confirmed that kaempferol’s antiinflammation and antioxidative stress involving the SIRT1/HMGB1/NF-κB axis.
Collapse
Affiliation(s)
- Chunli Yang
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Wenkai Yang
- Department of Cardiovascular Surgery, Affiliated Central People's Hospital of Zhanjiang of Guangdong Medical University, Zhanjiang, China
| | - Zhaohui He
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Huiwei He
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Xiaogang Yang
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Yuanhua Lu
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Hongbo Li
- Department of Intensive Care, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| |
Collapse
|
128
|
Miranda LE, Mente ED, Fernandes Molina CA, Sumarelli Albuquerque AA, Rubens de Nadai T, Arcêncio L, Basile-Filho A, Barbosa Evora PR. Methylene blue and the NO/cGMP pathway in solid organs transplants. Minerva Anestesiol 2020; 86:423-432. [PMID: 31994368 DOI: 10.23736/s0375-9393.20.13841-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The nitric oxide/cyclic guanosine monophosphate (NO/cGMP) pathway has a significative influence in hemodynamic changes that occur in transplants. Classically, the ischemia-reperfusion syndrome (IRS) is characterized by hypotension and low vascular resistance, when cGMP and nitric oxide (NO) are increased, contributing to oxidative stress, within an inflammatory context. These mechanisms occur in several types of transplants, such as liver, heart, lungs, kidney, which are a therapeutic choice in several clinical conditions when conventional treatments failed. It is well known the significant relation between graft dysfunction or rejection and ischemia-reperfusion injury that is linked to inflammatory response and NO/cGMP pathway activation. This review aims to study the NO/cGMP pathway in solid organ transplants. Finally, we inquire whether physicians do not underestimate the NO/cGMP pathway.
Collapse
Affiliation(s)
- Luiz E Miranda
- Division of Cardiothoracic Surgery, Department of Surgery and Anatomy, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Enio D Mente
- Division of Cardiothoracic Surgery, Department of Surgery and Anatomy, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Carlos A Fernandes Molina
- Division of Cardiothoracic Surgery, Department of Surgery and Anatomy, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Agnes A Sumarelli Albuquerque
- Division of Cardiothoracic Surgery, Department of Surgery and Anatomy, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Tales Rubens de Nadai
- Division of Cardiothoracic Surgery, Department of Surgery and Anatomy, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Livia Arcêncio
- Division of Cardiothoracic Surgery, Department of Surgery and Anatomy, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Anibal Basile-Filho
- Division of Cardiothoracic Surgery, Department of Surgery and Anatomy, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Paulo R Barbosa Evora
- Division of Cardiothoracic Surgery, Department of Surgery and Anatomy, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil -
| |
Collapse
|
129
|
Relaxin Positively Influences Ischemia-Reperfusion Injury in Solid Organ Transplantation: A Comprehensive Review. Int J Mol Sci 2020; 21:ijms21020631. [PMID: 31963613 PMCID: PMC7013572 DOI: 10.3390/ijms21020631] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/07/2020] [Accepted: 01/17/2020] [Indexed: 02/07/2023] Open
Abstract
In recent decades, solid organ transplantation (SOT) has increased the survival and quality of life for patients with end-stage organ failure by providing a potentially long-term treatment option. Although the availability of organs for transplantation has increased throughout the years, the demand greatly outweighs the supply. One possible solution for this problem is to extend the potential donor pool by using extended criteria donors. However, organs from such donors are more prone to ischemia reperfusion injury (IRI) resulting in higher rates of delayed graft function, acute and chronic graft rejection and worse overall SOT outcomes. This can be overcome by further investigating donor preconditioning strategies, graft perfusion and storage and by finding novel therapeutic agents that could reduce IRI. relaxin (RLX) is a peptide hormone with antifibrotic, antioxidant, anti-inflammatory and cytoprotective properties. The main research until now focused on heart failure; however, several preclinical studies showed its potentials for reducing IRI in SOT. The aim of this comprehensive review is to overview currently available literature on the possible role of RLX in reducing IRI and its positive impact on SOT.
Collapse
|
130
|
Yan X, Lu QG, Zeng L, Li XH, Liu Y, Du XF, Bai GM. Synergistic protection of astragalus polysaccharides and matrine against ulcerative colitis and associated lung injury in rats. World J Gastroenterol 2020; 26:55-69. [PMID: 31933514 PMCID: PMC6952295 DOI: 10.3748/wjg.v26.i1.55] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 11/29/2019] [Accepted: 12/14/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a main form of inflammatory bowel disease. Due to complicated etiology and a high rate of recurrence, it is quite essential to elucidate the underlying mechanism of and search for effective therapeutic methods for UC. AIM To investigate the effects of astragalus polysaccharides (APS) combined with matrine on UC and associated lung injury. METHODS UC was induced in rats by colon mucosal tissue sensitization combined with trinitro-benzene-sulfonic acid-ethanol. Then, the effects of the treatments of salazopyrine, APS, matrine, and APS combined with matrine on histopathological changes of lung and colon tissues, disease activity index (DAI), colon mucosal damage index (CMDI), serum endotoxin (ET) level, serum diamine oxidase (DAO) activity, the contents of tumor necrosis factor-α and interleukin-1β, and the activities of myeloperoxidase, superoxide dismutase, and malondialdehyde in lung tissues, as well as the protein expression of zonula occludens (ZO)-1, Occludin, and trefoil factor 3 (TFF3) were detected in UC rats. RESULTS The treatments of salazopyrine, APS, matrine, and APS combined with matrine reduced DAI scores and improved histopathological changes of colon and lung tissues, as well as decreased CMDI scores, ET levels, and DAO activities in UC rats. Moreover, in lung tissues, inflammatory response and oxidative stress injury were relieved after the treatments of salazopyrine, APS, matrine, and APS combined with matrine in UC rats. Furthermore, the expression of ZO-1, Occludin, and TFF3 in lung and colon tissues was increased after different treatments in UC rats. Notably, APS combined with matrine exerted a better protective effect against UC and lung injury compared with other treatments. CONCLUSION APS combined with matrine exert a synergistic protective effect against UC and lung injury, which might be associated with regulating TFF3 expression.
Collapse
Affiliation(s)
- Xin Yan
- College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan 063210, Hebei Province, China
| | - Qing-Ge Lu
- Department of Anorectal Medicine, Tangshan Traditional Chinese Medicine Hospital, Tangshan 063000, Hebei Province, China
| | - Li Zeng
- Department of Anorectal Medicine, Tangshan Traditional Chinese Medicine Hospital, Tangshan 063000, Hebei Province, China
| | - Xiao-Hai Li
- Department of Anorectal Medicine, Tangshan Traditional Chinese Medicine Hospital, Tangshan 063000, Hebei Province, China
| | - Yu Liu
- Department of Anorectal Medicine, Tangshan Traditional Chinese Medicine Hospital, Tangshan 063000, Hebei Province, China
| | - Xue-Feng Du
- Department of Anorectal Medicine, Tangshan Traditional Chinese Medicine Hospital, Tangshan 063000, Hebei Province, China
| | - Guo-Min Bai
- Department of Anorectal Medicine, Tangshan Traditional Chinese Medicine Hospital, Tangshan 063000, Hebei Province, China
| |
Collapse
|
131
|
Effect of left ventricular diastolic dysfunction on development of primary graft dysfunction after lung transplant. Curr Opin Anaesthesiol 2019; 33:10-16. [PMID: 31789901 DOI: 10.1097/aco.0000000000000811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW Primary graft dysfunction (PGD) is one of the most common complications after lung transplant and is associated with significant early and late morbidity and mortality. The cause of primary graft dysfunction is often multifactorial involving patient, donor, and operational factors. Diastolic dysfunction is increasingly recognized as an important risk factor for development of PGD after lung transplant and here we examine recent evidence on the topic. RECENT FINDINGS Patients with end-stage lung disease are more likely to suffer from cardiovascular disease including diastolic dysfunction. PGD as result of ischemia-reperfusion injury after lung transplant is exacerbated by increased left atrial pressure and pulmonary venous congestion impacted by diastolic dysfunction. Recent studies on relationship between diastolic dysfunction and PGD after lung transplant show that patients with diastolic dysfunction are more likely to develop PGD with worse survival outcome and complicated hospital course. SUMMARY Patients with diastolic dysfunction is more likely to suffer from PGD after lung transplant. From the lung transplant candidate selection to perioperative and posttransplant care, thorough evaluation and documentation diastolic dysfunction to guide patient care are imperative.
Collapse
|
132
|
Buchko MT, Stewart CJ, Hatami S, Himmat S, Freed DH, Nagendran J. Total parenteral nutrition in ex vivo lung perfusion: Addressing metabolism improves both inflammation and oxygenation. Am J Transplant 2019; 19:3390-3397. [PMID: 31420938 DOI: 10.1111/ajt.15572] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 07/15/2019] [Accepted: 08/04/2019] [Indexed: 01/25/2023]
Abstract
Ex vivo lung perfusion (EVLP) protocols generally limit metabolic supplementation to insulin and glucose. We sought to determine whether the addition of total parenteral nutrition (TPN) would improve lung function in EVLP. Ten porcine lungs were perfused using EVLP for 24 hours and supplemented with insulin and glucose. In the treatment group (n = 5), the perfusate was also supplemented with a continuous infusion of TPN containing lipids, amino acids, essential vitamins, and cofactors. Physiologic parameters and perfusate electrolytes were continuously evaluated. Perfusate lactate, lipid and branch chain amino acid (BCAA) concentrations were also analyzed to elucidate how substrates were being utilized over time. Lungs in the TPN group exhibited significantly better oxygenation. Perfusate sodium was more stable in the TPN group. In the control group, free fatty acids (FFA) were quickly depleted, reaching negligible levels early in the perfusion. Alternatively, BCAA in the control group rose continually over the perfusion demonstrating a shift toward proteolysis for energy substrate. In the TPN group, both FFA and BCAA concentrations remained stable at in vivo levels after initial stabilization. TNF-α concentrations were lower in the TPN group. The addition of TPN in EVLP allows for better electrolyte composition, decreased inflammation, and improved graft performance.
Collapse
Affiliation(s)
- Max T Buchko
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Mazankowski Alberta Heart Institute, Edmonton, AB, Canada
| | - Catherine J Stewart
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Sanaz Hatami
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Sayed Himmat
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Darren H Freed
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Mazankowski Alberta Heart Institute, Edmonton, AB, Canada.,Alberta Transplant Institute, Edmonton, AB, Canada.,Canadian National Transplant Research Program, Edmonton, AB, Canada
| | - Jayan Nagendran
- Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, AB, Canada.,Mazankowski Alberta Heart Institute, Edmonton, AB, Canada.,Alberta Transplant Institute, Edmonton, AB, Canada.,Canadian National Transplant Research Program, Edmonton, AB, Canada
| |
Collapse
|
133
|
Zeng J, Li X, Cheng Y, Ke B, Wang R. Activation of cannabinoid receptor type 2 reduces lung ischemia reperfusion injury through PI3K/Akt pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:4096-4105. [PMID: 31933805 PMCID: PMC6949786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/24/2019] [Indexed: 06/10/2023]
Abstract
Cannabinoid receptor-2 activation plays a protective role against ischemic reperfusion injury (IRI) in various organs, and exerts a protective effect against paraquat-induced acute lung injury, while the role of CB2 in lung IRI remains unclear. Hence, the present study was designed to explore the role of CB2 in lung IRI, and whether the PI3K pathway was involved. C57BL/6 mice were subjected to lung ischemia by clamping the left hilum for 1 hour, followed by 2 hours' reperfusion. Mice were pretreated with vehicle, CB2 agonist JWH133, or antagonist AM630 followed by JWH133. Arterial blood and left lung tissues were collected to detect the PaO2/FiO2 ratio, lung wet-to-dry weight ratio, lung pathologic scoring, pro-inflammatory cytokines, MDA, and SOD. Secondly, mice were pretreated with vehicle, JWH133, or both PI3K-inhibitor LY294002 and JWH133. Arterial blood and left lung tissues were collected for the above studies and protein expression of CB2 receptor, p-AKT, and AKT. After mice were pretreated with JWH133, IR-induced lung edema and lung histopathologic changes were significantly attenuated. Pretreatment with JWH133 improved PaO2/FiO2 ratio, decreased lung TNF-α, IL-6, MDA levels and MPO activities, and increased SOD activity. By contrast, the protective effect of JWH133 was blocked by pretreatment with CB2 antagonist AM630. Similarly, pretreatment with PI3K-inhibitor weakened the protection induced by JWH133, and downregulated the expression of p-AKT without altering CB2 expression. The study suggested that activation of CB2 receptor plays a protective role against IR-induced lung injury through reducing inflammation in mice. The PI3K/Akt pathway might be involved in the protective effect of CB2 receptors in lung IRI.
Collapse
Affiliation(s)
- Jieting Zeng
- Department of Anesthesiology, Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Xuehan Li
- Department of Anesthesiology, Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Yan Cheng
- Department of Anesthesiology, Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Bowen Ke
- Department of Anesthesiology, Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| | - Rurong Wang
- Department of Anesthesiology, Laboratory of Anesthesia and Intensive Care Medicine, West China Hospital of Sichuan University Chengdu, Sichuan, China
| |
Collapse
|
134
|
Wang Q, Zhu L, Xing F, Zhao P, Wang F. The comparison of the effects of local cooling and heating on apoptosis and pyroptosis of early-stage pressure ulcers in rats. J Cell Biochem 2019; 121:1649-1663. [PMID: 31560409 DOI: 10.1002/jcb.29399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 08/13/2019] [Indexed: 12/26/2022]
Abstract
The exploration of an effective method for preventing and treating pressure ulcers (PUs) is a hot topic in medical research. Recently, disputes about the choice of heat and cold therapies have emerged for the prevention and treatment of clinical PUs. The present study was designed to compare the effect of cool and heat therapies on pyroptosis and apoptosis of early-stage PUs in rats. Sixty SD rats of SPF grade were randomly divided into the sham group, model group, heating group, and cooling group. We established a rat model of early-stage PUs by using an ischemia-reperfusion method. At the end of the experiment, the tissue underneath the compressed region was collected for hematoxylin and eosin staining, transmission electron microscopy, immunohistochemistry, immunofluorescence staining, a TdT-mediated dUTP nick-end labeling assay, a Western blot analysis, and a mitochondrial swelling experiment. Our results suggested that the mitochondrial apoptotic pathway and pyroptosis were involved in the formation of early-stage PUs, and local heating increased the PU injury in rats, while local cooling reduced the PU injury in rats. This study showed that heat therapy might not be suitable for the clinical treatment and care of early-stage PUs, while cold therapy may be more appropriate.
Collapse
Affiliation(s)
- Qing Wang
- Department of Nursing, Institute of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan, Hebei, China
| | - Liang Zhu
- Department of Nursing, Institute of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan, Hebei, China
| | - Fengmei Xing
- Department of Nursing, Institute of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan, Hebei, China
| | - Ping Zhao
- Department of Nursing, Institute of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan, Hebei, China
| | - Fenglan Wang
- Department of Nursing, Institute of Nursing and Rehabilitation, North China University of Science and Technology, Tangshan, Hebei, China
| |
Collapse
|
135
|
Zampieri D, Azzollini N, Vuljan S, Pezzuto F, Fiori S, Mister M, Todeschini M, Rodriguez Ordonez PY, Marulli G, Rea F, Calabrese F, Casiraghi F. Vein Suturing Results in Worse Lung Graft Outcomes Compared to the Cuff Method. Eur Surg Res 2019; 60:106-116. [PMID: 31480059 DOI: 10.1159/000501805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 06/28/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND The rat orthotopic lung transplant model is not widely used yet because of the complexity of the procedure, in particular, venous anastomosis. Here, we performed a rat orthotopic lung transplantation using either the suture (ST) or cuff (CT) method for vein anastomosis. OBJECTIVES To compare the vein ST and CT techniques in terms of operative time, success, recipient survival, and early histological outcomes was the objective of this study. METHODS A total of 24 left lung transplants in rats were performed. Twelve syngeneic (Lewis to Lewis) and 12 allogeneic (Brown-Norway to Lewis) lung transplants were performed using either the vein ST or the CT procedure. Arterial and bronchial anastomoses were performed with the CT technique. Graft histological damage was evaluated 3-7 days post-transplant in all rat lungs. RESULTS The surgical success rate was 75% in both the ST and CT groups. Failures related mainly to vein bleeding (n = 2 in the ST group) and thrombosis (n = 1 in the ST group; n = 2 in the CT group). Total ischemia time was longer in the ST group (122 ± 25 min in ST group vs. 83 ± 10 min in CT group, mean ± SD), due to prolonged warm ischemia time (60 ± 12 min in the ST group vs. 21 ± 5 min in the CT group, mean ± SD), reflecting the time required to complete the vein ST procedure. The prolonged warm ischemia time resulted in significantly higher vascular inflammation in syngeneic grafts (2.3 ± 1.2 ST group vs. 0 in the CT group, mean ± SD) and in increased severity of ischemia/reperfusion injury and acute graft rejection (3.6 ± 0.4 in the ST group vs. 2.6 ± 0.4 in the CT group, mean ± SD) in allogeneic lung transplants. CONCLUSIONS The vein ST technique is a more time-consuming procedure than the CT method and the prolonged anastomosis time has a deleterious impact on transplant outcomes. These findings suggest that warm ischemia time - one of the modifiable transplant factors - should be considered a major risk factor in lung transplantation, particularly in the setting of donation after cardiac death.
Collapse
Affiliation(s)
- Davide Zampieri
- Department of Cardiologic, Thoracic Surgery Unit, Thoracic and Vascular Sciences, University of Padua, Padua, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Nadia Azzollini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Stefania Vuljan
- Department of Cardiologic, Thoracic Surgery Unit, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - Federica Pezzuto
- Department of Cardiologic, Thoracic Surgery Unit, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - Sonia Fiori
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Marilena Mister
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Marta Todeschini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | | | - Giuseppe Marulli
- Department of Cardiologic, Thoracic Surgery Unit, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - Federico Rea
- Department of Cardiologic, Thoracic Surgery Unit, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | - Fiorella Calabrese
- Department of Cardiologic, Thoracic Surgery Unit, Thoracic and Vascular Sciences, University of Padua, Padua, Italy
| | | |
Collapse
|
136
|
Smirnova NF, Eickelberg O. Three Is Better than One: An Improved Multiple-Hit Model of Primary Graft Dysfunction. Am J Respir Cell Mol Biol 2019; 61:141-142. [PMID: 30908931 PMCID: PMC6670037 DOI: 10.1165/rcmb.2019-0082ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Natalia F Smirnova
- 1Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAurora, Colorado
| | - Oliver Eickelberg
- 1Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAurora, Colorado
| |
Collapse
|
137
|
Gu X, Nan Y, Pang X, Zhang W, Zhang J, Zhang Y. EXPRESS: Products of oxidative stress and TRPA1 expression in the brainstem of rats after lung ischemia-reperfusion injury. Pulm Circ 2019; 9:2045894019865169. [PMID: 31267824 PMCID: PMC6681259 DOI: 10.1177/2045894019865169] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/27/2019] [Indexed: 12/30/2022] Open
Abstract
Lung ischemia–reperfusion injury is a common clinical concern. As the injury occurs, the pulmonary afferent nerves play a key role in regulating respiratory functions under pathophysiological conditions. The present study was to examine products of oxidative stress and expression of transient receptor potential A1 in the commissural nucleus of the solitary tract after lung ischemia–reperfusion injury; and further to determine molecular mediators linking to activation of oxidative stress and transient receptor potential ankyrin A1. A rat model of lung ischemia–reperfusion injury was used. Enzyme-linked immunosorbent assay and western blot analysis were employed to examine products of oxidative stress (i.e. 8-isoprostaglandin F2α and 8-hydroxy-2′-deoxyguanosine), and expression of transient receptor potential A1, Nrf2-antioxidant response element, and NADPH oxidase. 8-isoprostaglandin F2α and 8-hydroxy-2′-deoxyguanosine were amplified in the commissural nucleus of the solitary tract of lung ischemia–reperfusion injury rats, accompanied with downregulation of Nrf2-antioxidant response element, and upregulation of NOX4 and transient receptor potential A1. Blocking NADPH oxidase (subtype NOX4) decreased products of oxidative stress in the commissural nucleus of the solitary tract and attenuated upregulation of transient receptor potential A1 induced by lung ischemia–reperfusion injury. Our data revealed specific signaling pathways by which lung ischemia–reperfusion injury impairs Nrf2-antioxidant response and activates oxidative stress in the brainstem thereby leading to amplification of transient receptor potential A1 receptor likely via products of oxidative stress. Data suggest the abnormalities in the pulmonary afferent signals at the brainstem level which is likely to affect respiratory functions as lung ischemia–reperfusion injury occurs.
Collapse
Affiliation(s)
- Xiaoying Gu
- Department of Anesthesiology, The First
Hospital of Jilin University, Changchun, China
| | - Yu Nan
- Department of Gynecology and Obstetrics,
The Second Hospital of Jilin University, Changchun, China
| | - Xiaochuan Pang
- Clinical Laboratory, The First Hospital
of Jilin University, Changchun, China
| | - Wenwen Zhang
- Department of Anesthesiology, The First
Hospital of Jilin University, Changchun, China
| | - Jian Zhang
- Department of Anesthesiology, The First
Hospital of Jilin University, Changchun, China
| | - Yiyuan Zhang
- Department of Anesthesiology, The First
Hospital of Jilin University, Changchun, China
| |
Collapse
|
138
|
Zhang C, Yu S, Zheng B, Liu D, Wan F, Ma Y, Wang J, Gao Z, Shan Z. miR-30c-5p Reduces Renal Ischemia-Reperfusion Involving Macrophage. Med Sci Monit 2019; 25:4362-4369. [PMID: 31185006 PMCID: PMC6582680 DOI: 10.12659/msm.914579] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background Ischemia-reperfusion (I/R) leads to kidney injury. Renal I/R frequently occurs in kidney transplantations and acute kidney injuries. Recent studies reported that miR-30 stimulated immune responses and reductions in renal I/R related to anti-inflammation. Our study investigated the effects of miR-30c-5p on renal I/R and the relationship among miR-30c-5p, renal I/R, and macrophages. Material/Methods Sprague Dawley rats received intravenous tail injections of miR-30c-5p agomir. Then a renal I/R model were established by removing the left kidney and clamping the right renal artery. Serum creatinine (Cr) was analyzed using a serum Cr assay kit, and serum neutrophil gelatinase associated lipocalin (NGAL) was measured using a NGAL ELISA (enzyme-linked immunosorbent assay) kit. Rat kidney tissues were analyzed using hematoxylin and eosin staining. THP-1 cells treated with miR-30c-5p agomir and miR-30c-5p antagomir were measured with quantitative reverse transcription-polymerase chain reaction. Protein levels were analyzed by western blot. Results MiR-30c-5p agomir reduced serum Cr, serum NGAL, and renal I/R injury. MiR-30c-5p agomir inhibited the expression of CD86 (M1 macrophage marker), inducible nitric oxide synthase (iNOS), and tumor necrosis factor-alpha (TNF-α) and promoted the expression of CD206 (M2 macrophage marker), interleukin (IL)-4, and IL-10 in rat kidneys. MiR-30c-5p agomir reduced the expression of CD86 and iNOS, and increased the expression of CD206 and IL-10 in THP-1 cells. Conclusions We preliminarily demonstrated that miR-30c-5p agomir might decrease renal I/R through transformation of M1 macrophages to M2 macrophages and resulted in changes in inflammatory cytokines.
Collapse
Affiliation(s)
- Chengjun Zhang
- Department of Organ Transplantation, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland).,Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Shengqiang Yu
- Department of Organ Transplantation, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland).,Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Binyan Zheng
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Dongfu Liu
- Department of Organ Transplantation, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland).,Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Fengchun Wan
- Department of Organ Transplantation, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland).,Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Yue Ma
- Department of Organ Transplantation, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland).,Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Jiantao Wang
- Department of Organ Transplantation, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland).,Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Zhenli Gao
- Department of Organ Transplantation, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland).,Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Zhengfei Shan
- Department of Organ Transplantation, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland).,Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
139
|
Wei L, Li J, Han Z, Chen Z, Zhang Q. Silencing of lncRNA MALAT1 Prevents Inflammatory Injury after Lung Transplant Ischemia-Reperfusion by Downregulation of IL-8 via p300. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:285-297. [PMID: 31604167 PMCID: PMC6796730 DOI: 10.1016/j.omtn.2019.05.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 05/17/2019] [Indexed: 12/15/2022]
Abstract
Ischemia-reperfusion injury is a common early complication after lung transplantation. It was reported that long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is involved in ischemia-reperfusion injury and regulates inflammation. This study aimed to explore the role of MALAT1 in inflammatory injury following lung transplant ischemia-reperfusion (LTIR). A LTIR rat model was successfully established, with the expression of MALAT1 and interleukin-8 (IL-8) in lung tissues detected. Then, in vitro loss- and gain-of-function investigations were conducted to evaluate the effect of MALAT1 on pulmonary epithelial cell apoptosis and IL-8 expression. The relationship among MALAT1, p300, and IL-8 was tested. Moreover, a sh-MALAT1-mediated model of LTIR was established in vivo to examine inflammatory injury and chemotaxis infiltration. Both IL-8 and MALAT1 were highly expressed in LTIR. MALAT1 interacted with p300 to regulate the IL-8 expression by recruiting p300. Importantly, silencing of MALAT1 inhibited the chemotaxis of neutrophils by downregulating IL-8 expression via binding to p300. Besides, MALAT1 silencing alleviated the inflammatory injury after LTIR by downregulating IL-8 and inhibiting infiltration and activation of neutrophils. Collectively, these results demonstrated that silencing of MALAT1 ameliorated the inflammatory injury after LTIR by inhibiting chemotaxis of neutrophils through p300-mediated downregulation of IL-8, providing clinical insight for LTIR injury.
Collapse
Affiliation(s)
- Li Wei
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou 450003, P.R. China
| | - Jiwei Li
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou 450003, P.R. China.
| | - Zhijun Han
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou 450003, P.R. China
| | - Zhong Chen
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou 450003, P.R. China
| | - Quan Zhang
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University and People's Hospital of Henan University, Zhengzhou 450003, P.R. China
| |
Collapse
|
140
|
Scozzi D, Ibrahim M, Liao F, Lin X, Hsiao HM, Hachem R, Tague LK, Ricci A, Kulkarni HS, Huang HJ, Sugimoto S, Krupnick AS, Kreisel D, Gelman AE. Mitochondrial damage-associated molecular patterns released by lung transplants are associated with primary graft dysfunction. Am J Transplant 2019; 19:1464-1477. [PMID: 30582269 PMCID: PMC6482093 DOI: 10.1111/ajt.15232] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/12/2018] [Accepted: 12/08/2018] [Indexed: 02/07/2023]
Abstract
Primary graft dysfunction (PGD) is a major limitation in short- and long-term lung transplant survival. Recent work has shown that mitochondrial damage-associated molecular patterns (mtDAMPs) can promote solid organ injury, but whether they contribute to PGD severity remains unclear. We quantitated circulating plasma mitochondrial DNA (mtDNA) in 62 patients, before lung transplantation and shortly after arrival to the intensive care unit. Although all recipients released mtDNA, high levels were associated with severe PGD development. In a mouse orthotopic lung transplant model of PGD, we detected airway cell-free damaged mitochondria and mtDNA in the peripheral circulation. Pharmacologic inhibition or genetic deletion of formylated peptide receptor 1 (FPR1), a chemotaxis sensor for N-formylated peptides released by damaged mitochondria, inhibited graft injury. An analysis of intragraft neutrophil-trafficking patterns reveals that FPR1 enhances neutrophil transepithelial migration and retention within airways but does not control extravasation. Using donor lungs that express a mitochondria-targeted reporter protein, we also show that FPR1-mediated neutrophil trafficking is coupled with the engulfment of damaged mitochondria, which in turn triggers reactive oxygen species (ROS)-induced pulmonary edema. Therefore, our data demonstrate an association between mtDAMP release and PGD development and suggest that neutrophil trafficking and effector responses to damaged mitochondria are drivers of graft damage.
Collapse
Affiliation(s)
- Davide Scozzi
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department of Clinical & Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Mohsen Ibrahim
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department Medical-Surgical Science & Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - Fuyi Liao
- Department of Surgery, Washington University School, St. Louis, Missouri
| | - Xue Lin
- Department of Surgery, Washington University School, St. Louis, Missouri
| | - Hsi-Min Hsiao
- Department of Surgery, Washington University School, St. Louis, Missouri
| | - Ramsey Hachem
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Laneshia K Tague
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Alberto Ricci
- Department of Clinical & Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Hrishikesh S Kulkarni
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Howard J Huang
- Houston Methodist J. C. Walter Jr. Transplant Center, Houston, Texas
| | - Seiichiro Sugimoto
- Department of General Thoracic Surgery, Okayama University Hospital, Okayama, Japan
| | - Alexander S Krupnick
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Daniel Kreisel
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Andrew E Gelman
- Department of Surgery, Washington University School, St. Louis, Missouri
- Department Medical-Surgical Science & Translational Medicine, Sapienza University of Rome, Rome, Italy
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
141
|
Wang ML, Wei CH, Wang WD, Wang JS, Zhang J, Wang JJ. Melatonin attenuates lung ischaemia-reperfusion injury via inhibition of oxidative stress and inflammation. Interact Cardiovasc Thorac Surg 2019; 26:761-767. [PMID: 29346581 DOI: 10.1093/icvts/ivx440] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/22/2017] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES Lung ischaemia-reperfusion injury is a complex pathophysiological process due to the production of reactive oxygen species and the generation of inflammatory reaction. We investigated the protective effects and the corresponding mechanism of melatonin (MT), a potent free-radical scavenger, on lung injury induced by ischaemia-reperfusion in a mouse model. METHODS Adult male C57BL/6J mice (n = 30) were randomly and equally allocated into 5 groups: sham controls, IR, IR + 10 mg/kg MT, IR + 20 mg/kg MT and IR + 30 mg/kg MT. Lung ischaemia-reperfusion injury was induced by thoracotomy followed by clamping of the left hilum for 1 h and subsequent reperfusion for 2 h. RESULTS Histological scoring analysis showed that lung parenchymal damage was ameliorated in the melatonin pretreatment groups when compared with the IR group, with the IR + 20 mg/kg MT group showing the strongest effect among the melatonin pretreatment groups. Wet-to-dry weight ratio, detection of malondialdehyde, protein expressions of inflammatory factors (tumour necrosis factor-α, interleukin-1β, NF-κB and IKK-γ) and apoptotic factors (cleaved caspase-3 and Bax/Bcl-2), as well as TUNEL assay showed changes similar to those of the lung injury scores in all groups. In contrast, the examination of superoxide dismutase showed a pattern contrary to that of the lung injury score in all groups. In addition, immunohistochemistry staining showed that the expressions of the antioxidants glutathione peroxidase and glutathione reductase were increased in the melatonin pretreatment groups. CONCLUSIONS This study demonstrated that melatonin pretreatment attenuated lung ischaemia-reperfusion injury via inhibition of oxidative stress, inflammation and apoptosis.
Collapse
Affiliation(s)
- Ming-Liang Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun-Hua Wei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Dong Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Shun Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Zhang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Jun Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
142
|
Khaket TP, Kang SC, Mukherjee TK. The Potential of Receptor for Advanced Glycation End Products (RAGE) as a Therapeutic Target for Lung Associated Diseases. Curr Drug Targets 2019; 20:679-689. [DOI: 10.2174/1389450120666181120102159] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/17/2018] [Accepted: 11/02/2018] [Indexed: 12/27/2022]
Abstract
The receptor for advanced glycation end products (RAGE) is a multi-ligand pattern recognition
receptor that is highly expressed in lung epithelial cells. It helps alveolar epithelial cells to
maintain their morphology and specific architecture. However, in various pathophysiological conditions,
pulmonary tissues express a supraphysiological level of RAGE and its ligands including advanced
glycation end products, high mobility group box 1 proteins, and S100 proteins. On interaction
with RAGE, these ligands stimulate downstream signaling that generates inflammation and oxidative
stress leading to asthma, chronic obstructive pulmonary disease, lung cancers, idiopathic pulmonary
fibrosis, acute lung injury, pneumonia, bronchopulmonary dysplasia, cystic fibrosis, and sepsis. Thus,
pharmacological agents that can either suppress the production of RAGE or block its biological activity
would offer promising therapeutic value against pathogenesis of the aforementioned lungassociated
diseases. This review presents a comprehensive overview of the recent progress made in
defining the functions of RAGE in lung-associated diseases.
Collapse
Affiliation(s)
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, Korea
| | - Tapan Kumar Mukherjee
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana, Haryana, India
| |
Collapse
|
143
|
Abstract
Lung transplantation is a life-saving operation for patients with advanced lung disease. Pulmonary allografts eventually fail because of infection, thromboembolism, malignancy, airway complications, and chronic rejection, otherwise known as chronic lung allograft dysfunction (CLAD). Emerging evidence suggests that a highly-compromised airway circulation contributes to the evolution of airway complications and CLAD. There are two significant causes of poor perfusion and airway hypoxia in lung transplantation: an abnormal bronchial circulation which causes airway complications and microvascular rejection which induces CLAD. At the time of transplantation, the bronchial artery circulation, a natural component of the airway circulatory anatomy, is not surgically connected, and bronchi distal to the anastomosis become hypoxic. Subsequently, the bronchial anastomosis is left to heal under ischemic conditions. Still later, the extant microvessels in transplant bronchi are subjected to alloimmune insults that can further negatively impact pulmonary function. This review describes how airway tissue hypoxia evolves in lung transplantation, why depriving oxygenation in the bronchi and more distal bronchioles contributes to disease pathology and what therapeutic interventions are currently emerging to address these vascular injuries. Improving anastomotic vascular healing at the time of transplantation and preventing microvascular loss during acute rejection episodes are two steps that could limit airway hypoxia and improve patient outcomes.
Collapse
Affiliation(s)
- Shravani Pasnupneti
- VA Palo Alto Health Care System/Stanford University, 3801 Miranda Ave., Palo Alto CA 94304 USA
| | - Mark R. Nicolls
- VA Palo Alto Health Care System/Stanford University, 3801 Miranda Ave., Palo Alto CA 94304 USA
| |
Collapse
|
144
|
Mesenchymal stromal cells-derived exosomes alleviate ischemia/reperfusion injury in mouse lung by transporting anti-apoptotic miR-21-5p. Eur J Pharmacol 2019; 852:68-76. [PMID: 30682335 DOI: 10.1016/j.ejphar.2019.01.022] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/18/2018] [Accepted: 01/22/2019] [Indexed: 12/12/2022]
Abstract
MiR-21-5p is an anti-apoptotic miRNA known to mediate the protective effect of mesenchymal stromal cell-secreted exosomes (MSC-Exo) against oxidative stress-induced cell death. In the present research we employed murine lung ischemia/reperfusion (I/R) model and in vitro hypoxia/reoxygenation (H/R) model using primary murine pulmonary endothelial cells to investigate whether MSC-Exo could alleviate lung IRI by transporting miR-21-5p. Our data suggested that intratracheal administration of MSC-Exo or miR-21-5p agomir significantly reduced lung edema and dysfunction, M1 polarization of alveolar macrophages as well as secretion of HMGB1, IL-8, IL-1β, IL-6, IL-17 and TNF-α. Pre-challenge of MSCs by H/R significant increased miR-21-5p expression level in exosomes they secreted and the anti-IRI effect of these MSC-Exo, while pre-treatment of MSCs with miR-21-5p antagomir showed opposite effect. We further demonstrated that MSC-Exo ameliorated IRI in vivo or H/R induced apoptosis in vitro by inhibiting both intrinsic and extrinsic apoptosis pathway via miR-21-5p targeting PTEN and PDCD4, while artificial overexpressing PTEN or PDCD4 significantly attenuated the anti-apoptotic effect of MSC-Exo in vitro. Treatment with miR-21-5p agomir mimicked the IRI-reducing and anti-apoptotic effect of MSC-Exo. Our data suggested that MSC-Exo alleviate IRI in lung in an exosomal miR-21-5p-dependent manner. Treatment with MSC-Exo or miR-21-5p agomir might ameliorate IRI in lung.
Collapse
|
145
|
The Nox1/Nox4 inhibitor attenuates acute lung injury induced by ischemia-reperfusion in mice. PLoS One 2018; 13:e0209444. [PMID: 30571757 PMCID: PMC6301701 DOI: 10.1371/journal.pone.0209444] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/27/2018] [Indexed: 02/05/2023] Open
Abstract
Lung ischemia and reperfusion injury (LIRI) were mediated by several processes including over-production of reactive oxygen species (ROS) and inflammatory activation. ROS generated by nicotinamide adenine dinucletide phosphate (NADPH) oxidase (Nox) may play a pivotal role in pathophysiological changes in a range of disease. However, it was poorly understood in LIRI. Thus, the purpose of our study was to explore whether GKT137831, as a special dual inhibitor of Nox1 and 4, could alleviate LIRI in mice model and explore the minimal dose. According to the protocol, this study was divided into two parts. The first part was to determine the minimal dose of Nox1/4 inhibitor in attenuating LIRI via histopathology and apoptosis analysis. Eighteen C57BL/6J male wild-type mice were randomly divided in to sham, 2.5Nox+sham, 5.0Nox+sham, IR, 2.5Nox+IR and 5.0Nox+IR groups. According to the different group, mice were pretreated with corresponding dose of Nox1/4 inhibitors or normal saline. After LIRI, the results showed 5.0mg/kg Nox1/4 inhibitor could be considered as the minimal dose to alleviate injury by decreasing of lung injury score and the number of TUNEL-positive cells. The second part was to further verify the benefit of 5.0mg/kg Nox1/4 inhibitor in lung protective effects. Thirty-seven C57BL/6J male wild-type mice were divided in to sham, IR and 5.0Nox+IR groups randomly. The results showed that expressions of inflammatory, autophagy cytokines were markedly elevated and PH value was declined after LIRI. However, 5.0 mg/kg Nox1/4 inhibitor significantly attenuated cytokine production as reflected by immunohistochemistry, western blotting and Q-PCR analysis. In conclusion, our findings suggested that 5.0mg/kg Nox1/4 inhibitor contributed to protect lung tissue damage after LIRI via the suppression of inflammatory and autophagy activation.
Collapse
|
146
|
Takamori S, Shoji F, Okamoto T, Kozuma Y, Matsubara T, Haratake N, Akamine T, Katsura M, Takada K, Toyokawa G, Tagawa T, Maehara Y. HMGB1 blockade significantly improves luminal fibrous obliteration in a murine model of bronchiolitis obliterans syndrome. Transpl Immunol 2018; 53:13-20. [PMID: 30508580 DOI: 10.1016/j.trim.2018.11.007] [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] [Received: 08/19/2018] [Revised: 11/25/2018] [Accepted: 11/27/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although high-mobility group box-1 (HMGB1), which is a nuclear protein, was reported to enhance the allogeneic responses in transplantation, the effect of HMGB1 on bronchiolitis obliterans syndrome (BOS) is unknown. METHODS A murine heterotopic tracheal transplantation model was used. Protein concentrations of HMGB1, interferon-γ (IFN-γ), interleukin (IL)-10, and IL-17 were analyzed in the isografts, allografts, controls, and HMGB1-neutralizing antibody administered allografts (n = 6; Days 1, 3, 5, 7, 14, 21, and 28). The luminal fibrous occlusion was analyzed (n = 6; Days 7, 14, 21, and 28). Infiltrating CD8 and CD4 T lymphocytes around the allografts and serum levels of IFN-γ and IL-10 were evaluated (n = 6; Day 7). RESULTS The HMGB1 levels in the allografts were significantly increased compared with the isografts at Day 7. HMGB1 blockade did not change the IL-17 level, but decreased the IFN-γ/IL-10 ratio in the early phase (Days 5 and 7) and significantly improved the fibrous occlusion in the late phase (Days 14, 21, and 28). HMGB1 blockade significantly suppressed the CD8 T lymphocytes infiltration and decreased the serum IFN-γ/IL-10 ratio compared with the control at Day 7. CONCLUSIONS HMGB1 may be a trigger of the BOS pathogenesis and candidate target for the treatment of the disease.
Collapse
Affiliation(s)
- Shinkichi Takamori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Fumihiro Shoji
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan.
| | - Tatsuro Okamoto
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Yuka Kozuma
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Taichi Matsubara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Naoki Haratake
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Takaki Akamine
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Masakazu Katsura
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Kazuki Takada
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Gouji Toyokawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Tetsuzo Tagawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka 812-8582, Japan
| |
Collapse
|
147
|
Tamura T, Otulakowski G, Kavanagh BP. Could nanotechnology make vitamin E therapeutically effective? Am J Physiol Lung Cell Mol Physiol 2018; 316:L1-L5. [PMID: 30407864 DOI: 10.1152/ajplung.00430.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Vitamin E (VitE) has important antioxidant and anti-inflammatory effects and is necessary for normal physiological function. α-Tocopherol (α-T), the predominant form of VitE in human tissues, has been extensively studied. Other VitE forms, particularly γ-tocopherol (γ-T), are also potent bioactive molecules. The effects are complex, involving both reactive oxygen and nitrogen species, but trials of VitE have been generally negative. We propose that a nanoparticle approach to delivery of VitE might provide effective delivery and therapeutic effect.
Collapse
Affiliation(s)
- Tetsuya Tamura
- Program in Translational Medicine, The Research Institute, and the Departments of Critical Care Medicine and Anesthesia, Hospital for Sick Children, University of Toronto , Toronto , Canada
| | - Gail Otulakowski
- Program in Translational Medicine, The Research Institute, and the Departments of Critical Care Medicine and Anesthesia, Hospital for Sick Children, University of Toronto , Toronto , Canada
| | - Brian P Kavanagh
- Program in Translational Medicine, The Research Institute, and the Departments of Critical Care Medicine and Anesthesia, Hospital for Sick Children, University of Toronto , Toronto , Canada
| |
Collapse
|
148
|
The selective Nlrp3 inflammasome inhibitor Mcc950 attenuates lung ischemia-reperfusion injury. Biochem Biophys Res Commun 2018; 503:3031-3037. [DOI: 10.1016/j.bbrc.2018.08.089] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 08/12/2018] [Indexed: 12/21/2022]
|
149
|
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
|
150
|
The Anti-inflammatory Effect of Hydrogen on Lung Transplantation Model of Pulmonary Microvascular Endothelial Cells During Cold Storage Period. Transplantation 2018; 102:1253-1261. [DOI: 10.1097/tp.0000000000002276] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|